Scolaris Content Display Scolaris Content Display

旧大陆皮肤利什曼病的干预措施

Contraer todo Desplegar todo

Referencias

Adam 2009 {published data only}

Adam I, Hagelnur A. Artesunate plus sulfamethoxypyrazine/pyrimethamine for the treatment of cutaneous leishmaniasis: a double‐blind, placebo‐controlled clinical trial. International Journal of Antimicrobial Agents 2009;34(4):380‐1. [PUBMED: 19409761 ]CENTRAL

Al‐Fouzan 1991 {published data only}

al‐Fouzan AS, al‐Saleh QA, Najem NM, Rostom AI. Cutaneous leishmaniasis in Kuwait. Clinical experience with itraconazole. International Journal of Dermatology 1991;30(7):519‐21. [PUBMED: 1663089]CENTRAL

Al Hamdi 2010 {published data only}

Al Hamdi K, Awad AH, Moker HM. Evaluation of intralesional 0.2% ciprofloxacin as a treatment for cutaneous leishmaniasis. Eastern Mediterranean Health Journal 2010;16(1):89‐93. [PUBMED: 20214164]CENTRAL

Alkhawajah 1997 {published data only}

Alkhawajah AM, Larbi E, al‐Gindan Y, Abahussein A, Jain S. Treatment of cutaneous leishmaniasis with antimony: intramuscular versus intralesional administration. Annals of Tropical Medicine and Parasitology 1997;91(8):899‐905. [PUBMED: 9579209]CENTRAL

Alrajhi 2002 {published data only}

Alrajhi AA, Ibrahim EA, De Vol EB, Khairat M, Faris RM, Maguire JH. Fluconazole for the treatment of cutaneous leishmaniasis caused by leishmania major. New England Journal of Medicine 2002;346(12):891‐5. [PUBMED: 11907288]CENTRAL

Alsaleh 1995 {published data only}

Alsaleh QA, Dvorak R, Nanda A. Ketoconazole in the treatment of cutaneous leishmaniasis in Kuwait. International Journal of Dermatology 1995;34(7):495‐7. [PUBMED: 7591417]CENTRAL

Aronson 2010 {published data only}

Aronson NE, Wortmann GW, Byrne WR, Howard RS, Bernstein WB, Marovich MA, et al. A randomized controlled trial of local heat therapy versus intravenous sodium stibogluconate for the treatment of cutaneous Leishmania major infection. PLoS Neglected Tropical Diseases 2010;4(3):e628. [PUBMED: 20231896]CENTRAL

Asilian 1995 {published data only}

Asilian A, Jalayer T, Whitworth JA, Ghasemi RL, Nilforooshzadeh M, Olliaro P. A randomized, placebo‐controlled trial of a two‐week regimen of aminosidine (paramomycin) ointment for treatment of cutaneous leishmaniasis in Iran. American Journal of Tropical Medicine and Hygiene 1995;53(6):648‐51. [PUBMED: 8561269]CENTRAL

Asilian 2003 {published data only}

Asilian A, Jalayer T, Nilforoosshzadeh M, Ghassemi RL, Peto R, Wayling S, et al. Treatment of cutaneous leishmaniasis with aminosidine (paromomycin) ointment: double‐blind, randomized trial in the Islamic Republic of Iran. Bulletin of the World Health Organization 2003;81(5):353‐9. [PUBMED: 12856053]CENTRAL

Asilian 2004a {published data only}

Asilian A, Sadeghinia A, Faghihi G, Momeni A. Comparative study of the efficacy of combined cryotherapy and intralesional meglumine antimoniate (Glucantime) vs. cryothrapy and intralesional meglumine antimoniate (Glucantime) alone for the treatment of cutaneous leishmaniasis. International Journal of Dermatology 2004;43(4):281‐3. [PUBMED: 15090013]CENTRAL

Asilian 2004b {published data only}

Asilian A, Sharif A, Faghihi G, Enshaeieh Sh, Shariati F, Siadat AH. Evaluation of CO laser efficacy in the treatment of cutaneous leishmaniasis. International Journal of Dermatology 2004;43(10):736‐8. [PUBMED: 15485530]CENTRAL

Asilian 2006 {published data only}

Asilian A, Davami M. Comparison between the efficacy of photodynamic therapy and topical paromomycin in the treatment of Old World cutaneous leishmaniasis: a placebo‐controlled, randomized clinical trial. Clinical & Experimental Dermatology 2006;31(5):634‐7. [PUBMED: 16780497]CENTRAL

Asilian 2014 {published data only}

Asilian A, Omrani Sh, Nilforoushzadeh MA. Comparing the effects of topical miltefosine and glucantime in treatment of cutaneous leishmaniasis. Journal of Isfahan Medical School 2014;31(269):2257‐63. CENTRAL

Ben Salah 1995 {published data only}

Ben Salah A, Zakraoui H, Zaatour A, Ftaiti A, Zaafouri B, Garraoui A, et al. A randomized, placebo‐controlled trial in Tunisia treating cutaneous leishmaniasis with paromomycin ointment. American Journal of Tropical Medicine and Hygiene 1995;53(2):162‐6. [PUBMED: 7677218]CENTRAL

Ben Salah 2009 {published data only}

Ben Salah A, Buffet PA, Morizot G, Ben Massoud N, Zâatour A, Ben Alaya N, et al. WR279,396, a third generation aminoglycoside ointment for the treatment of Leishmania major cutaneous leishmaniasis: a phase 2, randomized, double blind, placebo controlled study. PLoS Neglected Tropical Diseases 2009;3(5):e432. [PUBMED: 19415122]CENTRAL

Ben Salah 2013 {published data only}

Ben Salah A, Ben Messaoud N, Guedri E, Zaatour A, Ben Alaya N, Bettaieb J, et al. Topical paromomycin with or without gentamicin for cutaneous leishmaniasis. New England Journal of Medicine 2013;368(6):524‐32. [PUBMED: 23388004 ]CENTRAL

Bumb 2013 {published data only}

Bumb RA, Prasad N, Khandelwal K, Aara N, Mehta RD, Ghiya BC, et al. Long‐term efficacy of single‐dose radiofrequency‐induced heat therapy vs. intralesional antimonials for cutaneous leishmaniasis in India. British Journal of Dermatology 2013;168(5):1114‐9. [PUBMED: 23298394]CENTRAL

Daie Parizi 2015 {published data only}

Daie Parizi MH, Karvar M, Sharifi I, Bahrampour A, Heshmat Khah A, Rahnama Z, et al. The topical treatment of anthroponotic cutaneous leishmaniasis with the tincture of thioxolone plus benzoxonium chloride (Thio‐Ben) along with cryotherapy: A single‐blind randomized clinical trial. Dermatologic Therapy 2015;28(3):140‐6. [PUBMED: 25847678]CENTRAL

Dandashli 2005 {published data only}

Dandashi A. Treatment of cutaneous leishmaniasis with fluconazole: a randomized double‐blind, placebo‐controlled trial. Journal of the European Academy of Dermatology and Venereology : JEADV 2005;19(Suppl 2):43. CENTRAL

Dastgheib 2012 {published data only}

Dastgheib L, Naseri M, Mirashe Z. Both combined oral azithromycin plus allopurinol and intramuscular Glucantime yield low efficacy in the treatment of Old World cutaneous leishmaniasis: a randomized controlled clinical trial. International Journal of Dermatology 2012;51(12):1508‐11. [PUBMED: 23171020]CENTRAL

Dogra 1990 {published data only}

Dogra J, Aneja N, Lal BB, Mishra SN. Cutaneous leishmaniasis in India: Clinical experience with itraconazole (R51 211 Janssen). International Journal of Dermatology 1990;29(9):661‐2. [PUBMED: 2177041]CENTRAL

Dogra 1991 {published data only}

Dogra J. A double‐blind study on the efficacy of oral dapsone in cutaneous leishmaniasis. Transactions of the Royal Society of Tropical Medicine and Hygiene 1991;85(2):212‐3. [PUBMED: 1887473]CENTRAL

Dogra 1992 {published data only}

Dogra J. Cutaneous leishmaniasis in India: evaluation of oral drugs (dapsone versus itraconazole). European Journal of Dermatology 1992;2:568‐9. CENTRAL

Dogra 1996 {published data only}

Dogra J, Saxena VN. Itraconazole and leishmaniasis: a randomised double‐blind trial in cutaneous disease. International Journal for Parasitology 1996;26(12):1413‐5. [PUBMED: 9024895]CENTRAL

Ejaz 2014 {published data only}

Ejaz A, Qadir SNUR, Malik N, Bari AU. Comparison of low‐dose meglumine antimoniate/ allopurinol combination therapy with full dose meglumine antimoniate alone in the treatment of cutaneous leishmaniasis ‐ A randomized controlled trial. Journal of Pakistan Association of Dermatologists 2014;24(2):108‐114. [EMBASE: 373774478]CENTRAL

El‐Sayed 2010 {published data only}

El‐Sayed M, Anwar AE. Intralesional sodium stibogluconate alone or its combination with either intramuscular sodium stibogluconate or oral ketoconazole in the treatment of localized cutaneous leishmaniasis: a comparative study. Journal of the European Academy of Dermatology and Venereology : JEADV 2010;24(3):335‐40. [PUBMED: 19744259]CENTRAL

Emad 2011 {published data only}

Emad M, Hayati F, Fallahzadeh MK, Namazi MR. Superior efficacy of oral fluconazole 400 mg daily versus oral fluconazole 200 mg daily in the treatment of cutaneous leishmania major infection: a randomized clinical trial. Journal of the American Academy of Dermatology 2011;64(3):606‐8. [PUBMED: 21315963]CENTRAL

Esfandiarpour 2002 {published data only}

Esfandiarpour I, Alavi A. Evaluating the efficacy of allopurinol and meglumine antimoniate (Glucantime) in the treatment of cutaneous leishmaniasis. International Journal of Dermatology 2002;41(8):521‐4. [PUBMED: 12207774]CENTRAL

Faghihi 2003 {published data only}

Faghihi G, Tavakoli‐kia R. Topical paromomycin vs intralesional meglumine antimoniate in cutaneous leishmaniasis. Clinical and Experimental Dermatology 2003;28(1):13‐6. [PUBMED: 12558620]CENTRAL

Farajzadeh 2015 {published data only}

Farajzadeh S, Esfandiarpour I, Haghdoost AA, Mohammadi S, Mohebbi A, Mohebbi E, Mostafavi M. Comparison between combination therapy of oral terbinafine and cryotherapy versus systemic meglumine antimoniate and cryotherapy in cutaneous leishmaniasis: a randomized clinical trial. Iranian Journal of Parasitology 2015;10(1):1‐8. [PUBMED: 25904940]CENTRAL

Fekri 2015 {published data only}

Fekri A, Rahnama Z, Khalili M, Dookhani AP, Khazaeli P, Beigi KB. The efficacy of co‐administration of topical niosomal dapsone gel and intralesional injection of glucantime in cutaneous leishmaniasis in comparison with cryotherapy plus intralesional injection of glucantime. Journal of Kerman University of Medical Sciences 2015;22(2):117‐32. [PUBMED: 603502875]CENTRAL

Firooz 2005 {published data only}

Firooz A, Khatami A, Khamesipour A, Nassiri‐Kashani M, Behnia F, Nilforoushzadeh M, et al. Intralesional injection of 2% zinc sulphate solution in the treatment of acute Old World cutaneous leishmaniasis: a randomized, double‐blind, controlled clinical trial. Journal of Drugs in Dermatology 2005;4(1):73‐9. [PUBMED: 15696988]CENTRAL

Firooz 2006 {published data only}

Firooz A, Khamesipour A, Ghoorchi MH, Nassiri‐Kashani M, Eskandari SE, Khatami A, et al. Imiquimod in combination with meglumine antimoniate for cutaneous leishmaniasis: a randomized assessor‐blind controlled trial. Archives of Dermatology 2006;142(12):1575‐9. [PUBMED: 17178983 ]CENTRAL

Gholami 2000 {published data only}

Gholami A, Khamesipour A, Momeni A, Ghazanfari T, Nilforoushzadeh MA, Darajeh Z, et al. Treatment of cutaneous leishmaniasis with 5% garlic cream: a randomized, double‐blind study. Iranian Journal of Dermatology 2000;3(3):2‐6. [CENTRAL: CN‐00454251]CENTRAL

Harms 1991 {published data only}

Harms G, Chehade AK, Douba M, Roepke M, Mouakeh A, Rosenkaimer F, et al. A randomized trial comparing a pentavalent antimonial drug and recombinant interferon‐gamma in the local treatment of cutaneous leishmaniasis. Transactions of the Royal Society of Tropical Medicine and Hygiene 1991;85(2):214‐6. [PUBMED: 1909469]CENTRAL

Iraji 2004 {published data only}

Iraji F, Vali A, Asilian A, Shahtalebi M, Momeni AZ. Comparison of intralesionally injected zinc sulphate with meglumine antimoniate in the treatment of acute cutaneous leishmaniasis. Dermatology 2004;209(1):46‐9. [PUBMED: 15237267]CENTRAL

Iraji 2005 {published data only}

Iraji F, Sadeghinia A. Efficacy of paromomycin ointment in the treatment of cutaneous leishmaniasis: results of a double‐blind, randomized trial in Isfahan, Iran. Annals of Tropical Medicine and Parasitology 2005;99(1):3‐9. [PUBMED: 15701249]CENTRAL

Jaffar 2006 {published data only}

Jaffar H. Rifampicin in cutaneous leishmaniasis ‐ a therapeutic trial in Saudi Arabia. Journal of Pakistan Association of Dermatologists 2006;16(1):4‐9. [EMBASE: 44265597]CENTRAL

Jaffary 2010 {published data only}

Jaffary F, Nilforoushzadeh MA, Ansari N, Rahimi M. Treatment of cutaneous leishmaniasis: cassia fistula fruit gel‐intralesional glucantime Vs. placebo gel‐ intralesional glucantime combination. Tehran University Medical Journal 2010;67(10):705‐11. CENTRAL

Jaffary 2014a {published data only}

Jaffary F, Nilforoushzadeh MA, Tavakoli N, Zolfaghari B, Shahbazi F. The efficacy of Achilles millefolium topical gel along with intralesional injection of glucantime in the treatment of acute cutaneous leishmaniasis major. Advanced Biomedical Research 2014;3:111. [PUBMED: 24804185 ]CENTRAL

Jaffary 2014b {published data only}

Jaffary F, Nilforoushzadeh MA, Moradi S, Derakhshan R, Ansari N. Concentrated extracts of Cassia fistula versus intralesional injection of meglumine antimoniate in treatment of acute cutaneous leishmaniasis. Journal of Skin and Stem Cell 2014;1(1):e16631. [EMBASE: 604236618]CENTRAL

Jebran 2014 {published data only}

Jebran AF, Schleicher U, Steiner R, Wentker P, Mahfuz F, Stahl HC, et al. Rapid healing of cutaneous leishmaniasis by high‐frequency electrocauterization and hydrogel wound care with or without DAC N‐055: a randomized controlled phase IIa trial in Kabul. PLoS Neglected Tropical Diseases 2014;8(2):e2694. [PUBMED: 24551257 ]CENTRAL

Jowkar 2012 {published data only}

Jowkar F, Dehghani F, Jamshidzadeh A. Is topical nitric oxide and cryotherapy more effective than cryotherapy in the treatment of old world cutaneous leishmaniasis?. Journal of Dermatological Treatment 2012;23(2):131‐5. [PUBMED: 20964568 ]CENTRAL

Kashani 2010 {published data only}

Kashani MN, Sadr B, Nilforoushzadeh MA, Arasteh M, Babakoohi S, Firooz A. Treatment of acute cutaneous leishmaniasis with intralesional injection of meglumine antimoniate: comparison of conventional technique with mesotherapy gun. International Journal of Dermatology 2010;49(9):1034‐7. [PUBMED: 20883265 ]CENTRAL

Khatami 2012 {published data only}

Khatami A, Rahshenas M, Bahrami M, Ghoorchi MH, Eskandari SE, Sharifi I, et al. Miltefosine in treatment of cutaneous leishmaniasis: a randomized controlled trial (Poster P‐31). 6th International Dermato‐Epidemiology Association Congress, Malmo, Sweden, 26‐28 August 2012. British Journal of Dermatology 2012;167(2):e23. [EMBASE: 71050777]CENTRAL

Khatami 2013 {published data only}

Khatami A, Talaee R, Rahshenas M, Khamesipour A, Mehryan P, Tehrani S, et al. Dressings combined with injection of meglumine antimoniate in the treatment of cutaneous leishmaniasis: a randomized controlled clinical trial. PloS One 2013;8(6):e66123. [PUBMED: 23826087]CENTRAL

Kochar 2000 {published data only}

Kochar DK, Aseri S, Sharma BV, Bumb RA, Mehta RD, Purohit SK. The role of rifampicin in the management of cutaneous leishmaniasis. QJM : Monthly Journal of the Association of Physicians 2000;93(11):733‐7. [PUBMED: 11077029]CENTRAL

Kochar 2006 {published data only}

Kochar DK, Saini G, Kochar SK, Sirohi P, Bumb RA, Mehta RD, et al. A double blind, randomised placebo controlled trial of rifampicin with omeprazole in the treatment of human cutaneous leishmaniasis. Journal of Vector Borne Diseases 2006;43(4):161‐7. [PUBMED: 17175700]CENTRAL

Larbi 1995 {published data only}

Larbi EB, al‐Khawajah A, al‐gindan Y, Jain S, Abahusain A, al‐Zayer A. A randomized, double‐blind, clinical trial of topical clotrimazole versus mizonazole for treatment of cutaneous leishmaniasis in the eastern province of Saudi Arabia. American Journal of Tropical Medicine and Hygiene 1995;52(2):166‐8. [PUBMED: 7872446]CENTRAL

Layegh 2007 {published data only}

Layegh P, Yazdanpanah MJ, Vosugh EM, Pezeshkpoor F, Shakeri MT, Moghiman T. Efficacy of azitromycin versus systemic meglumine antimoniate (Glucantime) in the treatment of cutaneous leishmaniasis. American Journal of Tropical Medicine and Hygiene 2007;77(1):99‐101. [PUBMED: 17620637]CENTRAL

Layegh 2009 {published data only}

Layegh P, Pezeshkpoor F, Soruri AH, Naviafar P, Moghiman T. Efficacy of cryotherapy versus intralesional meglumine antimoniate (Glucantime) for treatment of cutaneous leishmaniasis in children. American Journal of Tropical Medicine and Hygiene 2010;80(2):172‐5. [PUBMED: 19190206]CENTRAL

Layegh 2011 {published data only}

Layegh P, Rajabi O, Jafari MR, Emamgholi Tabar Malekshah P, Moghiman T, Ashraf H, et al. Efficacy of topical liposomal amphotericin B versus intralesional meglumine antimoniate (Glucantime) in the treatment of cutaneous leishmaniasis. Journal of Parasitology Research 2011;2011:656523. [DOI: 10.1155/2011/656523; PUBMED: 22174993 ]CENTRAL

Lynen 1992 {published data only}

Lynen L, Van Damme W. Local application of diminazene aceturate: an effective treatment for cutaneous leishmaniasis?. Annales de la Société Belge de Médecine Tropicale 1992;72(1):13‐9. [PUBMED: 1567264]CENTRAL

Maleki 2012 {published data only}

Maleki M, Karimi G, Tafaghodi M, Raftari S, Nahidi Y. Comparison of intralesional two percent zinc sulfate and glucantime injection in treatment of acute cutaneous leishmaniasis. Indian Journal of Dermatology 2012;57(2):118‐22. [PUBMED: 22615508]CENTRAL

Mapar 2010 {published data only}

Mapar MA, Omidian M. Intralesional injections of metronidazole versus meglumine antimoniate for the treatment of cutaneous leishmaniasis. Jundishapur Journal of Microbiology 2010;3(2):79‐83. [EMBASE: 2010406291]CENTRAL

Mashood 2001 {published data only}

Mashhood AA, Hussain K. Efficacy of allopurinol compared with pentostam in the treatment of old world cutaneous leishmaniasis. Journal of the College of Physicians and Surgeons‐‐Pakistan : JCPSP 2001;11(6):367‐70. [EMBASE: 2001264054]CENTRAL

Mohebali 2007 {published data only}

Mohebali M, Fotouhi A, Hooshmand B, Zarei Z, Akhoundi B, Rahnema A, et al. Comparison of miltefosine and meglumine antimoniate for the treatment of zoonotic cutaneous leishmaniasis (ZCL) by a randomized clinical trial in Iran. Acta Tropica 2007;103(1):33‐40. [PUBMED: 17586452]CENTRAL

Momeni 1996 {published data only}

Momeni AZ, Jalayer T, Emamjomeh M, Bashardost N, Ghassemi RL, Meghdadi M, et al. Treatment of cutaneous leishmaniasis with itraconazole. Randomized doubl‐blind study. Archives of Dermatology 1996;132(7):784‐6. [PUBMED: 8678570]CENTRAL

Momeni 2002 {published data only}

Momeni AZ, Reiszadae MR, Aminjavaheri M. Treatment of cutaneous leishmaniasis with a combination of allopurinol and low‐dose meglumine antimoniate. International Journal of Dermatology 2002;41(7):441‐3. [PUBMED: 12121563]CENTRAL

Momeni 2003 {published data only}

Momeni AZ, Aminjavaheri M, Omidghaemi MR. Treatment of cutaneous leishmaniasis with ketoconazole cream. Journal of Dermatological Treatment 2003;14(1):26‐9. [PUBMED: 12745852]CENTRAL

Mostafavi 2013 {published data only}

Mostafavi SA, Shatalebi MA, Attar AM, Hejazi H, Mottaghinejad A, Fadaei R, et al. Preparation and evaluation of clinical effects of glucantime gel mask. Journal of Isfahan Medical School 2013;31(231):389‐99. [EMBASE: 2014593999]CENTRAL

Mujtaba 1999 {published data only}

Mujtaba G, Khalid M. Weekly vs. fortnightly intralesional meglumine antimoniate in cutaneous leishmaniasis. International Journal of Dermatology 1999;38(8):607‐9. [PUBMED: 10487452]CENTRAL

Nassiri‐Kashani 2005 {published data only}

Nassiri‐Kashani M, Firooz A, Khamesipour A, Mojtahed F, Nilforoushzadeh M, Hejazi H, et al. A randomized, double‐blind, placebo‐controlled clinical trial of itraconazole in the treatment of cutaneous leishmaniasis. Journal of the European Academy of Dermatology and Venereology: JEADV 2005;19(1):80‐3. [PUBMED: 15649196]CENTRAL

Nilforoushzadeh 2004 {published data only}

Nilforoushzadeh MA. Efficacy of combined triple therapy (paromomycin ointment, cryotherapy and intralesional Glucantime) in comparison with intralesional Glucantime for treatment of acute cutaneous leishmaniasis. Iranian Journal of Dermatology 2004;3(7):136‐9. CENTRAL

Nilforoushzadeh 2006 {published data only}

Nilforoushzadeh MA, Jaffary F, Reiszadeh MR. Comparative effect of topical trichloroacetic acid and intralesional meglumine antimoniate in the treatment of acute cutaneous leishmaniasis. International Journal of Pharmacology 2006;2(6):633‐6. [EMBASE: 2007084731]CENTRAL
Nilforoushzadeh MA, Reiszadeh MR, Jafari F. Topical trichloroacetic acid compared with intralesional glucantime injection in the treatment of acute wet cutaneous leishmaniasis: An open clinical trial. Iranian Journal of Dermatology 2003;2(6):34‐9. CENTRAL

Nilforoushzadeh 2007 {published data only}

Nilforoushzadeh MA, Jaffary F, Moradi S, Derakhshan R, Haftbaradaran E. Effect of topical honey application along with intralesional injection of glucantime in the treatment of cutanneous leishmaniasis. BMC Complementary and Alternative Medicine 2007;7:13. [PUBMED: 17466071]CENTRAL

Nilforoushzadeh 2008 {published data only}

Nilforoushzadeh MA, Jaffary F, Ansari N, Siadat AH, Nilforoushan Z, Firouz A. A comparative study between the efficacy of systemic meglumine antimoniate therapy with standard or low dose plus oral omeprazole in the treatment of cutaneous leishmaniasis. Journal of Vector Borne Diseases 2008;45(4):287‐91. [PUBMED: 19248655]CENTRAL

Nilforoushzadeh 2012 {published data only}

Nilforoushzadeh MA, Naeeni FF, Sattar N, Haftbaradaran E, Jaffary F, Askari G. The effect of intralesional meglumine antimoniate (Glucantime) versus a combination of topical trichloroacetic acid 50% and local heat therapy by non‐ablative radiofrequency on cutaneous leishmaniasis lesions. Journal of Research in Medical Sciences 2012;17(1 Suppl 1):S97‐S102. [EMBASE: 2013471627]CENTRAL

Nilforoushzadeh 2013 {published data only}

Nilforoushzadeh MA, Siadat AH, Ansari N, Haftbaradaran E, Ahmadi E. A comparison between the effects of glucantime, topical trichloroacetic acid 50% plus glucantime, and fractional laser plus glucantime on cutaneous leishmaniasis lesions. Journal of Isfahan Medical School 2013;30(221):2450‐9. [EMBASE: 2013245548]CENTRAL

Nilforoushzadeh 2014a {published data only}

Nilforoushzadeh MA, Siadat AH, Haftbaradaran E, Minaravesh M. Comparative study of the efficacy of CO2 and fraxel lasers in treatment of cutaneous leishmaniasis scars. Journal of Isfahan Medical School 2014;31(269 SPEC.ISSUE):2277‐84. [EMBASE: 2014574591]CENTRAL

Nilforoushzadeh 2014b {published data only}

Nilforoushzadeh MA, Jaffary F, Derakhsahan R, Haftbaradaran E. Comparison between intralesional meglumine antimoniate and combination of trichloroacetic acid 50% and intralesional meglumine antimoniate in the treatment of acute cutaneous leishmaniasis: a randomized clinical trial. Journal of Stem Cells 2014;1(1):e16633. [DOI: 10.5812/jssc.16633; EMBASE: 2015013267]CENTRAL

Özgöztasi 1997 {published data only}

Özgöztasi O, Baydar I. A randomized clinical trial of topical paromomycin versus oral ketoconazole for treating cutaneous leishmaniasis in Turkey. International Journal of Dermatology 1997;36(1):61‐3. [EMBASE: 1997097334]CENTRAL

Ranawaka 2010 {published data only}

Ranawaka RR, Weerakoon HS. Randomized, double‐blind, comparative clinical trial on the efficacy and safety of intralesional sodium stibogluconate and intralesional 7% hypertonic sodium chloride against cutaneous leishmaniasis caused by L. donovani. Journal of Dermatological Treatment 2010;21(5):286‐93. [PUBMED: 20438389]CENTRAL

Ranawaka 2015 {published data only}

Ranawaka RR, Weerakoon HS, Silva SH. Randomized, double‐blind, controlled, comparative study on intralesional 10% and 15% hypertonic saline versus intralesional sodium stibogluconate in Leishmania donovani cutaneous leishmaniasis. International Journal of Dermatology 2015;54(5):555‐63. [PUBMED: 25600472]CENTRAL

Reithinger 2005 {published data only}

Reithinger R, Mohsen M, Wahid M, Bismullah M, Quinnell RJ, Davies CR, et al. Efficacy of thermotherapy to treat cutaneous leishmaniasis caused by Leishmania tropica in Kabul, Afghanistan: a randomized, controlled trial. Clinical Infectious Diseases 2005;40(8):1148‐55. [PUBMED: 15791515]CENTRAL

Sadeghian 2006a {published data only}

Sadeghian G, Nilforoushzadeh MA. Effect of combination therapy with systemic glucantime and pentoxifylline in the treatment of cutaneous leishmaniasis. International Journal of Dermatology 2006;45(7):819‐21. [PUBMED: 16863518]CENTRAL

Sadeghian 2006b {published data only}

Sadeghian G, Nilfroushzadeh MA, Siadat AH. A comparison between intralesional hypertonic sodium chloride solution and meglumine antimoniate in the treatment of cutaneous leishmaniasis. Egyptian Dermatology Online Journal 2;1:8. CENTRAL

Sadeghian 2007 {published data only}

Sadeghian G, Nilfroushzadeh MA, Iraji F. Efficacy of local heat therapy by radiofrequency in the treatment of cutaneous leishmaniasis, compared with intralesional injection of meglumine antimoniate. Clinical and Experimental Dermatology 2007;32(4):371‐4. [PUBMED: 17376205]CENTRAL

Safi 2012 {published data only}

Safi N, Davis GD, Nadir M, Hamid H, Robert LL, Case AJ. Evaluation of thermotherapy for the treatment of cutaneous leishmaniasis in Kabul, Afghanistan: a randomized controlled trial. Military Medicine 2012;177(3):345‐351. [PUBMED: 22479925]CENTRAL

Salmanpour 2001 {published data only}

Salmanpour R, Handjani F, Nouhpisheh MK. Comparative study of the efficacy of oral ketoconazole with intra‐lesional meglumine antimoniate (Glucantime) for the treatment of cutaneous leishmaniasis. Journal of Dermatological Treatment 2001;12:159‐62. [PUBMED: 12243707 ]CENTRAL

Salmanpour 2006 {published data only}

Salmanpour R, Razmavar MR, Abtahi N. Comparison of intralesional meglumine antimoniate, cryotherapy and their combination in the treatment of cutaneous leishmaniasis. International Journal of Dermatology 2006;45(9):1115‐6. [PUBMED: 16961529]CENTRAL

Shamsi Meymandi 2011 {published data only}

Shamsi Meymandi S, Zandi S, Aghaie H, Heshmatkhah A. Efficacy of CO(2) laser for treatment of anthroponotic cutaneous leishmaniasis, compared with combination of cryotherapy and intralesional meglumine antimoniate. Journal of the European Academy of Dermatology and Venereology : JEADV 2011;25(5):587‐91. [PUBMED: 20666876]CENTRAL

Shanehsaz 2015 {published data only}

Shanehsaz SM, Ishkhanian S. A comparative study between the efficacy of oral cimetidine and low‐dose systemic meglumine antimoniate (MA) with a standard dose of systemic MA in the treatment of cutaneous leishmaniasis. International Jornal of Dermatology 2015;54(7):834‐8. [PUBMED: 26108265]CENTRAL
Shanehsaz SM, Ishkhanian S. Therapeutic and adverse effects of standarddose and low‐dose meglumine antimoniate during systemic treatment of Syrian cutaneous leishmaniasis patients. Journal of Pakistan Association of Dermatologists 2014;24(2):115‐121. [EMBASE: 2014549292]CENTRAL

Sharquie 1997 {published data only}

Sharquie KE, Najim RA, Farjou IB. A compararive controlled trial of intralesionally‐ administered zinc sulphate, hypertonic sodium chloride and pentavalent antimony compound against cutaneous leishmaniasis. Clinical and Experimental Dermatology 1997;22(4):169‐73. [PUBMED: 9499605]CENTRAL

Sharquie 2001 {published data only}

Sharquie KE, Najim RA, Farjou IB, Al‐timimit DJ. Oral zinc sulphate in the treatment of acute cutaneous leishmaniasis. Clinical and Experimental Dermatology 2001;26(1):21‐6. [PUBMED: 11260171]CENTRAL

Shazad 2005 {published data only}

Shazad B, Abbaszadeh B, Khamesipour A. Comparison of topical paromomycin sulfate (twice/day) with intralesional meglumine antimoniate for the treatment of cutaneous leishmaniasis caused by L. major. European Journal of Dermatology 2005;15(2):85‐7. [PUBMED: 15757817]CENTRAL

Stahl 2014 {published data only}

Stahl HC, Ahmadi F, Schleicher U, Sauerborn R, Bermejo JL, Amirih ML, et al. A randomized controlled phase IIb wound healing trial of cutaneous leishmaniasis ulcers with 0.045% pharmaceutical chlorite (DAC N‐055) with and without bipolar high frequency electro‐cauterization versus intralesional antimony in Afghanistan. BMC Infectious Diseases 2014;14:619. [PUBMED: 25420793]CENTRAL

Yazdanpanah 2011 {published data only}

Yazdanpanah MJ, Banihashemi M, Pezeshkpoor F, Khajedaluee M, Famili S, Tavakoli Rodi I, et al. Comparison of oral zinc sulfate with systemic meglumine antimoniate in the treatment of cutaneous leishmaniasis. Dermatology Research & Practice 2011;269515:1‐4. [DOI: 10.1155/2011/269515; PUBMED: 21747837]CENTRAL

Zerehsaz 1999 {published data only}

Zerehsaz F, Salmanpour R, Farhad H, Ardehali S, Panjehshahin MR, Tabei SZ, et al. A double‐blind randomized clinical trial of a topical herbal extract (Z‐HE) vs. systemic meglumine antimoniate for the treatment of cutaneous leishmaniasis in Iran. International Journal of Dermatology 1999;38(8):610‐2. [PUBMED: 10487453]CENTRAL

Alavi‐Naini 2012 {published data only}

Alavi‐Naini R, Fazaeli A, O'Dempsey T. Topical treatment modalities for old world cutaneous leishmaniasis: a review. Prague Medical Report 2012;113(2):105‐18. [PUBMED: 22691282]CENTRAL

Banihashemi 2015 {published data only}

Banihashemi M, Yazdanpanah MJ, Amirsolymani H, Yousefzadeh H. Comparison of lesion improvement in lupoid leishmaniasis patients with two treatment approaches: trichloroacetic Acid and intralesional meglumine antimoniate. Journal of Cutaneous Medicine and Surgery 2015;19(1):35‐9. [PUBMED: 25775661]CENTRAL

Bumb 2010 {published data only}

Bumb RA, Mehta RD, Ghiya BC, Jakhar R, Prasad N, Soni P, et al. Efficacy of short‐duration (twice weekly) intralesional sodium stibogluconate in treatment of cutaneous Leishmaniasis in India. British Journal of Dermatology 2010;163(4):854‐8. [PUBMED: 20500797]CENTRAL

Dogra 1986 {published data only}

Dogra J, Lal BB, Misra SN. Dapsone in the treatment of cutaneous leishmaniasis. International Journal of Dermatology 1986;25(6):398‐400. [PUBMED: 3531044]CENTRAL

Dogra 1994 {published data only}

Dogra J, Aneja N. Leishmaniasis and itraconazole: a controlled clinical trial on cutaneous subtypes. International Journal of Antimicrobial Agents 1994;4(4):309‐11. [PUBMED: 18611622]CENTRAL

Dorlo 2012 {published data only}

Dorlo TP, Balasegaram M, Beijnen JH, de Vries PJ. Miltefosine: a review of its pharmacology and therapeutic efficacy in the treatment of leishmaniasis. Journal of Antimicrobial Chemotherapy 2012;67(11):2576‐97. [PUBMED: 22833634 ]CENTRAL

El On 1992 {published data only}

el‐On J, Halevy S, Grunwald MH, Weinrauch L. Topical treatment of Old World cutaneous leishmaniasis caused by Leishmania major: a double blind control study. Journal of the American Academy of Dermatology 1992;27(2 Pt 1):227‐31. [PUBMED: 1430361]CENTRAL

Frankenburg 1993 {published data only}

Frankenburg S, Gross A, Jonas F, Klaus S. Effect of topical paromomycin on cell‐mediated immunity during cutaneous leishmaniasis. International Journal of Dermatology 1993;32(1):68‐70. [PUBMED: 8425810]CENTRAL

Kim 2009 {published data only}

Kim DH, Chung HJ, Bleys J, Ghohestani RF. Is paromomycin an effective and safe treatment against cutaneous leishmaniasis? A meta‐analysis of 14 randomized controlled trials. PLoS Neglected Tropical Diseases 2009;3(2):e381. [PUBMED: 19221595]CENTRAL

Moosavi 2005 {published data only}

Moosavi Z, Nakhli A, Rassaii S. Comparing the efficiency of topical paromomycin with intralesional meglumine antimoniate for cutaneous leishmaniasis. International Journal of Dermatology 2005;44(12):1064‐5. [PUBMED: 16409282]CENTRAL

Nilforoushzadeh 2010 {published data only}

Ali NM, Fariba J, Elaheh H, Ali N. The efficacy of 5% trichoroacetic acid cream in the treatment of cutaneous leishmaniasis lesions. Journal of Dermatological Treatment 2012;23(2):136‐9. [PUBMED: 21034291]CENTRAL

Nilforoushzadeh 2011 {published data only}

Nilforoushzadeh MA, Jaffary F, Ansari N, Moradi S, Siadat AH. The comparison between trichloroacetic acid 50% and CO2 laser in the treatment of cutaneous leishmaniasis scar. Indian Journal of Dermatology 2011;56(2):171‐3. [PUBMED: 21716542]CENTRAL

Siavash 2013 {published data only}

Shanehsaz SM, Ishkhanian S. Electrocardiographic and biochemical adverse effects of meglumine antimoniate during the treatment of Syrian cutaneous leishmaniasis. Egyptian Dermatology Journal 2013;9(1):5. CENTRAL

Singh 1995 {published data only}

Singh S, Singh R, Sundar S. Failure of ketaconazole in oriental sore in India. Journal of Chemotherapy 1995;7(4):202‐3. [PUBMED: 8904156]CENTRAL

Trau 1987 {published data only}

Trau H, Schewach‐Millet M, Shoham J, Doerner T, Shor R, Passwell JH. Topical application of human fibroblast interferon (IFN) in cutaneous leishmaniasis. Israel Journal of Medical Sciences 1987;23(11):1125‐7. [PUBMED: 3325468]CENTRAL

Farajzadeh 2016a {published data only}

Farajzadeh S, Hakimi Parizi M, Haghdoost AA, Mohebbi A, Mohammadi S, Pardakhty A, et al. Comparison between intralesional injection of zinc sulfate 2 % solution and intralesional meglumine antimoniate in the treatment of acute old world dry type cutaneous leishmaniasis: a randomized double‐blind clinical trial. Journal of Parasitic Diseases 2016;40(3):935‐9. [PUBMED: 27605813]CENTRAL

Farajzadeh 2016b {published data only}

Farajzadeh S, Heshmatkhah A, Vares B, Mohebbi E, Mohebbi A, Aflatoonian M, et al. Topical terbinafine in the treatment of cutaneous leishmaniasis: triple blind randomized clinical trial. Journal of Parasitic Diseases 2016;40(4):1159‐64. [DOI: 10.1007/s12639‐014‐0641‐1; PUBMED: 27876906]CENTRAL

Hanif 2016 {published data only}

Hanif MM, Akram K, Mustafa G. Intralesional versus oral chloroquine in cutaneous leishmaniasis: comparison of outcome, duration of treatment and total dose of drug. Journal of the College of Physicians and Surgeons Pakistan 2016;26(4):260‐2. [PUBMED: 27097693]CENTRAL

Jaffary 2016 {published data only}

Jaffary F, Nilforoushzadeh MA, Siadat A, Haftbaradaran E, Ansari N, Ahmadi E. A comparison between the effects of Glucantime, topical trichloroacetic acid 50% plus Glucantime, and fractional carbon dioxide laser plus Glucantime on cutaneous leishmaniasis lesions. Dermatology Research and Practice 2016;2016:6462804. [PUBMED: 27148363]CENTRAL

Na‐Bangchang 2016 {published data only}

Na‐Bangchang K, Ahmed O, Hussein J, Hirayama K, Kongjam P, Aseffa A, et al. Exploratory, phase II controlled trial of shiunko ointment local application twice a day for 4 weeks in Ethiopian patients with localized cutaneous leishmaniasis. Evidence‐Based Complementary and Alternative Medicine 2016;2016:5984709. [PUBMED: 27195014]CENTRAL

Rajabi 2016 {published data only}

Rajabi O, Layegh P, Hashemzadeh S, Khoddami M. Topical liposomal azithromycin in the treatment of acute cutaneous leishmaniasis. Dermatologic Therapy 2016;29(5):358‐63. [DOI: 10.1111/dth.12357; PUBMED: 27073044]CENTRAL

Refai 2016 {published and unpublished data}

Refai W, Madarasingha N, Weerasingha S, Senarath U, De Silva A, Fernandopulle R, et al. Efficacy, safety and cost‐effectiveness of thermotherapy for L. donovani‐induced cutaneous leishmaniasis: a randomized controlled clinical trial. International Journal of Infectious Diseases 2016;45:74. [CENTRAL: CN‐01142616]CENTRAL

Sattar 2012 {published data only}

Sattar FA, Ahmed F, Ahmed N, Sattar SA, Malghani MA, Choudhary MI. A double‐blind, randomized, clinical trial on the antileishmanial activity of a Morinda citrifolia (Noni) stem extract and its major constituents. Natural Product Communications 2012;7(2):195‐6. [PUBMED: 22474954]CENTRAL

ACTRN12614001288617 {unpublished data only}

ACTRN12614001288617. A clinical trial to assess the safety and effect of heat therapy in comparison to standard intra‐lesional sodium stibogluconate for cutaneous leishmaniasis [In patients with cutaneous leishmaniasis, thermotherapy was compared with standard intra‐lesional therapy with regard to efficacy and safety]. anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12614001288617 Date first received: 10 December 2014. CENTRAL

IRCT138904091159N7 {unpublished data only}

IRCT138904091159N7. Comparison between two groups in the treatment of cutaneous leishmaniasis [Comparison between the efficacy of intralesional placebo and nitric oxide releasing patch versus placebo patch and glucantime in the treatment of cutaneous leishmaniasis]. www.irct.ir/searchresult.php?id=1159&number=7 Date first received: 2 September 2013. CENTRAL

IRCT2013092414746N1 {unpublished data only}

IRCT2013092414746N1. Treatment of patients with ZCL [The effect of MJ1 (Topical dairy extract) versus routine care for treatment of cutaneous leishmaniasis (rural) in Isfahan Iran :a randomized controled clinical trial (RCTs) ‐]. apps.who.int/trialsearch/Trial2.aspx?TrialID=IRCT2013092414746N1 Date first received: 20 February 2014. CENTRAL

NCT00840359 {unpublished data only}

NCT00840359. Study of the Efficacy of Daylight Activated Photodynamic Therapy in the Treatment of Cutaneous Leishmaniasis [Phase 2 Study of the Efficacy of Daylight Activated Photodynamic Therapy in the Treatment of Cutaneous Leishmaniasis]. clinicaltrials.gov/ct2/show/NCT00840359 Date first received: 8 February 2009. CENTRAL

NCT01050777 {unpublished data only}

NCT01050777. Efficacy of Topical Liposomal Form of Drugs in Cutaneous Leishmaniasis [Pilot Study of Efficacy of Topical Nano‐liposomal Meglumine Antimoniate (Glucantime) or Paromomycin in Combination With Systemic Glucantime for the Treatment of Anthroponotic Cutaneous Leishmaniasis (ACL) Caused by Leishmania Tropica]. clinicaltrials.gov/ct2/show/NCT01050777 Date first received: 13 January 2010. CENTRAL

SLCTR/2014/028 {unpublished data only}

SLCTR/2014/028. Treatment for leishmaniasis [Randomized, double blind, controlled study on efficacy and safety of intralesional metronidazole vs intralesional sodium stibogluconate in L. donovani cutaneous leishmaniasis]. slctr.lk/trials/276 Date first received: 18 October 2014. CENTRAL

Adam 2008

Adam I, Elhardello OA, Elhadi MO, Abdalla E, Elmardi KA, Jansen FH. The antischistosomal efficacies of artesunate–sulfamethoxypyrazine–pyrimethamine and artemether–lumefantrine administered as treatment for uncomplicated Plasmodium falciparum malaria. Annals of Tropical Medicine and Parasitology 2008;102(1):39‐44. [PUBMED: 18186976]

Al‐Waiz 2004

Al‐Waiz M, Sharquie KE, Al‐Assir M. Treatment of cutaneous leishmaniasis by intralesional metronidazole. Saudi Medical Journal 2004;25(10):1512‐3. [PUBMED: 15494841]

Alrajhi 2003

Alrajhi AA. Cutaneous leishmaniasis of the Old World. Skin Therapy Letter 2003;8(2):1‐4. [PUBMED: 12728282]

Alvar 2006

Alvar J, Yactayo S, Bern C. Leishmaniasis and poverty. Trends in Parasitology 2006;22(12):552‐7. [PUBMED: 17023215]

Alvar 2012

Alvar J, Vélez ID, Bern C, Herrero M, Desjeux P, Cano J, et al. Leishmaniasis worldwide and global estimates of its incidence. PLoS One 2012;7(5):e35671. [PUBMED: 22693548]

Amin 2006

Amin SP, Phelps RG, Goldberg DJ. Mesotherapy for facial skin rejuvenation: a clinical, histologic, and electron microscopic evaluation. Dermatologic Surgery 2006;32(12):1467‐1472. [PUBMED: 17199654]

Aram 1987

Aram H, Leibovici V. Ultrasound‐induced hyperthermia in the treatment of cutaneous leishmaniasis. Cutis 1987;40(4):350‐3. [PUBMED: 3677796]

Arevalo 2007

Arevalo I, Tulliano G, Quispe A, Spaeth G, Mathlashewski, Llanos‐cuantas A, et al. Role of imiquimod and parenteral meglumine antimoniate in the intitial treatment of cutaneous lesihmaniasis. Clinical Infectious Diseases 2007;44(12):1549‐54. [PUBMED: 17516397]

Babajev 1991

Babajev KB, Babajev OG, Korepanov VI. Treatment of cutaneous leishmaniasis using a carbon dioxide laser. Bulletin of the World Health Organization 1991;69(1):103‐6. [PUBMED: 1905204]

Badaro 1990

Badaro R, Falcoff E, Badaro FS, Carvalho EM, Pedral‐Sampaio D, Barral A, et al. Treatment of visceral leishmaniasis with pentavalent antimony and interferon gamma. New England Journal of Medicine 1990;322(1):16‐21. [PUBMED: 2104665]

Baryza 1995

Baryza MJ, Baryza GA. The Vancouver Scar Scale: an administration tool and its interrater reliability. Journal of Burn Care Rehabilitation 1995;16(5):535‐8. [PUBMED: 8537427]

Bassiouny 1982

Bassiouny A, El Meshad M, Talaat M, Kutty K, Metawaa B. Cryosurgery in cutaneous leishmaniasis. British Journal of Dermatology 1982;107(4):467‐74. [PUBMED: 7126453]

Berman 1981

Berman JD, Neva FA. Effect of temperature on multiplication of Leishmania amastigotes within monocyte‐derived macrophages in vitro. American Journal of Tropical Medicine and Hygiene 1981;30(2):318‐21. [PUBMED: 7235124]

Bigby 2003

Bigby M, Williams H. Appraising Systematic Reviews and Meta‐analyses. Archives of Dermatology 2003;139(6):795‐798. [PUBMED: 12810513]

Blum 2012

Blum J, Lockwood DN, Visser L, Harms G, Bailey MS, Caumes E, et al. Local or systemic treatment for New World cutaneous leishmaniasis? Re‐evaluating the evidence for the risk of mucosal leishmaniasis. International Health 2012;4(3):153‐63. [PUBMED: 24029394]

Blum 2014

Blum J, Buffet P, Visser L, Harms G, Bailey MS, Caumes E, et al. LeishMan recommendations for treatment of cutaneous and mucosal leishmaniasis in travelers, 2014. Journal of Travel Medicine 2014;21(2):116‐29. [PUBMED: 24745041]

Bogenrieder 2003

Bogenrieder T, Lehn N, Landthaler M, Stolz W. Treatment of Old World cutaneous leishmaniasis with intralesionally injected meglumine antimoniate using a Dermojet device. Dermatology 2003;206(3):269‐72. [PUBMED: 12673089]

Borelli 1987

Borelli D. A clinical trial of itraconazole in the treatment of deep mycoses and leishmaniasis. Reviews of Infectious Diseases 1987;9(Suppl 1):S57‐63. [PUBMED: 3027848]

Bryceson 1994

Bryceson AD, Murphy A, Moody AH. Treatment of 'Old World' cutaneous leishmaniasis with aminosidine ointment: results of an open study in London. Transactions of the Royal Society of Tropical Medicine and Hygiene 1994;88(2):226‐8. [PUBMED: 8036683]

Bygbjerg 1980

Bygbjerg IC, Knudsen L, Kieffer M. Failure of rifampicin therapy to cure cutaneous leishmaniasis. Archives of Dermatology 1980;116(9):988. [PUBMED: 7416770]

Cardo 2006

Cardo LJ. Leishmania: risk to the blood supply. Transfusion 2006;46(9):1641‐45. [PUBMED: 16965594]

Cauwenberg 1986

Cauwenberg G, De Doncker P. Itraconazole (R 51 211): a clinical review of its anti mycotic activity in dermatology, gynecology, and internal medicine. Drug Development Research 1986;8:317‐23. [DOI: 10.1002/ddr.430080136]

Chalmers 2009

Chalmers I, Glasziou P. Avoidable waste in the production and reporting of research evidence. Lancet 2009;374(9683):86‐9. [PUBMED: 19525005]

Chunge 1985

Chunge CN, Gachihi G, Muigai R, Wasunna K, Rashid JR, Chulay JD, et al. Visceral leishmaniasis unresponsive to antimonial drugs. III. Successful treatment using a combination of sodium stibogluconate plus allopurinol. Transactions of the Royal Society of Tropical Medicine and Hygiene 1985;79(5):715‐8. [PUBMED: 3006296]

Crawford 2005

Crawford R, Holmes D, Meymandi S. Comparative study of the efficacy of combined imiquimod 5% cream and intralesional meglumine antimoniate versus imiquimod 5% cream and intralesional meglumine antimoniate alone for the treatment of cutaneous leishmaniasis. Journal of the American Academy of Dermatology 2005;52:S118.

Croft 2006

Croft SL, Seifert K, Yardley V. Current scenario of drug development for Leishmaniasis. Indian Journal of Medical Research 2006;123(3):399‐410. [PUBMED: 16778319]

Daie Parizi 1992

Daie Parizi MH. Treatment of cutaneous leishmaniasis with local application of the tincture of Thioxolone and benzoxonium chloride (first research in the world) at annual congress of Iranian society of pediatrics, Tehran, Iran, Oct 1992. The Annual Book of Congress 1992;1:121‐5.

Daie Parizi 1996

Daie Parizi MH, Shamsaddini S. Comparison between topical treatment with Thioxolone, Benzoxonium Chloride tincture and intralesional injection of Meglumine Antimoniate on cutaneous Leishmaniasis. Journal of Kerman University of Medical Sciences 1996;3(1):7‐14.

Davis 2003

Davies CR, Kaye P, Croft SL, Sundar S. Leishmaniasis: new approaches to disease control. BMJ 2003;326(7385):377‐82. [PUBMED: 12586674 ]

de Vries 2015

de Vries HJ, Reedijk SH, Schallig HD. Cutaneous leishmaniasis: recent developments in diagnosis and management. American Journal of Clinical Dermatology 2015;16(2):99‐109. [PUBMED: 25687688]

Delamere 2008

Delamere FM, Sladden MJ, Dobbins HM, Leonardi‐Bee J. Interventions for alopecia areata. Cochrane Database of Systematic Reviews 2008;2:CD004413. [DOI: 10.1002/14651858.CD004413.pub2]

den Boer 2011

den Boer M, Argaw D, Jannin J, Alvar J. Leishmaniasis impact and treatment access. Clinical Microbiology and Infection 2011;17(10):1471–7. [PUBMED: 21933305]

Desjeux 1996

Desjeux P. Leishmaniasis. Public health aspects and control. Clinical Dermatology 1996;14(5):417‐23. [PUBMED: 8889319]

Dorlo 2011

Dorlo TP, van Thiel PP, Schoone GJ, Stienstra Y, van Vugt M, Beijnen JH, et al. Dynamics of parasite clearance in cutaneous leishmaniasis patients treated with miltefosine. PLoS Neglected Tropical Diseases 2011;5(12):e1436. [PUBMED: 22180803]

Egger 1997

Egger M, Davey Smith G, Schneider M, Minder C. Biasin meta‐analysis detected by a simple, graphical test. BMJ 1997;315(7109):629–34. [PUBMED: 9310563]

el‐On 1985

el‐On J, Weinrauch L, Livshin R, Even‐Paz Z, Jacobs GP. Topical treatment of recurrent cutaneous leishmaniasis with ointment containing paromomycin and methylbenzethonium chloride. British Medical Journal (Clinical Research Ed.) 1985;291(6497):704‐5. [PUBMED: 3929905]

el‐Safi 1990

el‐Safi SH, Murphy AG, Bryceson AD, Neal RA. A double‐blind clinical trial of the treatment of cutaneous leishmaniasis with paromomycin ointment. Transactions of the Royal Society of Tropical Medicine and Hygiene 1990;84(5):690‐1. [PUBMED: 2278070]

Faber 2003

Faber WR, Oskam L, van Gool T, Kroon NC, Knegt‐Junk KJ, Hofwegen H, et al. Value of diagnostic techniques for cutaneous leishmaniasis. Journal of the American Academy of Dermatology 2003;49(1):70‐4. [PUBMED: 12833011]

Fatima 2005

Fatima F, Khalid A, Nazar N, Abdalla M, Mohomed H, Toum AM, et al. In vitro assessment of anti ‐ cutaneous leishmaniasis activity of some Sudanese plants. Turkiye Parazitoloji Dergisi 2005;29(1):3‐6. [PUBMED: 17167733]

FDA 2014

US Food, Drugs Administration. News and Events ‐ FDA News Release 2014. www.fda.gov/NewsEvents/Newsroom/PrressAnnouncements/ucm389671.htm (acessed prior to 10 October 2016).

Garnier 2002

Garnier T, Croft SL. Topical treatment for cutaneous leishmaniasis. Current Opinion in Investigational Drugs (London, England : 2000) 2002;3(4):538‐44. [PUBMED: 12090720]

González 2009

González U, Pinart M, Rengifo‐Pardo M, Macaya A, Alvar J, Tweed JA. Interventions for American cutaneous and mucocutaneous leishmaniasis. Cochrane Database of Systematic Reviews 2009, Issue 2. [DOI: 10.1002/14651858.CD004834.pub2]

González 2010

González U, Pinart M, Reveiz L, Rengifo‐Pardo M, Tweed J, Macaya A, et al. Designing and reporting clinical trials on treatments for cutaneous leishmaniasis. Clinical Infectious Diseases 2010;51(4):409‐19. [PUBMED: 20624067]

González 2015

González U, Pinart M, Sinclair D, Firooz A, Enk C, Vélez ID, et al. Vector and reservoir control for preventing leishmaniasis. Cochrane Database of Systematic Reviews 2015, Issue 8. [DOI: 10.1002/14651858.CD008736.pub2]

Goodman 2007

Goodman AC. Beware the "Texas sharp shooter" in rate ratios of progression. BMJ 2007;334(7591):440. [PUBMED: 17332546]

Gradoni 2017

Gradoni L, López‐Vélez R, Mokni M. World Health Organization. Manual on case management and surveillance of the leishmaniasis in the WHO European Region. www.who.int/leishmaniasis/resources/EURO_WHO_Leish_manual_on_case_management_and_surveillance_9789289052511_2017.pdf?ua=1 (accessed prior to 15 November 2017).

Guyatt 2008

Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck‐Ytter Y, Alonso‐Coello P, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008;336(7650):924‐6. [PUBMED: 18436948]

Hepburn 2001

Hepburn NC. Management of cutaneous leishmaniasis. Current Opinion in Infectious Diseases 2001;14(2):151‐4. [PUBMED: 11979125]

Herwaldt 1999

Herwaldt BL. Leishmaniasis. Lancet 1999;354(9185):1191‐9. [PUBMED: 10513726]

Higgins 2003

Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ 2003;327(7414):557‐60. [PUBMED: 12958120]

Higgins 2011

Higgins JP, Altman DG, Gotzsche PC, Juni P, Moher D, Oxman AD, et al. The Cochrane Collaboration's tool for assessing risk of bias in randomised trials. BMJ 2011;343:d5928. [PUBMED: 22008217]

Ingram 2015

Ingram JR, Woo PN, Chua SL, Ormerod AD, Desai N, Kai AC, et al. Interventions for hidradenitis suppurativa. Cochrane Database of Systematic Reviews 2015, Issue 10. [DOI: 10.1002/14651858.CD010081.pub2]

Jha 1983

Jha TK. Evaluation of allopurinol in the treatment of kala‐azar occurring in North Bihar, India. Transactions of the Royal Society of Tropical Medicine and Hygiene 1983;77(2):204‐7. [PUBMED: 6868102]

Jiang 2002

Jiang S, Meadows J, Anderson SA, Mukkada AJ. Antileishmanial activity of the antiulcer agent omeprazole. Antimicrobial agents and chemotherapy 2002;46(8):2569‐74. [PUBMED: 12121934]

Junaid 1986

Junaid AJ. Treatment of cutaneous leishmaniasis with infrared heat. International Journal of Dermatology 1986;25(7):470‐2. [PUBMED: 3771049]

Kager 1981

Kager PA, Rees PH, Wellde BT, Hockmeyer WT, Lyerly WH. Allopurinol in the treatment of visceral leishmaniasis. Transactions of the Royal Society of Tropical Medicine and Hygiene 1981;75(4):556‐9. [PUBMED: 6275579]

Karamian 2007

Karamian M, Motazedian MH, Mehrabani D, Gholami K. Leishmania major infection in a patient with visceral leishmaniasis: treatment with Amphotericin B. Parasitology Research 2007;101(5):1431‐4. [PUBMED: 17659388]

Keiser J 2007

Keiser J, Utzinger J. Artemisinins and synthetic trioxolanes in the treatment of helminth infections. Current Opinion in Infectious Diseases 2007;20(6):605–12. [PUBMED: 17975411]

Kermode 2004

Kermode M. Unsafe injections in low‐income country health settings: need for injection safety promotion to prevent the spread of blood‐borne viruses. Health Promotion International 2004;19(1):95‐103. [PUBMED: 14976177]

Khalid 2005

Fatima F, Khalid A, Nazar N, Abdalla M, Mohomed H, Toum AM, et al. In vitro assessment of anti ‐cutaneous leishmaniasis activity of some Sudanese plants. Türkiye Parazitoloji Dergisi 2005;29(1):3‐6. [PUBMED: 17167733]

Khatami 2007

Khatami A, Firooz A, Gorouhi F, Dowlati Y. Treatment of acute Old World cutaneous leishmaniasis: a systematic review of the randomized controlled trials. Journal of the American Academy of Dermatology 2007;57(2):335.e1‐29. [PUBMED: 17337090]

Laguna 1999

Laguna F, López‐Vélez R, Pulido F, Salas A, Torre‐Cisneros J, Torres E, et al. Treatment of visceral leishmaniasis in HIV‐infected patients: a randomized trial comparing meglumine antimoniate with amphotericin B. Spanish HIV‐Leishmania Study Group. AIDS 1999;13(9):1063‐9. [PUBMED: 10397536]

Leibovici 1986

Leibovici V, Aram H. Cryotherapy in acute cutaneous leishmaniasis. International Journal of Dermatology 1986;25(7):473‐5. [PUBMED: 3533799]

Lessa 2001

Lessa HA, Machado P, Lima F, Cruz AA, Bacellar O, Guerreiro J, et al. Successful treatment of refractory mucosal leishmaniasis with pentoxifylline plus antimony. American Journal of Tropical Medicine and Hygiene 2001;65(2):87‐9. [PUBMED: 11508396]

Mapar 2001

Mapar MA, Kavoosi H, Dabbagh MA. Assessment of the effect of topical opium in treatment of cutaneous leishmaniasis. Iranian Journal of Dermatology 2001;4(16):23‐8.

Masmoudi 2013

Masmoudi A, Hariz W, Marrekchi S, Amouri M, Turki H. Old World cutaneous leishmaniasis: diagnosis and treatment. Journal of Dermatological Case Reports 2013;7(2):31‐41. [PUBMED: 23858338]

Meawad 1997

Meawad OB. Selective heat therapy in cutaneous leishmaniasis: a preliminary experience using the 585 nm pulsed dye laser. Journal of the European Academy of Dermatology and Venereology: JEADV 1997;8(3):241‐4. [EMBASE: 27286057]

Migdal 2011

Migdal C, Serres M. Reactive oxygen species and oxidative stress. Medical Science 2011;27(4):405‐412. [PUBMED: 21524406]

Minodier 2007

Minodier P, Parola P. Cutaneous leishmaniasis treatment. Travel Medicine and Infectious Disease 2007;5(3):150‐8. [PUBMED: 17448941]

Modabber 2007

Modabber F, Buffet PA, Torreele E, Milon G, Croft SL. Consultative meeting to develop a strategy for treatment of cutaneous leishmaniasis. Institute Pasteur, Paris. 13‐15 June, 2006. Kinetoplastid Biology and Disease 2007;6:3. [PUBMED: 17456237]

Monge‐Maillo 2013

Monge‐Maillo B, López‐Vélez R. Therapeutic options for old world cutaneous leishmaniasis and new world cutaneous and mucocutaneous leishmaniasis. Drugs 2013;73(17):1889‐920. [PUBMED: 24170665]

Monge‐Maillo 2015

Monge‐Maillo B, López‐Vélez R. Miltefosine for visceral and cutaneous leishmaniasis: drug characteristics and evidence‐based treatment recommendations. Clinical Infectious Diseases 2015;60(9):1398‐404. [PUBMED: 25601455]

Moore 2001

Moore OA, Smith LA, Campbell F, Seers K, McQuay KJ, Moore RA. Systematic review of the use of honey as a wound dressing. BMC Complementary and Alternative Medicine 2001;1:2. [PUBMED: 11405898]

Moskowitz 1999

Moskowitz PF, Kurban AK. Treatment of cutaneous leishmaniasis: Retrospective and advances for the 21st century. Clinical Dermatology 1999;17(3):305‐15. [PUBMED: 10384870]

Mosleh 2008

Mosleh IM, Geith E, Natsheh L, Schönian G, Abotteen N, Kharabsheh S. Efficacy of a weekly cryotherapy regimen to treat Leishmania major cutaneous leishmaniasis. Journal of the American Academy of Dermatology 2008;58(4):617‐24. [PUBMED: 18249466]

Munir 2008

Munir A, Janjua SA, Hussain I. Clinical efficacy of intramuscular meglumine antimoniate alone and in combination with intralesional meglumine antimoniate in the treatment of old world cutaneous leishmaniasis. Acta Dermatovenerologica Croatica: ADC 2008;16(2):60‐4. [PUBMED: 18541100]

Musa 2005

Musa AM, Khalil EA, Mahgoub FA, Hamad S, Elkadaru AM, El Hassan AM. Efficacy of liposomal amphotericin B (AmBisome) in the treatment of persistent post‐kala‐azar dermal leishmaniasis (PKDL). Annals of Tropical Medicine and Parasitology 2005;99(6):563‐9. [PUBMED: 16156969]

Najim 1998

Najim RA, Sharquie KE, Farjou IB. Zinc sulphate in the treatment of cutaneous leishmaniasis: an in vitro and animal study. Memórias do Instituto Oswaldo Cruz 1998;93(6):831‐7. [PUBMED: 9921312]

Neva 1984

Neva FA, Petersen EA, Corsey R, Bogaert H, Martinez D. Observations on local heat treatment for cutaneous leishmaniasis. American Journal of Tropical Medicine and Hygiene 1984;33(5):800‐4. [PUBMED: 6091468]

Olliaro 2013

Olliaro P, Vaillant M, Arana B, Grogl M, Modabber F, Magill A, et al. Methodology of clinical trials aimed at assessing interventions for cutaneous leishmaniasis. PLoS Neglected Tropical Diseases 2013;7(3):e2130. [PUBMED: 23556016]

Pace 2014

Pace D. Leishmaniasis. Journal of Infection 2014;69(Suppl 1):S10‐S18. [PUBMED: 25238669]

Passwell 1986

Passwell JH, Shor R, Shoham J. The enhancing effect of interferon‐beta and ‐gamma on the killing of Leishmania tropica major in human mononuclear phagocytes in vitro. Journal of Immunology 1986;136(8):3062‐6. [PUBMED: 3082979]

Pieper 2003

Pieper B, Caliri MH. Nontraditional wound care: A review of the evidence for the use of sugar, papaya/papain, and fatty acids. Journal of Wound, Ostomy, and Continence Nursing 2003;30(4):175–83. [PUBMED: 12851592]

Pigott 2014

Pigott DM, Golding N, Messina JP, Battle KE, Duda KA, Balard Y, et al. Global database of leishmaniasis occurrence locations, 1960‐2012. Scientific Data 2014;30(1):140036. [PUBMED: 25984344]

Ponte‐Sucre 2003

Ponte‐Sucre A. Physiological consequences of drug resistance in Leishmania and their relevance for chemotherapy. Kinetoplastid Biology and Disease 2003;2(1):14. [PUBMED: 14613496]

Ranawaka 2011

Ranawaka RR, Weerakoon HS, Opathella N. Liquid nitrogen cryotherapy on Leishmania donovani cutaneous leishmaniasis. Journal of Dermatological Treatment 2011;22(4):241‐5. [PUBMED: 20818996]

Rathi 2005

Rathi SK, Pandhi RK, Chopra P, Khanna N. Post‐kala‐azar dermal leishmaniasis: a histopathological study. Indian Journal of Dermatology, Venereology and Leprology 2005;71(4):250‐53. [PUBMED: 16394433]

Reithinger 2005b

Reithinger R, Aadil K, Kolaczinski J, Mohsen M, Hami S. Social impact of leishmaniasis, Afghanistan. Emerging Infectious Diseases 2005;11(4):634‐6. [PUBMED: 15834984]

Reithinger 2007

Reithinger R, Dujardin JC, Louzir H, Pirmez C, Alexander B, Brooker S. Cutaneous leishmaniasis. Lancet Infectious Diseases 2007;7(9):581‐96. [PUBMED: 17714672]

Rodriguez 1990

Rodriguez ME, Inguanzo P, Ramos A, Perez J. Treatment of cutaneous leishmaniasis with CO2 laser radiation. Revista Cubana de Medicina Tropical 1990;42(2):197‐202. [PUBMED: 2128547]

Rodriguez‐Cuartero 1990

Rodriguez‐Cuartero A, Pérez‐Blanco FJ, López‐Fernández A. Co‐trimoxazole for visceral leishmaniasis. Infection 1990;18(1):40. [PUBMED: 2312176]

Rohrich 2003

Rohrich RJ, Janis EJ, Reisman NR. Use of off‐label and non‐approved drugs and devices in plastic surgery. Plastic and Reconstructive Surgery 2003;112(1):241‐243. [PUBMED: 12832901]

Saab 2015

Saab M, El Hage H, Charafeddine K, Habib RH, Khalifeh I. Diagnosis of cutaneous leishmaniasis: why punch when you can scrape?. American Journal of Tropical Medicine and Hygiene 2015;92(3):518‐22. [PUBMED: 25561563]

Sacks 1983

Sacks DL, Barral A, Neva F. Thermosensitivity patterns of Old vs. New World cutaneous strains of Leishmania growing within mouse peritoneal macrophages in vitro. American Journal of Tropical Medicine and Hygiene 1983;32(2):300‐4. [PUBMED: 6837841]

Sampaio 1960

Sampaio SA, Godoy JT, Paiva L, Dillon NL, da Lacaz CS. The treatment of American (mucocutaneous) leishmaniasis with amphotericin B. Archives of Dermatology 1960;82:627‐35. [PUBMED: 13745957]

Sampaio 1997

Sampaio RN, Marsden PD. Treatment of the mucosal form of leishmaniasis without response to glucantime, with liposomal amphotericin B. Revista da Sociedade Brasileira de Medicina Tropical 1997;30(2):125‐8. [PUBMED: 9148335]

Savioli 2006

Savioli L, Engels D, Daumerie D, Jannin J, Alvar J, Asiedu K, et al. Response from Savioli and colleagues from the Department of Neglected Tropical Diseases, World Health Organization. PLoS medicine 2006;3(6):e283. [PUBMED: 16789805]

Schallig 2002

Schallig HD, Oskam L. Molecular biological applications in the diagnosis and control of leishmaniasis and parasite identification. Tropical Medicine and International Health 2002;7(8):641‐51. [PUBMED: 12167091]

Schmidt‐Ott 1999

Schmidt‐Ott R, Klenner T, Overath P, Aebischer T. Topical treatment with hexadecylphosphocholine (Miltex) efficiently reduces parasite burden in experimental cutaneous leishmaniasis. Transactions of the Royal Society of Tropical Medicine and Hygiene 1999;93(1):85‐90. [PUBMED: 10492799]

Schork 1967

Schork MA, Ramington RD. The determination of sample size in disease studies in which drop out or non‐adherence is a problem. Journal of Chronic Diseases 1967;20(4):233‐9. [PUBMED: 6023231]

Schulz 2010

Schulz KF, Altman DG, Moher D, for the CONSORT Group. CONSORT 2010 Statement: updated guidelines for reporting parallel group randomized trials. Annals of Internal Medicine 2010;152(11):726‐32. [DOI: 10.7326/0003‐4819‐152‐11‐201006010‐00232]

Sharifi 2010

Sharifi I, Fekri AR, Aflatoonian MR, Khamesipour A, Mahboudi F, Dowlati Y, et al. Leishmaniasis recidivans among school children in Bam, South‐east Iran, 1994‐2006. International Journal of Dermatology 2010;49(5):557‐561. [PUBMED: 20534092]

Sharquie 1988

Sharquie KE, Al‐Talib K, Chu AC. Intralesional therapy of cutaneous leishmaniasis with sodium stibogluconate antimony. British Journal of Dermatology 1988;119(1):53‐7. [PUBMED: 2841964]

Sharquie 1995

Sharquie KE. A new intralesional therapy for cutaneousd leishmaniasis with hypertonic sodium chloride solution. Journal of Dermatology 1995;22(10):732‐7. [PUBMED: 8586751]

Sharquie 1996

Sharquie KE, Al‐Azzawi KE. Intralesional therapy for cutaneous leishmaniasis with 2% zinc sulfate solution. Journal of Pan‐Arab League of Dermatologists 1996;7:41‐6.

Sharquie 1998

Sharquie KE, Al‐Hamamy H, El‐Yassin D. Treatment of cutaneous leishmaniasis by direct current electrotherapy: the Baghdadin device. Journal of Dermatology 1998;25(4):234‐7. [PUBMED: 9609980]

Simonsen 1999

Simonsen L, Kane A, Lloyd J, Zaffran M, Kane M. Unsafe injections in the developing world and transmission of bloodborne pathogens: a review. Bulletin of the World Health Organization 1999;77(10):789‐800. [PUBMED: 10593026]

Sindermann 2006

Sindermann H, Engel J. Development of miltefosine as an oral treatment for leishmaniasis. Transactions of the Royal Society of Tropical Medicine and Hygiene 2006;100(Suppl 1):S17‐20. [PUBMED: 16730362]

Solomon 2011

Solomon M, Pavlotsky F, Leshem E, Ephros M, Trau H, Schwartz E. Liposomal amphotericin B treatment of cutaneous leishmaniasis due to Leishmania tropica. Journal of the European Academy of Dermatology and Venereology 2011;25(8):973‐7. [PUBMED: 21129042]

Soto 2004

Soto J, Arana BA, Toledo J, Rizzo N, Vega JC, Diaz A, et al. Miltefosine for new world cutaneous leishmaniasis. Clinical Infectious Diseases 2004;38(9):1266‐72. [PUBMED: 15127339]

Stojkovic 2007

Stojkovic M, Junghanss T, Krause E, Davidson RN. First case of typical Old World cutaneous leishmaniasis treated with miltefosine. International Journal of Dermatology 2007;46(4):385‐7. [PUBMED: 17442078]

Storer 2005

Storer E, Wayte J. Cutaneous leishmaniasis in Afghani refugees. Australasian Journal of Dermatology 2005;46(2):80–3. [PUBMED: 15842398]

Stotland 2009

Stotland M, Shalita AR, Kissling RF. Dapsone 5% gel: a review of its efficacy and safety in the treatment of acne vulgaris. American Journal of Clinical Dermatology 2009;10(4):221‐7. [PUBMED: 19489655]

Sundar 2007a

Sundar S, Chakravarty J, Rai VK, Agrawal N, Singh SP, Chauhan V, et al. Amphotericin B treatment for Indian visceral leishmaniasis: response to 15 daily versus alternate‐day infusions. Clinical infectious Diseases 2007;45(5):556‐61. [PUBMED: 17682988]

Sundar 2007b

Sundar S, Jha TK, Thakur CP, Sinha PK, Bhattacharya SK. Injectable paromomycin for Visceral leishmaniasis in India. New England Journal of Medicine 2007;356(25):2571‐81. [PUBMED: 17582067]

Thakur 1996

Thakur CP, Pandey AK, Sinha GP, Roy S, Behbehani K, Olliaro P. Comparison of three treatment regimens with liposomal amphotericin B (AmBisome) for visceral leishmaniasis in India: a randomized dose‐finding study. Transactions of the Royal Society of Tropical Medicine and Hygiene 1996;90(3):319‐22. [PUBMED: 8758093]

Urcayo 1982

Urcayo FG, Zaias N. Oral ketoconazole in the treatment of cutaneous leishmaniasis. International Journal of Dermatology 1982;21(7):414‐6. [PUBMED: 6290403]

Uzun 2004

Uzun S, Durdu M, Culha G, Allahverdiyev AM, Memisoglu HR. Clinical features, epidemiology, and efficacy and safety of intralesional antimony treatment of cutaneous leishmaniasis: recent experience in Turkey. Journal of Parasitology 2004;90(4):853‐9. [PUBMED: 15357081]

van Griensven 2014

van Griensven J, Carrillo E, López‐Vélez R, Lynen L, Moreno J. Leishmaniasis in immunosuppressed individuals. Clinical Microbiology and Infection 2014;20(4):286‐299. [PUBMED: 24450618]

van Zuuren 2015

van Zuuren EJ, Fedorowicz Z, Carter B, van der Linden MMD, Charland L. Interventions for rosacea. Cochrane Database of Systematic Reviews 2015, Issue 4. [DOI: 10.1002/14651858.CD003262.pub5]

Vaneau 2007

Vaneau M, Chaby G, Guillot B, Martel P, Senet P, Téot L, et al. Consensus panel recommendations for chronic and acute wound dressings. Archives of Dermatology 2007;143(10):1291‐4. [PUBMED: 17938343]

Vardy 2001

Vardy D, Barenholz Y, Naftoliev N, Klaus S, Gilead L, Frankenburg S. Efficacious topical treatment for human cutaneous leishmaniasis with ethanolic lipid amphotericin B. Transactions of the Royal Society of Tropical Medicine and Hygiene 2001;95(2):184‐6. [PUBMED: 11355557]

Weigel 2001

Weigel MM, Armijos RX. The traditional and conventional medical treatment of cutaneous leishmaniasis in rural Ecuador. Revista Panamericana de Salud Publica [Pan American Journal of Public Health] 2001;10(6):395‐404. [PUBMED: 11820108]

Weinrauch 1983a

Weinrauch L, Livshin R, El‐On J. Cutaneous leishmaniasis treatment with ketoconazole. Cutis 1983;32(3):288‐90, 294. [PUBMED: 6313298]

Weinrauch 1983b

Weinrauch L, Livshin R, Even‐Paz Z, El‐On J. Efficacy of ketoconazole in cutaneous leishmaniasis. Archives of Dermatology 1983;275(5):353‐4. [PUBMED: 6318670]

WHO 2002

World Health Organization (WHO). Leishmaniasis. World Health Report 2002. www.who.int/whr/2002/annex/en/ (accessed 2 April 2004).

WHO 2007

World Health Organization (WHO). Control of leishmaniasis. Report by the Secretariat 22 March 2007. apps.who.int/gb/archive/pdf_files/WHA60/A60_10‐en.pdf (accessed before 11 October 2016).

WHO 2008

World Health Organization (WHO). Report of the Fifth Consultative Meeting on Leishmania/HIV Co‐infection. Addis Ababa, Ethiopia, 20‐22 March 2007. www.who.int/leishmaniasis/resources/Leishmaniasis_hiv_coinfection5.pdf (accessed before 11 October 2016). [WHO/CDS/NTD/IDM/2007.5]

WHO 2010

World Health Organization (WHO). Control of the leishmaniases: report of a meeting of the WHO Expert Committee on the Control of Leishmaniases, Geneva 22‐26 March 2010. WHO technical report series 949. apps.who.int/iris/bitstream/10665/44412/1/WHO_TRS_949_eng.pdf (accessed before 11 October 2016). [ISBN 9789241209496 ]

WHO 2011

World Health Organization (WHO). Application for Inclusion of Miltefosina on WHO Model List of Essential Medicines. Submitted to the EML Secretariat for consideration. November 2010. www.who.int/selection_medicines/committees/expert/18/applications/Miltefosine_application.pdf (accessed 11 October 2016).

Wortmann 2010

Wortmann G, Zapor M, Ressner R, Fraser S, Hartzell J, Pierson J, et al. Lipsosomal amphotericin B for treatment of cutaneous leishmaniasis. American Journal of Tropical Medicine and Hygiene 2010;83(5):1028‐33. [PUBMED: 21036832]

Zakraoui 1995

Zakraoui H, Ben Salah A, Ftaiti A, Marrakchi H, Zaatour A, Zaafouri B, et al. Spontaneous course of lesions of Leishmania major cutaneous leishmaniasis in Tunisia. Annales de Dermatologie et Venereogie 1995;122(6‐7):405‐7. [PUBMED: 8526421]

Zanger 2011

Zanger P, Kotter I, Raible A, Gelanew T, Schonian G, Kremsner PG. Case report: Successful treatment of cutaneous leishmaniasis caused by Leishmania aethiopica with liposomal amphothericin B in an immunocompromised traveler returning from Eritrea. American Journal of Tropical Medicine and Hygiene 2011;84(5):692‐4. [PUBMED: 21540377]

Zeglin 2009

Zeglin O. Infectiology and Tropical Dermatology Part 17: cutaneous leishmaniasis ‐ a casuistry with after assessment [Infektiologie und Tropendermatologie ‐ Teil 17: kutaneleishmaniasis ‐ eine kasuistik mit nachbegutachtung]. Dermatology 2009;15(4):246ff.

Gonzalez 2008

González U, Pinart M, Reveiz L, Alvar J. Interventions for Old World cutaneous leishmaniasis. Cochrane Database of Systematic Reviews 2008, Issue 4. [DOI: 10.1002/14651858.CD005067.pub3]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Adam 2009

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Omdurman Hospital for Tropical Diseases, Sudan

Study period: August 2007 to March 2008 (7 months)

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: people with cutaneous leishmaniasis, confirmed microscopically by finding amastigotes in slit skin smears, if they had not received any previous treatment (Antimonial)

Exclusion criteria: pregnant women, people weighing < 10 kg, malnourished people and those with a history of allergy to sulphonamides or artemisinins

N randomised: 41 (group 1: n = 20, group 2: n = 21)

Withdrawals: 0

N assessed: 41 (100%) (group 1: n = 20, group 2: n = 21)

Mean age (SD): group 1: 30.6 years (18.0), group 2: 28.5 years (15.3)

Baseline data:

  • N lesions (mean, SD): group 1: 2.1 (1.4); group 2: 2.2 (1.3)

  • Size of lesion (median, IQR): group 1: 3.4 cm (1.5); group 2: 3.2 cm (1.3)

  • Creatinine (mean, SD): group 1: 0.9 mg/dL (0.3), group 2: 0.9 mg/dL (0.2 )

  • Alanine aminotransferase (mean, SD): group 1: 36.7 IU (9.5), group 2: 37.3 IU (4.5)

Interventions

Type of interventions:

  • Group 1: AS + SMP (100 mg artesunate + 250 mg/12.5 mg sulphamethoxypyrazine/pyrimethamine) (Co‐Arinate®; Dafra Pharma NV, Turnhout, Belgium). 4 tablets on 4 consecutive days were administered and repeated 4 times with 2‐week intervals without treatment

  • Group 2: matched placebo

Duration of intervention: 8 weeks

Co‐interventions: pentosan was administered to participants who failed treatment with the study drug as well as to participants who had received placebo tablets

Outcomes

Healing rates: disappearance and/or shrinkage of the lesions. Lesions were identified, measured and numbered by their specific location on the participant's body before treatment and after 36 days and 72 days. Reported at the end of treatment.

Adverse effects: participants were questioned about expected adverse effects for 3 days (days 5–7) following administration of the doses. These were considered drug‐related if they were not reported at presentation.

Notes

Study funding sources: Dafra Pharma NV/SA, Turnhout, Belgium

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A computer‐generated block‐randomisation, blinded to the treating physician, was used to allocate patients to the two treatment arms"

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "a double‐blind, placebo controlled clinical trial"; "blinded to the treating physician…"

Comment: not fully reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Dropouts are unclear

Selective reporting (reporting bias)

Unclear risk

Protocol not available; not registered in a prospective clinical trial registry

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Al Hamdi 2010

Methods

Study design: randomised, prospective clinical trial

Setting/location: outpatient clinic of Basra teaching hospital, south Iraq

Study period: April 2004 to March 2005 (11 months)

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania major and L tropica

Inclusion criteria: all patients with cutaneous leishmaniasis who were diagnosed clinically by the same dermatologist

Exclusion criteria: ≥ 20 lesions; pregnancy, lactation; hypersensitivity to pentavalent antimonials or local anaesthetic; serious medical illness, lesion in proximity to mucous membranes, face, or cartilage; implanted metallic devices; unwillingness to avoid procreation for at least 2 months

N randomised: 38 participants, 70 lesions: group 1 = 35, group 2 = 35

Withdrawals: group 1: 14 lesions; group 2: 8 lesions

N assessed: lesions assessed: group 1: 21 (60%); group 2: 27 (77%)

Age range: 1.5 to 64 years (1.5–45 years in group 1 and 3–64 years in group 2) with a mean of 21.1 years

Sex: 52.5% males, 47.5% females

Baseline data:

  • N lesions treated at start of study: group 1, 35; group 2, 35

  • N lesions per participant: group 1 ‐ 1:7, 2:5, 3:1, > 3:7; group 2 ‐ 1:7, 2:8, 3:2, > 3:5

  • N lesions by site ‐ face and neck: group 1: 8, group 2: 3; upper limbs: group 1: 19, group : 10; lower limbs: group 1: 7, group 2: 18; trunk group 1: 1, group 2: 4

  • Mean lesion size before study: group 1, 1.74 cm, group 2, 1.70 cm

  • Range of lesion duration before study: group 1: 1 month–1 year; group 2: 1 month–5 years

Interventions

Type of interventions:

  • Group 1: hypertonic sodium chloride solution (HSCS) (7%) (7 g dissolved in 100 mL distilled water and autoclaved)

  • Group 2: ciprofloxacin solution (2 mg/mL)

Both drugs were injected into the lesions in amounts of 0.1–0.5 mL according to the size of the lesion

Duration of intervention: 8 weeks

Outcomes

Clinical cure: resolution of active lesion with or without scarring. A scoring system was specially designed as follows. The diameter of lesions was recorded in mm using a ruler and scored as: 0 (total healing); 1 (0 cm to < 0.5 cm); 2 (0.5 cm to < 1 cm); 3 (1 cm to < 1.5 cm); 4 (1.5 cm to < 2 cm); 5 (2 cm to < 2.5 cm); or 6 (≥ 2.5 cm). The degree of induration was assessed by palpation in comparison with the participant's normal skin and given the following scores: 0, 0.5, 1, 1.5, 2, or 3. The degree of erythema was assessed visually and scored as: 0, 0.5, 1, 1.5, 2, or 3. Ulceration was scored as: 1 (present) or 0 (absent). The scores of these 4 parameters were added to give a total score for each lesion.

Time points reported: the changes in total score between weeks: lesions were assessed at the start and again at 2‐week intervals after treatment for 8 weeks: 0, 2, 4, 6, and 8 weeks of treatment. Follow‐up continued for 8 weeks until complete healing took place

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Not reported; probably an open trial

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Not reported; probably an open trial. No information on how lesions were assessed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

After excluding participants who defaulted on treatment, 21/35 lesions were analysed in group 1 (HSCS) and 27/35 in group 2 (ciprofloxacin)

Selective reporting (reporting bias)

Unclear risk

Protocol not available. Not registered in a clinical trial registry. Pre‐specified outcomes of the review were reported.

Tables were not available in the links of the journal, and .pdf does not work. No adverse effects reported in the text

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Al‐Fouzan 1991

Methods

Study design: randomised controlled trial

Setting/location: Kuwait

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L tropica or L major in the area

Inclusion criteria: positive for leishmanial parasites (amastigotes) on microscopic examination. Women of childbearing age were instructed to use potent and adequate contraceptive measures before the initiation of treatment.

Exclusion criteria: not described

N randomised: 24. Oral itraconazole: 15; placebo: 9

Withdrawals: 0

N assessed: N = 24. Oral itraconazole: 15; placebo: 9

Age range: 12‐52 years

Sex: male/female: 13/11

Baseline data: single or multiple lesions, active being nodule, nodule‐ulcerative, or ulcerative. The site of lesions including both groups was 75% on upper limbs; 46% on lower limbs; 25% on the face, and 4% on the trunk. The duration of the lesion varied between 1 and 14 months.

Interventions

Type of interventions:

  • Group 1: oral itraconazole 200 mg twice daily. There was a 12‐year‐old boy who was given a dose of 100 mg once daily (3 mg/kg per day)

  • Group 2: placebo capsules twice daily during meals.

Duration of intervention: 6‐8 weeks

Duration of follow‐up: 12 weeks post‐treatment

Outcomes

Primary outcome: percentage of participants 'cured' 2 months after treatment. The response to treatment was graded as excellent (reduction in size of lesion by 80% up to complete clearance); good (reduction in size of lesion by 50%) and poor when there is minimal or no change of lesion.

Secondary outcomes: duration of remission and percentage of people with treated lesions that recur within 6 months and 1, 2, and 3 years (for a period up to 3 months after suspension of the drug)

Adverse effects

Time points reported: 8 weeks and 12 weeks post‐treatment

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The participants were randomly divided into two groups"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Quote: "The patients were randomly divided into two groups"

No further information about allocation concealment was provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The drug and the placebo were supplied in capsules with the same shape.

No information about blinding of personnel was provided but it is not likely to add risk of bias being oral administration

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No blinding of outcome assessment was described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No missing outcome data

Selective reporting (reporting bias)

Low risk

Relevant outcomes were reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Alkhawajah 1997

Methods

Study design: randomised controlled trial

Setting/location: Al‐Ahssa, Saudi Arabia

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L major in the area

Inclusion criteria: only a few (1‐3) simple lesions, on a non‐facial site; clinically confirmed CL by direct slit smears and/or in skin‐punch biopsies of the active, infiltrated edge of a representative lesion.

Exclusion criteria: multiple or disseminated lesions, lesions aged > 6 months, pregnancy, chronic illness, immunologically compromised condition, hyperallergic reaction to the trial drugs, treatment with regular medications which may affect specific therapy, treatment with antileishmanial drugs within the previous 6 months, the presence of scars of previously healed lesions

N participants (lesions) randomised: 80. IMMA group: 40 (77); ILMA group: 40 (70)

Withdrawals: 13 (13). IMMA group: 9 (9); ILMA group: 4 (4)

N assessed: 67. IMMA group: 31 (68); ILMA group: 36 (66)

Mean age (range): range 13‐42 years. IMMA group: 29.8 years (15‐41); ILMA group: 31.5 years (13‐42)

Sex: male: 48, female: 19

Baseline data:

  • Lesion type in the IMMA group were: nodular 20%; nodular‐ulcerative 24%; flat‐ulcerative 19%; and plaque‐like 5%

  • Lesion type in the ILMA group were: nodular 24%; nodular‐ulcerative 18%; flat‐ulcerative 15%; and plaque‐like 9%

  • Lesion site (% of lesions) in the IMMA group: shoulder 7; upper arm 11; lower arm 14; elbow 1; hand 10; thigh 5; knee 4; leg 12; and foot 4

  • Lesion site (% of lesions) in the ILMA group: shoulder 5; upper arm 9; lower arm 11; elbow 2; hand 8; thigh 7; knee 3; leg 12; and foot 9

  • IMMA group: median size of lesion (MSL) 1.8 cm²; median duration of lesions before therapy (MDLBT): 72.7 days

  • ILMA group: MSL: 1.2 cm². MDLBT: 67.9 days

Interventions

Type of interventions:

  • Group 1: IMMA 15 mg/kg/d daily on 6 days/week up to 12 injections

  • Group 2: ILMA 0.2 to 0.8 mL/lesion every other day over a 30 day period or until lesion had blanched

Duration of intervention:

  • Group 1: 15 days

  • Group 2: 30 days

Duration of follow‐up: 1 month post‐treatment

Outcomes

Primary outcome: percentage of lesions 'cured' at the end of treatment

Secondary outcomes: prevention of scarring

Adverse effects

Time points reported: days 15, 30

Notes

Study funding sources: none described

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "enrolled patients were randomly assigned to one of two treatment groups"

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Quote: "enrolled patients were randomly assigned to one of two treatment groups"

Comment: no further information about allocation concealment was provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information about blinding of participants and personnel was provided but the different administration via of the drug is impossible to blind, although it is unlikely to add risk of bias

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Lesions were assessed, whenever a patient came for his or her injection (s), by an observer who was unaware of the treatment the patient was receiving."

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information about dropouts

Selective reporting (reporting bias)

Unclear risk

No information about adverse effects was provided

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Alrajhi 2002

Methods

Study design: randomised controlled trial

Setting/location: Al‐Ahsaa and Ryyadh, Saudi Arabia

Study period: 15 months

Sample size calculation: to detect a difference of 22% in the rate of healing between the placebo group and the treatment group, assuming a healing rate of 45% in the placebo group, with a power of 90% and a two‐sided type I error of 5%, 101 subjects were needed in each group. To compensate for loss to follow‐up, 25% more participants were to be enrolled in each group.

Participants

Type of Leishmania:L major 56 participants (27%)

Inclusion criteria: age > 12 years, presence of lesions parasitologically confirmed leishmaniasis, non‐use of antileishmanial therapy during previous 2 months

Exclusion criteria: pregnancy, potential for pregnancy, breastfeeding; presence of lesions on the face or ears; presence of more than 10 lesions; history of liver disease; elevated serum, creatinine concentration, abnormal results on liver‐function tests; allergy to fluconazole

N randomised: 209. 106 were assigned to the fluconazole group, and 103 were assigned to the placebo group

Withdrawals: 63 received the container of capsules at the first visit and never returned for follow‐up: 37 participants assigned to receive placebo and 26 participants of fluconazole group

One participant in the placebo group, who was therefore excluded from the analyses.

N assessed: 145. Fluconazole group: 80, placebo group: 65

Age and sex: not described

Baseline data:

  • Intervention group: MNL: 3.1, MSL: 17 mm, MDLBT: 9.2 weeks

  • Control group: MNL: 3.7, MSL: 19 mm, MDLBT: 7.7 weeks

Interventions

Type of interventions:

  • Group 1: fluconazole orally 200 mg

  • Group 2: placebo 200 mg

Duration of intervention: 6 weeks

Co‐interventions: SSG was offered during follow‐up if oral therapy was considered to have failed (14 participants in the fluconazole group and 33 in the placebo group)

Duration of follow‐up: 1 year post‐treatment

Outcomes

Healing rates:

  • Percentage of participants 'cured' 3 months after treatment

  • Speed of healing (time taken to be 'cured')

Adverse effects

Time points reported: 6 weeks, 3 months of follow‐up, 1 year post‐treatment

Notes

Baseline imbalances: because of the criteria for inclusion and the limited number of women at risk for Leishmania in the study areas, there was only one female participant. Most of the participants were foreign construction workers or farmers originally from countries where CL is not endemic. One of 5 participants was a local national.

Study funding sources: supported in part by a grant (no. 146‐1414) from Pfizer and the Ministry of Health of Saudi Arabia

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "the randomisation sequence was generated from a random‐number table"

Allocation concealment (selection bias)

Unclear risk

Quote: "The randomisation sequence was generated from a random‐number table"

Comment: no further information about allocation concealment was provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quotes: "assigned to receive either fluconazole (Diflucan, Pfizer, New York) in the form of a 200‐mg capsule once daily for six weeks or a matching placebo"

"An independent observer evaluated the rates of compliance and side effects by interviewing patients and counting their remaining capsules."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No further information about blinding of outcome assessment was provided

Incomplete outcome data (attrition bias)
All outcomes

High risk

High rate of drops out: 63/209 (30.14%). Missing outcome data imbalanced in numbers across groups. Fluconazole orally 200 mg: 26. Placebo group: 37. An ITT analysis was performed

Selective reporting (reporting bias)

Low risk

Relevant outcomes were reported

Other bias

Low risk

Other items assessed correctly reported

Alsaleh 1995

Methods

Study design: randomised controlled trial

Setting/location: Kuwait

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania spp not specified

Inclusion criteria: only the smear‐positive cases included in the study

Exclusion criteria: participants younger than 14 years and pregnant nursing women

N randomised: 33. Group 1, ketoconazole 600 mg: 18; Group 2, ketoconazole 800 mg: 15

Withdrawals: 7. Group 1: 3, Group 2: 4

N assessed: 26. Group 1: 15, Group 2: 11

Age range: 14‐66 years

Sex (male/female): 26/7

Severity of illness: 1‐8 lesions. Site of the lesions in the ketoconazole 600 mg group: 46% on the upper extremities and 24/% on the lower extremities. In the ketoconazole 800 mg: 58% on the upper extremities; 21% on the lower extremities and 21% on head and neck

Ketoconazole 600 mg: MNL: 3.56 (range 2‐8). MDLBT: 3.4 months (range 1.5‐7)

Ketoconazole 800 mg: MNL: 3.27 (range 1‐6). MDLBT: 4.5 months (range 1‐12)

Interventions

Type of interventions:

  • Group 1: ketoconazole 600 mg daily

  • Group 2: ketoconazole 800 mg daily

Duration of intervention: 6 weeks or until the participant was cured (whatever occurred early)

Duration of follow‐up: 6 months

Outcomes

Healing rates: percentage of participants 'cured' at the end of treatment ( If there was more than 90% improvement of these parameters: re‐epithelisation and decrease in the size and inflammation of the lesions, with a negative smear for Leishmania parasites)

Secondary outcomes: duration of remission and percentage of people with treated lesions that recur within 6 months

Adverse effects

Time points reported: 1, 2, 4, 6, 8 weeks post‐treatment

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

No information about allocation concealment was provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information about blinding was provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information about blinding was provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing outcome data balanced in numbers across intervention groups

Selective reporting (reporting bias)

Unclear risk

Insufficient information to judge.

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Aronson 2010

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Walter Reed Army Medical Center (WRAMC) in Washington

Study period: 24 months (2004‐6)

Sample size calculation: a sample size of 27 participants per treatment group was planned, assuming a 73% cure rate for ThermoMed (TM), 99% for SSG, controlling for a probability of a type I error at alpha = 0.05 and was predicted to have 80% power to determine a 26% difference in outcome.

Participants

Type of Leishmania: L major

Inclusion criteria: eligible participants were Department of Defense healthcare beneficiaries with parasitologically confirmed cutaneous leishmaniasis. At Walter Reed Army Medical Center (WRAMC) in Washington. All participants were likely infected in Iraq or Kuwait. All were treatment naive.

Exclusion criteria: 20 lesions; pregnancy, lactation; hypersensitivity to pentavalent antimonials or local anaesthetic; serious medical illness; lesion in proximity to mucous membranes, face, or cartilage; implanted metallic devices; unwillingness to avoid procreation for at least 2 months

N randomised: 56, IVSSG: 28. Localised TM device heat treatment: 28

Participants with clinical failure at 2 months were offered cross‐over treatment. Afer 2 months: SSG: 29 (−1, +3), TM: 25 (−3, +1).

Withdrawals: 2. TM: 1 (lesion not amenable to heat), SSG: 1 (not confirmed L major)

N assessed: 54 (96.43%) completed treatment: TM: 27 (96.43%), SSG: 27 (96.43%). 53 (94.64%) completed 12 months follow‐up: TM: 27 (96.43%), SSG: 26 (92.86%)

Median age (range): TM: 25 years (20‐53); SSG: 24 years (18‐57)

Sex: TM: males: 28 (100%), SSG: males: 27 (96%), females: 1 (4%)

Baseline data:

  • Mean number of lesions (range): TM: 2 (1‐14), SSG: 3 (1‐17)

  • Mean duration of lesions (range): TM: 126 days (45–231), SSG: 138 days (50–270)

  • Amastigotes present (%): TM: 22 (79), SSG: 18 (64)

  • Culture isoenzyme L major* (%): TM: 12/23 (52), SSG: 12/24 (50)

  • L major spp PCR positive: TM: 28 (100), SSG: 27 (96)

  • Complicated leishmaniasis, presence of significant regional adenopathy or subcutaneous nodules (%): TM: 5 (18), SSG: 8 (29)

  • Location of the target lesion: TM: head and neck 11 (12%), arms 40 (43%), legs 27 (29%), back 11 (12%), chest 5 (5%). SSG: head and neck 6 (6%), arms 65 (67%), legs 14 (14%), back 3 (3%), chest 9 (9%)

  • MSL (range): TM: 155 mm² (9–1014), SSG: 110 mm² (9–1720)

Interventions

Type of interventions:

  • Group 1: quote: "Localized ThermoMed (TM) device heat treatment: one treatment session using the ThermoMed Model 1.8 device (TM). Prior to thermotherapy, each lesion was cleansed, anaesthestized, moistened, and overlying eschar was removed. 2 trained physicians performed TM treatments. TheTM probe was placed on the skin, covering the lesion with 50uCTM treatments applied for 30 s in a grid fashion extending 4 mm into border skin. The lesion size determined the number of applications. Each lesion was then covered with a dressing (Coverlet, Beiersdorf‐Jobst, Wilton CT) that was changed daily."

  • Group 2: IVSSG 20 mg/kg/d for 10 doses (GlaxoSmithKline, UK)

Duration of intervention: TM: 1 session. SSG: 10 days.

Co‐interventions: participants randomised to thermotherapy received oral antibiotics for secondary bacterial infections of the leishmaniasis lesion(s) prior to treatment. SSG arm participants were treated concurrently with antibiotics.

Follow‐up: 2, 6, and 12–24 months post‐treatment.

Outcomes

Clinical cure of the lesions: clinical cure was defined as complete epithelialisation or visually healed at 2 ± 1 month after completion of therapy and no reactivation in 12 months after the start of treatment. Clinical failure was less than complete epithelialisation or visually not healed at 2 ± 1 month after treatment completion. Relapse failure was defined as skin lesion persistence at the treatment site or elsewhere in the period up to 12 months after start of therapy, regardless of appearance at 2 ± 1 month after treatment completion

Laboratory cure of the lesions: microbiological cure: they looked for an eradication of the infection

Time to healing: survival analysis of time to healing for the 2 treatment arms

Adverse effects: toxicity profile

Time points reported: 2, 6, 12 months. Toxicity profile: daily physician evaluations

Notes

Study funding sources: this trial was supported by Walter Reed Army Medical Center, The North Atlantic Regional Medical Command, and the US Army Medical and Materiel Development Agency.

Possible conflicts of interest: the authors have declared that no competing interests exist

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The statistician (RH) generated the randomisation plan in blocks of 4 subjects using www.randomization.com. The research pharmacist made assignments using the randomisation plan in sequential order.

Allocation concealment (selection bias)

Low risk

The allocation sequence was unavailable to investigators until completion of the trial

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Not blinded

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Sequential photographs were independently assessed by blinded leishmaniasis experts, who were clinicians experienced in the treatment of CL, with a tiebreaker assessment when needed.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2/56 withdrawals. An ITT analysis was performed

Selective reporting (reporting bias)

Low risk

Clinical Trial Registration: ClinicalTrials.gov NCT 00884377; all prespecified outcomes were reported

Other bias

Low risk

Other items assessed correctly reported

Asilian 1995

Methods

Study design: randomised controlled trial

Setting/location: 8 primary health centres around Borkhar, north of Isfahan, Iran

Study period: 14 months

Sample size calculation: not described

Participants

Type of Leishmania: infections here were thought to be caused entirely by L major parasites, although there is probably some L tropica infection within the city of Isfahan

Inclusion criteria: 2 years or older, single lesion that was parasitologically positive, < 5 cm in diameter, at least 3 cm from the eyes, lesion present < 4 months

Exclusion criteria: pregnant or nursing mothers, previously treated for leishmaniasis, intercurrent illness or a history of allergy to aminoglycoside

N randomised: 251, aminosidine group: 126 (134 lesions); placebo group: 125 (134 lesions)

Withdrawals: not described

N assessed: aminosidine group: 123 lesions; placebo group: 123 lesions

Age (years): aminosidine group: < 15 years: 114, >15 years: 12; placebo group: < 15 years: 105, > 15 years: 20

Sex (male/female): aminosidine group: 64/62; placebo group: 67/58

Baseline data:

  • 12 to 17 participants had papular lesions; 12 to 16 nodular; 76 to 79 nodule‐ulcerative; 11 to 12 flat‐ulcerative, and 13 to 20 plaque‐like lesions.

  • Site of lesion: aminosidine group: limb: 82, head: 35, trunk: 9; placebo group: limb: 90, head: 28, trunk: 7

  • PR (15% aminosidine and 10% urea): MNL: 1, MDLBT: 1.5 weeks

  • Vehicle: MNL: 1, MDLBT: < 4 weeks

Interventions

Type of interventions:

  • Group 1: paromomycin (PR) (15% aminosidine and 10% urea) in petroleum ointment twice a day

  • Group 2: vehicle

Duration of intervention: 14 days

Co‐interventions: additional treatment, usually parenteral antimony, was given if lesions were judged to have worsened (25 participants in the PR‐treated group and 28 in the placebo group)

Duration of follow‐up: 105 days after starting treatment

Outcomes

Healing rates: percentage of participants 'cured' 2.5 months after treatment. Definite cure was defined as complete epithelialisation on days 45 or 105

Adverse effects

Tertiary outcomes: microbiological or histopathological cure of skin lesion

Time points reported: 15, 45, 105 days

Notes

Study funding sources: this work was supported in part by he UNDP/World Bank/WHO Special Program for Research and Training in Tropical Diseases (TDR) and Isfahan University of Medical Sciences

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomisation of treatment was carried out in Geneva (Switzerland) but did not state how that was done.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Identical ointment tubes were numbered and allocated to consecutive eligible participants.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing outcome data balanced in numbers across intervention groups, with similar reasons for missing data across groups (16 drops out of 251, 10%)

Selective reporting (reporting bias)

Unclear risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes, including those that were pre‐specified

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Asilian 2003

Methods

Study design: randomised controlled trial

Setting/location: 3 primary health centres around Borkhar district north of Isfahan, Iran

Study period: not described

Sample size calculation: determined on the basis of a 2‐week cure rate of 50% at day 45 and unexpected 4‐week cure rate of 70%

Participants

Type of Leishmania: L major

Inclusion criteria: participants with a single parasitologically confirmed lesion, < 5 cm in diameter

Exclusion criteria: lesions duration > 4 months and < 2 years, pregnant or nursing mothers, treated previously, any intercurrent illness or history of allergy to aminoglycoside

N randomised: 233. 117 were allocated to receive 4 weeks of active treatment and 116 to receive 2 weeks of active treatment

Withdrawals: 17, 9 in 4 weeks of active treatment and 8 in 2 weeks of active treatment

N assessed: 216, 108 in each group.

Mean age (SD): 4 weeks of active treatment: 9.2 years (8.9); 2 weeks of active treatment: 9.0 years (8.8)

Sex ratio (male/female): 4 weeks of active treatment: 47/53; 2 weeks of active treatment: 44/56

Baseline data: ulcerated lesion (SD): 4 weeks of active treatment 85 (79); 2 weeks of active treatment 95 (88)

Interventions

Type of interventions:

  • Group 1: PR (aminosidine) ointment for 4 weeks n = 117. Mean number of lesions: 1.

  • Group 2: PR (aminosidine) ointment for 2 weeks followed by 2 weeks of paraffin = 116. Mean number of lesions: 1

Co‐interventions: if lesions were bad enough, they were treated with antimonate

Duration of follow‐up: 105 days after starting treatment

Outcomes

Healing rates: 'Clinical cure' was defined as > 50% re‐epithelialisation of the original lesion, and 'clinical and parasitological cure' as either complete re‐epithelialisation or clinical cure plus a parasitologically negative smear. The primary study endpoints were clinical cure and clinical and parasitological cure at day 29, when the 4 weeks of active treatment ended

Adverse effects

Tertiary outcomes: microbiological or histopathological cure of skin lesions

Time points reported: days 29, 45, 105

Notes

Study funding sources: this investigation was supported by the UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation list after participants returned their first used tube after 2 weeks

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The vehicle and active ointments looked and smelled identical.

Outcome is not likely to be influenced by the lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The clinical and parasitological evaluators were blinded to each other's assessment.

All efforts were made to reduce the introduction of bias into this study; however, duration of the lesion, which was self‐reported by the participants or their guardians, could introduce bias if the durations were significantly different in the 2 arms by chance.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing outcome data balanced in numbers across intervention groups, with similar reasons for missing data across groups.

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes, including those that were pre‐specified

Other bias

Low risk

Other items assessed correctly reported

Asilian 2004a

Methods

Study design: randomised controlled trial

Setting/location: Skin Diseases and Leishmania Research Center of Isfahan (Iran)

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania but spp not declared

Inclusion criteria: confirmed diagnosis parasitologically and clinically, lesions with duration of < 8 weeks

Exclusion criteria: participants with a history of > 8 weeks, those with allergy to antimonials, lactating or pregnant women

N randomised participants (lesions): 400 (539). Combined cryotherapy and ILMA: 100 (149), cryotherapy alone: 200 (230), ILMA alone: 100 (160)

Withdrawals: 30 participants (46 lesions)

N assessed (lesions): 370 (493). Group 1: 93 (132); group 2: 185 (210); group 3: 92 (151)

Age: range 2‐65 years. Mean: group 1, 32 years; group 2, 27 years; group 3, 25 years

Sex (male/female): 186/184. Group 1, 46/47; group 2, 95/90; group 3, 45/47

Baseline data: not described

Interventions

Type of interventions:

  • Group 1: combined cryotherapy + ILMA: cryotherapy involved the application of liquid nitrogen via a cotton swab for 10‐25 s until the lesion and 1–2 mm of surrounding normal tissue appeared frozen. Then, after thawing, ILMA was administered, enough to blanch the lesion and a 1 mm rim of surrounding normal skin (only participants in groups 1 and 3 received ILMA). Generally, 0.5‐2 cm³ of the solution (Glucantime) was required for individual lesions, depending on their size.

  • Group 2: cryotherapy alone

  • Group 3: ILMA alone

Different modalities of treatment were not given to the same participant in different lesions (i.e. each participant received ILMA alone, cryotherapy alone, or combined cryotherapy and ILMA).

Duration of intervention: fortnightly until complete cure or for up to 6 weeks

Duration of follow‐up: 6 months

Outcomes

Healing rates: percentage of participants 'cured' 2.5 months after treatment

Adverse effects: mild adverse side effects, such as postinflammatory hypopigmentation and hyperpigmentation

Tertiary outcomes: microbiological or histopathological cure of skin lesions

Notes

Baseline imbalances: in number of participants and lesions among the groups

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The 400 patients were randomly divided into three groups"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Quote: "The 400 patients were randomly divided into three groups"

Comment: no further information was provided.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Treatment doesn't appear as blinded and the different treatments are difficult to blind.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information about blinding of outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No good information about adverse effects.

Selective reporting (reporting bias)

Unclear risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes, including those that were pre‐specified.

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Asilian 2004b

Methods

Study design: randomised controlled trial

Setting/location: Skin and Leishmaniasis Research Center of Isfahan (Iran)

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: not reported

Inclusion criteria: age 7‐70 years, disease confirmed clinically and by laboratory methods, lesions present, surface area of lesions ≤ 5 cm²

Exclusion criteria: disease duration > 4 months; pregnant or breastfeeding; chronic disease; immune suppression; and sporotrichoid forms

N participants randomised (lesions): 233 (433). 123 (183) were included in the CO₂ laser group and 110 (250) in the IMMA group

Withdrawals: 59 (112). CO₂ laser group: 40 (72), IMMA group: 19 (40)

N assessed (lesions): 174 (321). CO₂ laser group: 83 (111), IMMA group: 91 (210)

Age: range 12‐60 years

Sex (male/female): 55/68 included in the CO₂ laser group; 40/70 in the IMMA group (control group)

Baseline imbalances: no

Severity Illness: in 47% of cases, participants had one lesion and in 53% of cases they had 2‐5 lesions. There were more lesions on the upper limbs (43%), and lesions were < 5 cm². In the remaining, the lesion duration was 2‐4 months.

Mean number of lesions: CO₂ laser group: 1.49; IMMA group: 2.27

Interventions

Type of interventions:

  • Group 1: CO₂ laser (30 W, continuous) was applied to the lesion and an area 2‐3 mm around it. This procedure was repeated until the ulcer bed turned brown. After completion of the procedure, the ulcer was covered with 2% erythromycin ointment.

  • Group 2: IMMA 50 mg/kg/d for 15 days and after 15 days of rest, this treatment was repeated.

Co‐interventions: in the first group, lesions were locally anaesthetised by injection of 1%–2% lidocaine

Duration of follow‐up: 24 weeks

Outcomes

Healing rates:

  • Percentage of lesions 'cured' 1.5 months after treatment

  • Speed of healing (time taken to be 'cured')

  • Prevention of scarring

Adverse effects

Notes

Study funding sources: none declared

Possible conflicts of interest: none described

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Comment: the author was contacted. The randomisation was through coin flip method.

Allocation concealment (selection bias)

Unclear risk

Comment: the author was contacted. The randomisation was through coin flip method, but the method to conceal the allocation was not described.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No data provided, but the treatment is unlikely to be blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided about blinding of outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

High risk

Imbalance in the missing data between the groups

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes, including those that were pre‐specified.

Other bias

Unclear risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Asilian 2006

Methods

Study design: randomised controlled trial

Setting/location: Department of Dermatology, Isfahan University of Medical Sciences (Iran)

Study period: September 2004 to May 2005 (8 months)

Sample size calculation: after following a formula, sample size and number of lesions would be a maximum of 20 participants and 40 lesions, respectively, for each group

Participants

Type of Leishmania: L major

Inclusion criteria: confirmation of typical CL by positive Giemsa‐stained direct smear for Leishman‐Donovan bodies

Exclusion criteria: > 2 lesions, lesions with > 20 mm induration diameter, duration of the disease > 2 months, previous use of any anti‐leishmanial treatments, pregnant or nursing women, children < 5 years of age, serious concomitant medical problems, history of seizure, photosensitivity

N participants randomised (lesions): 60 (99). Photodynamic therapy (PDT): 20 (31); topical paromomycin: 20 (35); vehicle: 20 (33)

Withdrawals (n = 3): 1 participant with one lesion in topical paromomycin group and 2 participants with 3 lesions (one participant had 2 lesions and the other had one) in vehicle group did not complete the study because they used the ointment irregularly

N assessed (lesions): 57 (95). Photodynamic therapy (PDT): 20 (31); topical paromomycin: 19 (34); placebo: 18 (30)

Age: range 5‐59 years. Mean (SD): PDT 22.2 years (15); topical paromomycin: 24.2 years (17);

vehicle 22.3 years (15)

Sex (n, (%)): PDT: female: 12 (60)/male: 8 (40); topical paromomycin: female: 8 (42.1)/male: 11 (57.9); vehicle: female: 11 (61.1)/male: 7 (38.9)

Severity of illness: the most common sites of lesions (< 10) were on the extremities.

Total number of lesions: PDT 31; topical paromomycin 34; vehicle 30

MNL: PDT 1.55; topical paromomycin 1.75, vehicle 1.65

Mean duration of lesions (SD): PDT 38 days (11); topical paromomycin 35 days (11); vehicle 36 days (11.6)

Interventions

Type of interventions:

  • Group 1: PDT ( 10% 5‐aminolevulinic acid (5‐ALA) hydrochloride in a water‐in‐oil cream, applied topically). Lesions irradiated using visible red light at 100 J/cm² per treatment session, repeated weekly for 4 weeks

  • Group 2: PR (15% PR sulphate + 12% MBCL) in a soft white paraffin‐based ointment applied topically twice daily at 1 mm thickness over the total surface of the lesion(s) for 28 days.

  • Group 3: vehicle applied twice daily at 1 mm thickness over the total surface of the lesion(s) for 28 days.

Duration of intervention: 4 weeks

Duration of follow‐up: these groups were followed for 2 months after the end of treatment

Outcomes

Healing rates: percentage of lesions 'cured' 2 months after treatment; prevention of scarring

Scale: 'complete improvement' was defined as loss of induration and other signs of inflammation, complete re‐epithelialisation and return to normal skin texture as well as 'parasitological cure', i.e. a negative Giemsa‐stained direct smear. 'Partial improvement' was considered as flattening, reduction in size and induration without complete re‐epithelialisation. All lesions showing no decrease in size or induration were regarded as 'treatment failures'.

Adverse effects

Tertiary outcomes: microbiological or histopathological cure of skin lesions

Time points reported: days 7, 14, 21, 28, 90

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The participants were randomly divided into three groups of 20 subjects each, using computer‐based randomisation."

Allocation concealment (selection bias)

Unclear risk

No information about allocation concealment.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "Subjects in both the paromomycin and placebo groups and the clinician treating them were blinded with respect to the topical treatment received. However, it was not possible to blind subjects in the PDT group."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The dermatologist was not aware of the type of the treatment that the patient was receiving."

Incomplete outcome data (attrition bias)
All outcomes

High risk

No defaults were included in the analyses. No good information about adverse effects.

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes, including those that were pre‐specified.

Other bias

Low risk

Other items assessed correctly reported

Asilian 2014

Methods

Study design: randomised, prospective, clinical trial

Setting/location: Skin and Leishmaniasis Research Center of Isfahan, Barkhar Health Center and Department of Dermatology, Isfahan University of Medical Sciences (Iran)

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: fewer than 4 lesions of < 1 month old and < 3 cm, lesions not located on the face, > 15 years of age, lesions located neither on muscles nor were sporotrichoid lesions; receiving no medication for leishmaniasis

Exclusion criteria: history of susceptibility to MA or miltefosine; pregnant or breastfeeding; immunosuppressed; had taken systemic medication interfering with p‐450 enzyme; or history of kidney, liver, and heart failure

N randomised: 64. Topical miltefosine: 32, ILMA: 32

Withdrawals: 0

N assessed: 64 (100%). Topical miltefosine: 32, ILMA: 32

Mean age (SD): 23.12 years (13.30)

Sex: topical miltefosine: 17 males (53%) and 15 females (47%). ILMA: 16 males (50%) and 16 females (50%)

Baseline data: mean (SD) size of the lesions before treatment in the group treated with miltefosine and ILMA was 4.4 cm (3.1) and 2.3 cm (2.2), respectively.

Interventions

Type of interventions:

  • Group 1: topical miltefosine (ointment 6%), once daily (28 days) in a way that the lesion was completely smeared with the ointment

  • Group 2: ILMA, twice a week (up to 28 days)

Duration of intervention: 28 days

Outcomes

Clinical cure: cure of cutaneous leishmaniasis was defined as follows:

  • Complete cure: the lesion was flat, no induration observed, epidermal crease observed.

  • Partial cure: reduction in size of the lesions, but no epidermal creases observed.

  • Uncured: clinically no reduction observed in the lesion size or even an increase in the lesion size observed.

Adverse effects: participants were questioned about expected adverse effects for 3 days (days 5–7) following administration of the doses. These were considered drug‐related if they were not reported at presentation.

Time points reported: at the end of treatment, 1 month after treatment

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The patients were randomly divided into two groups"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Open trials. Photography was done from all the lesions both at the first visit and all the follow‐up visits

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

High risk

Protocol not available; not registered; in clinical trial registry; adverse effects not reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Ben Salah 1995

Methods

Study design: randomised controlled trial

Setting/location: Sidi‐ Bouzid. Tunisia

Study period: 9 months

Sample size calculation: the ideal sample size was estimated to be 120 based on a rate of success of treatment of 80%, 30%‐45% self‐healing in the vehicle group (type I error = 0.01and type II error = 0.10), and 10%‐20% loss to follow‐up

Participants

Type of Leishmania: L major

Inclusion criteria: aged 2‐60 years, single lesion diagnosed by the presence of parasite in stained dermal smears, no previous anti‐leishmanial treatment

Exclusion criteria: known allergy, adverse reactions to aminoglycoside antibiotics, multiple lesions, an active lesion measuring > 5 cm in diameter, if their ulcerated lesion had already persisted for more than 4 months, lesions < 3 cm from the eye, who by the physician's judgment required systemic antimonial treatment: participants with serious concomitant diseases, under medication for other illnesses likely to interfere with this study; pregnant women or nursing mothers.

N randomised: 132. Paromomycin group: 66; vehicle: 66

Withdrawals: 17. Paromomycin group: 9; vehicle: 8

N assessed: 115. Paromomycin group: 57; vehicle: 58

Mean age (SD): paromomycin group: 19.2 years ( 2.31); vehicle: 18.2 years (1.65)

Sex (ratio M:F): paromomycin group: 1.04; vehicle: 1.07

Baseline data:

  • Location of the lesions: paromomycin group: upper limbs: 47.4%, lower limbs: 38.6% trunk: 5.3%, face: 8.8%. Vehicle: upper limbs: 41.4%, lower limbs: 51.7%, trunk: 3.5%, face: 3.5%

  • Description of the lesions: paromomycin group: papular: 21.0%, nodular: 21.0%, nodo‐ulcerative: 73.7%, flat and ulcerative: 5.2%. Vehicle: papular: 19.0%, nodular: 17.2% nodo‐ulcerative: 74.1%, flat and ulcerative 7.0%

  • Days from appearance of lesion to onset of treatment (mean (SD)): paromomycin group: 39.7 (2.95). Vehicle: 33.5 (2.75)

Interventions

Type of interventions:

  • Group 1: 15% PR and 10% urea in soft white paraffin ointment

  • Group 2: vehicle (10% urea in soft white paraffin)

Duration of intervention: twice daily for 14 days

Duration of follow‐up: 105 days

Outcomes

Healing rates: percentage of participants with complete re‐epithelisation of the lesion, 2.5 months after treatment initiation (105 days)

Parasitological cure: percentage of lesions with negative smear and culture, 2.5 months after treatment initiation (105 days)

Adverse effects

Time points reported: days 15, 45, 105

Notes

Study funding sources: this investigation received funding from the United Nations Development Program/World Bank/World Health Organization Special Program for Research and TraininginTropical Diseases(grant ID:TDK910677)

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The tubes containing drug or placebo were supplied by the WHO/TDR, randomly numbered, and were given in numerical order to patients as they were admitted into the study."

Allocation concealment (selection bias)

Low risk

Quote: "The tubes containing drug or placebo were supplied by the WHO/TDR, randomly numbered, and were given in numerical order to patients as they were admitted into the study."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "The code remained unknown to patients and investigators until the study had been completed"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The code remained unknown to patients and investigators until the study had been completed"

Incomplete outcome data (attrition bias)
All outcomes

High risk

No defaults were included in the analysis

Selective reporting (reporting bias)

Low risk

Relevant outcomes were reported

Other bias

Low risk

Other items assessed correctly reported

Ben Salah 2009

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Sidi Bouzid,Tunisia, and Paris, France

Study period: 22 months

Sample size calculation: the protocol calculated a sample size of 50 participants per group with 80 percent power and a Type I error rate of 5 percent to detect a 30% difference in the proportion of participants achieving CCR, assuming a CCR proportion of 35% in the vehicle group and 65 percent in WR279, 396 participants, with a 5% expected rate of loss to follow‐up

Participants

Type of Leishmania: L major, L tropica, L infantum

Inclusion criteria: aged 5‐75 years, presence of parasitologically confirmed CL, lesions that were primarily ulcerative (i.e. not purely verrucous or nodular) and measured ≥ 1 cm² and ≤ 5 cm².

Exclusion criteria: history of known or suspected hypersensitivity or idiosyncratic reactions to aminoglycoside; previous use of antileishmanial drugs (within 3 months) or nephrotoxic or ototoxic drugs; prior diagnosis of leishmaniasis; more than 5 lesions, or a lesion in the face that in the opinion of the attending dermatologist could potentially cause significant disfigurement; significant medical problems as determined by history or laboratory studies; breastfeeding and pregnancy

N randomised: 92. WR279,396: 50; vehicle: 42

Withdrawals (n = 2): WR279,396: 1; vehicle: 1

N assessed (%): 90 (95.7). WR279,396: 49 (94.2), vehicle: 41 (97.6)

Age < 18 years ‐ n (%): WR278,396: 47 (94), vehicle: 33 (79)

Sex: male: 54, female: 38

Severity of illness: N lesions: 1: 54, 2: 16, 3: 13, 4 or 5: 9

Total lesion area (median, IQR): WR279,396: 128 (85 to 223), vehicle: 154 (70 to 264)

Index lesion area (median, IQR): WR279,396: 92 (55 to 141), vehicle: 115 (50 to 172)

Leishmania spp ‐ n (%): L major: WR279,396: 32 (64), vehicle: 24 (57). L Infantum: WR279,396: 1 (2), placebo: 0 (0). L tropica: WR279,396: 1 (2), vehicle: 0 (0). Unidentified: WR279,396: 16 (32), placebo: 18 (43)

Interventions

Type of interventions:

  • Group 1: WR279,396 is an off‐white to yellowish, thick cream containing 15% (w/w) paromomycin‐sulphate (Farmitalia) and 0.5% (w/w) gentamicin‐sulphate (Schering) as active components.

  • Group 2: the vehicle consisted of a cream without the active components and trace amounts of colorings agents to match the appearance and maintain the blind.

Duration of intervention: 20 days

Outcomes

Clinical cure: defined as complete re‐epithelialisation (i.e. length 6 width of ulceration = 060) of the index lesion by day 50 or a 50% re‐epithelialisation by day 50 followed by complete re‐epithelialisation on or before day 100 with no relapse ever having occurred from day 50 through day 180. Relapse was defined as an increase in the area of ulceration relative to the previous measurement. Participants who did not complete the 180‐day period of observation were considered to have failed to achieve complete clinical response (CCR) because relapse could not be fully assessed.

Complete clinical response at the index lesion according to baseline characteristics: influence of baseline factors on effect of treatment.

Time to healing: time course of complete re‐epithelialisation measured at 20, 50, 100, 180 days since start of treatment.

Adverse effects: immediate: observed within 30 min of application. Delayed: observed just prior to next application

Notes

Study funding sources: the Office of the Surgeon General (OTSG), Chief, Human Subjects Protection Division, U.S. Army MRMC, Fort Detrick, MD 21702‐5012. IND50,098 HSRRB Protocol#1791. Co‐sponsor: Institute Pasteur, Rue du Dr. Roux, Paris, France

Possible conflicts of interest: MG has no financial competing interests

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A sequence of genuine random numbers for the randomisation procedure was obtained from the 'fourmilab.ch/hotbits' website by a member of the Department of Chemical Information, Walter Reed Army Institute of Research, Silver Spring, Maryland and purged of duplicates.

The random numbers are generated by a process which takes advantage of the inherent uncertainty in the quantum mechanical laws of nature. Specifically, they are generated by timing successive pairs of radioactive decays detected by a Geiger‐Muëller tube interfaced to a computer. This process is better than the pseudo‐random number algorithms typically used in computer programs. The randomisation of the study drugs was done by an independent group, Fischer BioServices, Rockville, Maryland a contractor to The U.S. Army Medical Research Acquisition Activity (USAMRAA), Ft. Detrick, Maryland."

Allocation concealment (selection bias)

Low risk

The randomisation of the study drugs was done by an independent group; however, allocation concealment is not fully described.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The vehicle lacked the active components and trace amounts of colorings agents to match the appearance and maintain the blind.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Investigators, who were blinded to whether participants received WR279,396 or placebo‐vehicle, evaluated lesions for clinical response on D20 (i.e. the end of the treatment period), D50 (i.e., 30 days after the conclusion of treatment), D100, and D180". Investigators measured all lesions in 2 perpendicular directions and took photographs at the following time points: prior to therapy, at the end of therapy

Incomplete outcome data (attrition bias)
All outcomes

Low risk

49 of 50 participants randomised to WR279,396 and 41 of 42 participants randomised to placebo‐vehicle completed the study. With one exception, applications of study drugs were conducted according to the protocol

Selective reporting (reporting bias)

Low risk

ClinicalTrials.gov NCT00703924. Pre‐specified outcomes were reported

Other bias

Low risk

Other items assessed correctly reported

Ben Salah 2013

Methods

Study design: randomised controlled trial

Setting/location: Sidi Bouzid, Tunisia

Study period: January 2008 to July 2011 (42 months)

Sample size calculation: the sample size of 375 participants was based on estimated rates of final clinical cure of 94% in the paromomycin – gentamicin group and 71% in the vehicle‐control group, as shown in a previous study. On the basis of these rates, a sample size of 125 participants in each of these 2 groups provided a statistical power of 99% to detect a significant difference in the rates of final clinical cure rates (94% vs 71%).

Participants

Type of Leishmania: L major

Inclusion criteria: aged 5‐65 years; good health besides cutaneous leishmaniasis if female; absence of pregnancy and lactation; the presence of ≤ 5 lesions, with an index lesion that was ulcerative; lesions measured 1‐5 cm in diameter; lesions confirmed to contain Leishmania by means of culture or microscopical examination of lesion material

Exclusion criteria: included clinically significant lymphadenopathy or mucosal involvement, against which a topical agent would not be expected to be effective

N randomised: 383 (129 were assigned to paromomycin–gentamicin; 128 were assigned to paromomycin; 126 were assigned to vehicle control)

Withdrawals: 8 participants (4, paromomycin–gentamicin; 3, paromomycin; 1, vehicle control)

N assessed: 375 participants (125 in each of the 3 groups)

Mean age (SD): 24 years (16) (paromomycin–gentamicin, 23 years (16); paromomycin, 25 years (16); vehicle 23 years (15)

Sex: male 193 (51%) (paromomycin–gentamicin, 56 (45%); paromomycin, 68 (54%); vehicle control, 69 (55%))

Baseline data:

  • Paromomycin–gentamicin: total N of lesion in group: 243. Mean (SD) area of all lesion ulcers per participants: 126 mm² (121)

  • Paromomycin: total N of lesion in group: 272. Mean (SD) area of all lesion ulcers per participants: 90 mm² (75)

  • Vehicle control: total N of lesion in group: 282. Mean (SD) area of all lesion ulcers per participants: 98 mm² (112)

Interventions

Type of interventions:

  • Group 1: paromomycin‐gentamicin topical cream were manufactured by Teva Pharmaceuticals in accordance with Good Manufacturing Practices

  • Group 2: paromomycin alone topical cream were manufactured by Teva Pharmaceuticals in accordance with Good Manufacturing Practices

  • Group 3: vehicle control manufactured by Teva Pharmaceuticals in accordance with Good Manufacturing Practices

Duration of intervention: 20 days

Outcomes

Final cure of index lesion

Total re‐epithelialisation of ulcerated lesions at 42 days

Adverse effects

Time points reported: 168 days

Notes

Study funding sources: the study was sponsored by the Office of the Surgeon General, Department of the Army

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A permuted block randomisation schema was generated using nQuery, employing random block sizes, for the first 330 randomisation numbers. As 7 subjects were randomised that did not receive treatment at the time when the new randomisation list was generated, the plan in generating the new list was to balance the assignments in the new list for these 7 subjects to achieve the 1:1:1 allocation balance overall. The second randomisation was performed using SAS Version 9.2."

Allocation concealment (selection bias)

Low risk

The randomisation of the study drugs was done by an independent group.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The placebo consisted of the vehicle without the active components and trace amounts of colorings agents to match the appearance and maintain the blind.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "The modified intention‐to‐treat population consisted of patients who received at least one dose of study treatment. We tested two hypotheses using a fixed testing‐sequence procedure with an overall two‐sided alpha level of 0.05 or less."

Selective reporting (reporting bias)

Low risk

ClinicalTrials.gov number, NCT00606580. Pre‐specified outcomes were reported

Other bias

Low risk

Other items assessed correctly reported

Bumb 2013

Methods

Study design: randomised, phase I, open‐label, clinical trial

Setting/location: Department of Dermatology at SP Medical College and PBM Hospital, Bikaner, India

Study period: June 2009 to December 2010 (19 months)

Sample size calculation: not described

Participants

Type of Leishmania: L tropica

Inclusion criteria: aged ≥ 4 years, 4 or fewer lesions, a parasitologically confirmed diagnosis of CL by demonstration of organisms (L tropica bodies) in the lesion smear or biopsy

Exclusion criteria: included lesion size > 5 cm diameter, prior treatment failure with SSG, treatment for CL within 2 months of enrolment into the study, any chronic condition that might prevent the patient from completing the study therapy and subsequent follow‐up

N randomised: 100; radiofrequency‐induced heat therapy (RFHT): 50; ILSSG: 50

Withdrawals: not described

N assessed: 100

Median (range) age: RFHT: 20 years (4‐70); ILSSG: 20.5 years (4‐85)

Sex: RFHT: male 27, female 23; ILSSG: male 20, female 30

Baseline data:

  • Number of lesions: median (range); RFHT: 1 (1‐7); ILSSG: 1 (1‐4)

  • Number of lesions per participant (%): 1 lesion: RFHT, 30 (60); ILSSG: 32 (64). 2 lesions: RFHT, 14 (28); ILSSG: 13 (26). > 2 lesions: RFHT, 6 (12); ILSSG, 5 (10)

  • Median size of lesions (range): RFHT, 2 cm (0.5‐5); ILSSG, 2 cm (0.5‐14)

  • Median duration of illness (range): RFHT, 3 months (1‐9); ILSSG, 3 months (1‐18)

Interventions

Type of interventions:

  • Group 1: the RFHT group received a single application of a controlled and localised delivery of radiofrequencies into lesions for 30–60 s depending on the thickness of the lesion. The application was performed under local anaesthesia (1% lidocaine) using a current field radiofrequency generator (ThermoMed 1.8)

  • Group 2: ILSSG group, the participants were treated with an intralesional injection of SSG (50 mg/cm² of lesion), twice a week for 7 injections

Duration of intervention: RFHT, 5 days; ILSSG, 4 weeks

Co‐interventions: all participants were prescribed oral nonsteroidal anti‐inflammatory drugs and topical antibacterial cream (fusidic acid cream) for 5 days

Outcomes

Cure rate, assessed as follows:

  • Complete cure: the total re‐epithelialisation of the lesions

  • Partial cure: decreased induration and erythema

Time points reported: 6, 8, 10, 12, 16, and 20 weeks after the initiation of treatment, and at 5, 6, 9, 12 and 18 months post‐treatment

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The patients were N randomised in a 1:1 ratio"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "The clinician who recorded healing was blinded to the modality of treatment."

Comment: not clear how the assessor was blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

No information about numbers or reasons for withdrawals during treatment. It seems that there was withdrawals only in the follow‐up: 44% at 18 months in RFHT group and 16% at 18 months in the SSG group. Losses to follow‐up were heterogeneous between groups and no ITT analyses were performed.

Selective reporting (reporting bias)

Low risk

Outcomes of interest were reported in results

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Daie Parizi 2015

Methods

Study design: randomised single blind parallel clinical trial

Setting/location: Leishmaniasis Center in Dadbin Health Care Clinic in Kerman, Iran

Study period: 15 months. Participants recruitment: 8 months (December 2012 to August 2013)

Sample size calculation: based on the data from previous study, and by considering the confidence interval of 95%, sample size was determined as 23 lesions per group. Regarding to a possible loss in follow‐up period, finally 32 lesions were allocated in each group

Participants

Type of Leishmania: L amastigote (Leishman‐Donovan bodies)

Inclusion criteria: positive direct smear or documented skin biopsy for L amastigote (Leishman‐Donovan bodies), > 2 years of age, no previous therapy for leishmaniasis, ulcerated lesions for those lesions that were included in Thio‐Ben + cryotherapy (TC) group

Exclusion criteria: pregnancy, lesions on the eyelids and lips, having more than 5 lesions, positive history of confirmed immunodeficiency disorders or immunosuppression, disease duration for more than a year, positive history of allergy or hypersensitivity to thioxolone, benzoxonium chloride, or pentavalent antimony compounds.

N randomised: 64 lesions of 47 participants, Thio‐Ben + cryotherapy (TC) group (32 lesions, 22 participants), ILMA (32 lesions, 25 participants)

Withdrawals: 16 lesions of 9 participants (10 lesions from TC and 6 from ILMA group) were removed from the study arms because of their poor adherence to the trial protocol and lost to follow‐up. Additionally, one participant (with one lesion) was excluded because of developing a hypersensitive reaction to MA in the course of the treatment.

Lesions assessed: 48 lesions in 47 participants. TC: 22, ILMA: 25

Mean age (SD): TC 23.5 years (16.4), ILMA 25.4 years (19.4)

Sex (n (%)): TC: male 6 (27.2%), female: 16 (72.7%); ILMA: male 19 (76%), female: 6 (24%)

Baseline data:

  • Location of lesion: TC: face 5 (22.7%), elbow 1 (4.5%), hand 13 (59.1%), foot 1 (4.5%), ear 1 (4.5%), arm 1 (4.5%), neck 0. ILMA: face 6 (24%), elbow 3 (12%), hand 10 (40%), foot 2 (8%), ear 1 (4%), arm 2 (8%), neck 1 (4%)

  • Mean (SD) duration of disease: TC, 4.1 months (2.9); ILMA, 3.1 months (2.4)

Interventions

Type of interventions:

  • Group 1: 1–2 mL of tincture of Thio‐Ben (depending on the size of the lesion) topically by a cotton swab that was held with a mild pressure on the lesion for about 3–4 min, every other day

  • Group 2: ILMA containing 8.1% Sb5+ (81 mg/mL), once a week with the dose of 0.5–2 mL per lesion

Co‐intervention: in addition, at the beginning, and then every 2 weeks, cryotherapy with liquid nitrogen (−195°C) was performed for all lesions in both groups

Duration of intervention: 3 months or until the lesion was cured, whichever came first

Duration of follow‐up: 6 months after the termination of the treatment to evaluate the incidence of relapse (reported at 1 month, 2 months, 5 months)

Outcomes

Clinical cure of the lesions:

  • Complete cure: complete subsidence of induration and re‐epithelialisation of the lesion

  • Partial cure: reduction of the size of lesion by 50% and more

  • Improvement: reduction of the size of lesion by less than 50%

  • No response: no significant reduction of the size of lesion

  • Deterioration: any increase in the size of the lesion

Adverse effects

Relapse: participants that developed with relapse of the lesions at the previously involved area

Time points reported: clinical cure was reported when it occurs or for a maximum of 3 months.

Adverse effects were assessed at follow‐up during therapy; they were the safety endpoints of the treatments, and 6 months after the termination of the treatment

Notes

Study funding sources: this study was supported and funded by the Vice Chancellor for Research, Kerman University of Medical Sciences. The founder had no financial or proprietary interest in any material or method used in this study and had no role in study design, data collection and analysis, or preparation of the manuscript.

Possible conflicts of interest: no conflict of interest

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised allocation was performed by blocked randomisation method with block size of 2

Allocation concealment (selection bias)

Low risk

Randomised allocation was performed by blocked randomisation method and was prepared by the analyst of the research team who had no clinical involvement in the trial.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Blinding was impossible

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Assessing the outcomes and data analyst were blinded to group assignment. The photographs were reviewed by the dermatologist of the research team who was blinded to study groups. The photographs did not contain any indicator that could help differentiate the group of the lesion.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The numbers of withdrawals were high in both groups: 31.25% (10/32) in TC group and 18.75 % (6/32) in the ILMA group. Participants were removed from the study arms because of their poor adherence to the trial protocol and lost to follow‐up. No intention‐to‐treat analysis was done.

Selective reporting (reporting bias)

Low risk

The study published reported our outcomes

Other bias

Low risk

Other items assessed correctly reported

Dandashli 2005

Methods

Study design: randomised controlled trial

Setting/location: Aleppo, Syria

Study period: June 2009 to December 2010 (19 months)

Sample size calculation: not described

Participants

Type of Leishmania: L tropica

Inclusion criteria: not described

Exclusion criteria: not described

N randomised: 79

Withdrawals: 14

N assessed: 65. 46 participants (264 lesions) in the fluconazole group and 19 participants (102 lesions) in the placebo group

Age (years): not described

Sex: not described

Baseline data: not described

Interventions

Type of interventions:

  • Group 1: fluconazole orally 200 mg/d for 6 weeks

  • Group 2: placebo orally for 6 weeks

Duration of follow‐up: not reported

Outcomes

Healing rates: percentage of lesions 'cured' (follow‐up not reported)

Adverse effects

Time points reported: not described

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

This is an abstract

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomly assigned to treatment"

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Quote: "randomly assigned to treatment"

Comment: no further information provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information about blinding was provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information about blinding was provided

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "Side effects were mild and similar in both groups"

Comment: no further information on adverse effects was provided. Withdrawals: 14/79 (17.7%) and no ITT analyses were performed

Selective reporting (reporting bias)

Unclear risk

No protocol available. Not possible to allocate to high or low risk.

Other bias

High risk

Sample size calculation and baseline comparability were not correctly reported

Dastgheib 2012

Methods

Study design: prospective randomised controlled trial

Setting/location: dermatology clinic in Saadi Hospital, an academic center in Fars Province, Iran

Study period: December 2008 to March 2010 (15 months)

Sample size calculation: not described

Participants

Type of Leishmania: L major

Inclusion criteria: participants that presented with skin lesions suspicious of CL, not receiving any previous treatment, positive for leishmaniasis with direct smears

Exclusion criteria: pregnant and nursing women; children < 12 years old; lesions on the face; disease for more than 3 months; more than 5 active lesions; participants with any serious systemic disease or previous history of sensitivity to MA, allopurinol, or azithromycin; any difficulty in laboratory results before initial treatment (CBC diff, LFT, BUN, Cr); those who refused to sign the written informed consent form

N randomised: 86 participants

Withdrawals: 14 participants (6 in azithromycin + allopurinol group and 8 IMMA group) one lost follow‐up because of adverse effect

N assessed: 71 participants (36 azithromycin + allopurinol group and 35 IMMA group)

Mean age (SD; range): 38.2 years (12.6); range 16‐64. Azithromycin + allopurinol group: 39.7 years (12.6); IMMA: 36.8 years (12.8)

Sex: 28 females and 53 males (azithromycin + allopurinol group: 13 females and 23 males; IMMA group: 15 females and 20 males

Baseline data: most participants in azithromycin + allopurinol group had more than 3 lesions (72.3%) and in the IMMA group most participants also had more than 3 lesions (65.8%)

Interventions

Type of interventions:

  • Group 1: azithromycin capsules (Tehran Chemie Pharmaceutical Company, Tehran, Iran) at a daily dose of 10 mg/kg (maximum dose of 500 mg) + allopurinol tablets (Hakim Pharmaceutical Company, Tehran, Iran) at a daily dose of 10 mg/kg (maximum doses 800 mg)

  • Group 2: IMMA (Aventis Laboratories, France) at a dose of 20 mg/kg of antimony

Duration of intervention: group 1, 2 months; group 2, 20 days.

Co‐interventions: in case of any secondary bacterial infection, participants were with oral cephalexin for 10 days after which the antileishmanial was administered

Outcomes

Cure rate, assessed as follows:

  • Complete response: complete re‐epithelialisation and relief of induration

  • Partial response: more than 50% re‐epithelialisation and decrease of induration and size of lesion

  • No response: < 50% decrease of induration and size of lesion or worsening of lesion was considered as no response

Adverse effects

Time points reported: 2 months after completing treatment

Notes

Study funding sources: funded by deputy of research, Shiraz University of Medical Sciences

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A total of 71 participants who met the inclusion criteria in the trial were randomly divided into two treatment groups according to simple even and odd number allocation"

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Quote: "Fourteen of 86 patients dropped out due to poor compliance (six patients in the combination therapy and eight in the Glucantime group). The number and reason for their withdrawal were almost the same in both treatment groups. One patient also developed GI complications and headache while taking the combination therapy of azithromycin and allopurinol; therefore, overall 71 subjects completed the study."

Comment: no ITT analyses were performed. However, withdrawals accounted for <20% and were homogeneous among the treatment groups.

Selective reporting (reporting bias)

Low risk

Our primary outcomes (cure and adverse effects) were described in Methods and reported in Results

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Dogra 1990

Methods

Study design: randomised controlled trial

Setting/location: India

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L tropica in the area

Inclusion criteria: participants with cutaneous Leishmania confirmed by the presence of L tropica bodies in the slit skin smear stained with Leishman stain.

Exclusion criteria: women of child‐bearing age

N randomised: 20

Withdrawals: 0

N assessed: 20 (100%). Intervention group: 15, control group: 5

Age: range 14‐56 years.

Baseline data: single or multiple lesions, the duration of the lesions varied from 4 to 16 weeks

Interventions

Type of interventions:

  • Group 1: itraconazole orally 4 mg/kg per day for 6 weeks (max 200 mg). MNL: 2. MDLBT: 9 weeks

  • Group 2: control group (no treatment)

Duration of follow‐up: 6 weeks, but 3 months for relapses assessment

Outcomes

Primary outcome: percentage of participants 'cured' at the end of treatment. The essential criteria for declaring the participant was cured was complete disappearance of the induration or redness in the nodular form and complete healing in the ulcerative form, accompanied by smear positivity conversion

Secondary outcomes: duration of remission and percentage of people with treated lesions that recur within 3 months

Adverse effects

Tertiary outcomes: microbiological or histopathological cure of skin lesions

Time points reported: clinical cure: 4 weeks, clinical and parasitological cure: 6 weeks

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Proportion of 2 to 4:1

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Selective reporting (reporting bias)

Unclear risk

All of the study's pre‐specified (primary and secondary) outcomes that are of interest in the review have been reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Dogra 1991

Methods

Study design: randomised controlled trial

Setting/location: India

Study period: not described

Sample size calculation: the number of subjects was determined by the standard method of Schork 1967

Participants

Type of Leishmania: L tropica in the area

Inclusion criteria: demonstration of Leishmania from skin lesion by the slit smear technique.

Exclusion criteria: pregnant women and children < 12 years old, suffering from any chronic illness, immunocompromised, allergic to sulphones, prior therapy for cutaneous Leishmania in any form, patients with scars of healed leishmanial lesions, lesions of > 4 months duration

N randomised: 120. 60 in each group

Withdrawals: 0

N assessed: 120 (100%), 60 in each group

Age: range 15‐56 years

Sex: 52 males/68 females

Baseline data: the duration of the lesions ranged from 3 weeks to 3 months. Lesions were situated mainly on the exposed parts of the body (face, arms and feet). 46 participants (24 in dapsone group and 22 in placebo group) had a single lesion while 74 participants had multiple lesions (maximum 13).

Interventions

Type of interventions:

  • Group 1: dapsone tablets (100 mg)

  • Group 2: placebo tablets

Duration of intervention: every 12 h for 6 weeks

Duration of follow‐up: 6 weeks

Outcomes

Primary outcome: percentage of participants 'cured' at the end of treatment

Secondary outcome: adverse effects

Tertiary outcomes: microbiological or histopathological cure of skin lesions

Time points reported: clinical response: days 15 and 45. Clinical and parasitological response: 6 weeks

Notes

Informed consent obtained: yes

Study funding sources: —

Possible conflicts of interest: —

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "... and was randomly allocated to receive either tablets of dapsone (100 mg) or placebo tablets which were identical in appearance"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Quote: "... and was randomly allocated to receive either tablets of dapsone (100 mg) or placebo tablets which were identical in appearance"

Comment: no further information provided.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "double‐blind therapeutic trial"

Comment: participants looks like blinded but no description about personnel blinding were provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "double‐blind therapeutic trial"

Comment: no further information about blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All relevant outcome data were provided

Selective reporting (reporting bias)

Low risk

Relevant outcomes were reported

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Dogra 1992

Methods

Study design: randomised controlled trial

Setting/location: India

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L tropica in the area

Inclusion criteria: participants with localised CL and only smear positive cases

Exclusion criteria: not being smear positive

N randomised: 60, 20 in each group

Withdrawals: 0

N assessed: 60 (100%), 20 in each group

Age (years): ≥ 15 years

Sex: not described

Severity of illness: 8, 11, and 12 participants had multiple lesions in the itraconazole, dapsone and placebo groups respectively.

Interventions

Type of interventions:

  • Group 1: itraconazole 4 mg/kg/d (max. 200 mg)

  • Group 2: dapsone 4 mg/kg in 2 doses/d

  • Group 3: placebo control group

Duration of intervention: 6 weeks

Duration of follow‐up: 6 weeks, but 3 months for assessment of relapses

Outcomes

Healing rates: percentage of participants 'cured' at the end of treatment. Strict clinical and parasitological criteria were followed to asses cure.

Adverse effects

Time points reported: healing rates at the end of treatment

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Low risk

Relevant outcomes were reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Dogra 1996

Methods

Study design: randomised controlled trial

Setting/location: India

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania:L major and tropica in the area

Inclusion criteria: participants with localised CL. Demonstration of parasites from skin lesions by slit smear examination

Exclusion criteria: women of child‐bearing age, children < 18 years old, participants suffering from any chronic illness, immunocompromised, prior therapy for CLs in any form, scars of healed leishmanial lesions, lesions of 4 months or more duration, participants showing abnormality in liver function tests

N randomised: 20

Withdrawals: 0

N assessed: 20 (100%)

Age: range 19‐62 years

Sex: 15 males/5 females

Baseline data: the duration of the lesions ranged from 2 weeks to 16 weeks, they were mainly seen on exposed parts of the body. 9 had a single lesion and 11 participants had a multiple lesion.

Interventions

Type of interventions:

  • Group 1: itraconazole orally (2 100 mg capsules) for 6 weeks n = 10. 4/10 participants had single lesions.

  • Group 2: placebo orally (2 capsules) for 6 weeks n = 10. 5/10 participants had single lesions.

Duration of intervention: 6 weeks

Duration of follow‐up: 3 months

Outcomes

Primary outcome: percentage of participants 'cured' 3 months after treatment

Secondary outcome: adverse effects

Tertiary outcomes: microbiological or histopathological cure of skin lesions

Time points reported: clinical response: day 15. Clinical and parasitological response: 6 weeks

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were allocated randomly to receive capsules of itraconazole 100 mg or identical placebo".

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Quote: "Patients were allocated randomly to receive capsules of itraconazole 100 mg or identical placebo".

Comment: no further information provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "double‐blind therapeutic trial"

Comment: participants looks like blinded but no description about personnel blinding was provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "double‐blind therapeutic trial"

Comment: participants looks like blinded but no description about assessment blinding was provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No missing outcome data

Selective reporting (reporting bias)

Low risk

All relevant outcomes were reported

Other bias

High risk

Sample size calculation, reporting of Leishmania spp involved and baseline comparability was not correctly reported

Ejaz 2014

Methods

Study design: randomised controlled trial

Setting/location: Pakistan at dermatology departments of Combined Military Hospital, Kharian Camtonment, Combines Military Hospital, Quetta and Combined Military Hospital, Muzaffarabad

Study period: January 2008 to December 2010 (35 months)

Sample size calculation: assuming equal‐sized groups, keeping the equivalence trial design, standardised difference of 0.05 and power at 90%, sample size was calculated as 150 participants in each arm using Altman nomogram. A total of 300 participants were needed for the study and 24 extra participants were recruited to cater for dropouts.

Participants

Type of Leishmania: not described

Inclusion criteria: men and women over 18 years of age having parasitologically proven CL requiring systemic therapy, willing for admission to hospital for the study and regular follow‐up visits, and consenting not to use any other treatment for CL during trial

Exclusion criteria: pregnant, compromised immune system (i.e. diabetics or cancer patients), diffuse cutaneous or visceral leishmaniasis, complete or incomplete treatment with antimony compounds in the last 3 months, history of hepatic, renal, or cardiovascular disease

N randomised: 324. Group 1: 151; group 2: 173

Withdrawals: not described

N assessed: 324 (100%)

Age: mean 27.9 years (SD 6.5), range 17‐48 years

Sex: all were male soldiers

Baseline data: most participants (39.2%) had a single lesion, but one participant had 15 lesions. Maximum lesions were found on exposed parts of body, arms and legs accounted for 42.3% and 47.2% of lesions respectively. Plaques were the most common morphological pattern seen (82.7%).

Mean (SD) size of lesions at baseline was 28.8 mm (16.1). Group 1: 29.7 mm (16.4); group 2: 28 mm (15.8). Mean induration at baseline was 17.5 mm (11.6). Group 1: 17.7 mm (11.5); group 2: 18 mm (12.8)

Interventions

Type of interventions:

  • Group 1: IMMA (Glucantime, Lot No. 888, Aventis Laboratories, France) 20 mg/kg/d until clinical resolution or for 28 days maximum

  • Group 2: IMMA 10 mg/kg/d + oral allopurinol (Zyloric – 300, Batch No. 2ZMAF, GlaxoSmithKline Pakistan Limited) 20 mg/kg/d

Duration of intervention: 28 days maximum

Outcomes

Successful treatment: was defined as complete re‐epithelialisation of the ulcer and disappearance of the induration, or reduction of more than 50% of the ulcer and the indurations areas in relation to the last clinical evaluation

Adverse effects: serious adverse event was defined as life‐threatening, or prolongation of existing hospitalisation, or causing persistent or significant disability. Non‐serious adverse effects, not qualifying the above criteria were clinically judged by the investigator to be definitely related, probably related, possibly related, or not related to the trial medication.

Time points reported: follow‐up phase lasted 6 months

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Simple randomisation through a random number table and allocation ratio being 1:1.

Allocation concealment (selection bias)

Unclear risk

"Randomisation sequence generation and allocation of medicine was done by lead investigators at the three research set ups"

Comment: no information on the method of allocation concealment was provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Outcome assessors were blinded from the treatment groups; however, it is not clear how this was done. No placebo was provided.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "There were 18 dropouts due to various complications, 9 belonging to each group"

Comment: ITT analyses were performed.

Selective reporting (reporting bias)

Unclear risk

Protocol not available. The trial was registered with www.anzctr.org.au and the registration number is ACTRN12607000295448. Some outcomes registered in the protocol were not reported: successful treatment; therapeutic failure; reactivation

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

El‐Sayed 2010

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: the Saudi Hospital in Sanaa, Yemen

Study period: 21 months; participants recruited from June 2006 to June 2007

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania spp

Inclusion criteria: all had the clinical signs of cutaneous leishmaniasis and a majority of their cutaneous lesions were found smear positive for mastigotes. 4 participants with a negative smear preparation were diagnosed by tissue culture using NNN (Novey, McNeal and Nicolle) medium

Exclusion criteria: skin lesions of more than 8 weeks duration, allergic to antimonial drugs, lactating or pregnant

N participants randomised (lesions): N = 30: ILSSG, n = 10 participants (12 lesions), ILSSG + IMSSG, n = 10 (15), ILSSG + oral ketoconazole: n = 10 (13)

Withdrawals: no

N assessed: 30 (100%)

Age: range 12‐50 years (mean 23.5, SD 14). Group 1: 12‐50 (24.3, SD 15); group 2: 13‐48 years (22.5, SD 14); group 3: 12‐49 years (25.1, SD 12)

Sex (male/female): group 1, 5/5; group 2, 7/3; group 3, 4/6

Baseline data:

  • Group 1: duration of disease (weeks) 2–5. Site: face 10, upper extremities: 2. Type of ulceration: nodules 5, plaques 7

  • Group 2: duration of disease (weeks) 3‐7. Site: face 14, upper extremities: 1. Type of ulceration: nodules 8, plaques 7

  • Group 3: duration of disease (weeks) 2–8. Site: face 12, upper extremities: 1. Type of ulceration: nodules 6, plaques 7

Interventions

Type of interventions:

  • Group 1: ILSSG (100 mg/mL) alone, following a treatment schedule on alternate day's injection

  • Group 2: ILSSG + IMSSG injections. A part of the dose (calculated as 20 mg/kg/d) was injected intralesionally on days 1, 3, 5 as in group 1. The remaining amount of the total dose was given intramuscularly simultaneously on the same day.

  • Group 3: ILSSG as in group 1 + oral ketoconazole (200 mg 3 times daily) for 4 weeks

Duration of intervention: in all groups, treatment was continued until clearance or for a maximum of 3 treatment cycles at 4 week intervals (12 weeks)

Follow‐up: performed monthly for 6 months after the last treatment

Outcomes

Clinical cure: the cure was indicated by complete re‐epithelialisation, the disappearance of oedema, induration and other signs of inflammation and a negative Giemsa‐stained direct smear of a scraping of the skin at the lesion site. In the 4 participants diagnosed only by a positive culture before enrolment, the cure was indicated by a negative culture at the end of the 12 weeks.

Adverse effects

Time points reported: 4, 8, 13 weeks. Adverse effects: at the end of treatment

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "A follow‐up assessment was performed by the treating clinician, the patient and by comparing the serial photographs"

Comment: open trial

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants completed the study

Selective reporting (reporting bias)

Low risk

All relevant outcomes were reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Emad 2011

Methods

Study design: randomised controlled trial

Setting/location: Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L major

Inclusion criteria: PCR‐proven L major cutaneous infection, age > 12 years, not using anti‐Leishmania therapies during the past 2 months, duration of lesions < 4 moths

Exclusion criteria: pregnancy, breastfeeding, presence of lesions on ears and/or face, number of lesions >10, history of liver or kidney disease

N randomised: 120 participants (fluconazole 200 mg: 60; fluconazole 400 mg: 60)

Withdrawals: 2 in the 400 mg group

N assessed: 118 participants

Mean (SD) age: fluconazole 200 mg, 36.45 years (15.34); fluconazole 400 mg, 35.38 years (13.81)

Sex (male/female): 65/55 (fluconazole 200 mg, 30/30; fluconazole 400 mg, 35/25)

Baseline data:

  • Mean number of lesions (SD): fluconazole 200 mg, 3.10 (2.08); fluconazole 400 mg, 3.21 (2.09)

  • Mean duration of lesions (SD): fluconazole 200 mg, 7.12 weeks (2.51); fluconazole 400 mg, 7.43 weeks (2.6)

  • Size of lesions (SD): fluconazole 200 mg, 19.84 mm (8.97); fluconazole 400 mg, 21.87 mm (8.93)

Interventions

Type of interventions:

  • Group 1: high dose fluconazole 200 mg twice daily

  • Group 2: low dose fluconazole100 mg twice daily

Duration of intervention: 6 weeks

Outcomes

Complete healing: defined as complete re‐epithelialisation of the lesions at intervals of 2, 4 and 6 weeks

Adverse effects

Time points reported: 6 weeks

Notes

Study funding sources: grant from Shiraz University of Medical Sciences, Shiraz, Iran

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The patients were then randomized into two equal groups to receive either fluconazole 100 mg or 200 mg twice daily."

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2 dropouts were reported in the fluconazole 400 mg group. ITT analysis were performed.

Selective reporting (reporting bias)

Unclear risk

Our primary outcomes (Complete healing and adverse effects) were described in Methods and reported in Results.

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Esfandiarpour 2002

Methods

Study design: randomised controlled trial

Setting/location: Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L tropica in the area

Inclusion criteria: positive direct smear or documented skin biopsy, not pregnant or nursing at the time of therapy, no serious systemic illness, no sensitivity to antimonial drugs or AL

Exclusion criteria: not having received treatment for at least 2 months before entry into the study.

N randomised: 150. 50 in each group

Withdrawals: 0

N assessed: 150 (100%). 50 in each group

Age: mean age 14 years. Group 1 (AL): 14, group 2 (IMMA): 14.5, group 3 (AL + IMMA): 14

Sex: male/female: 69/81. 23/27 in each group

Baseline data: the common sites of involvement were the face and extremities. By groups were as follows: oral AL: face: 62%; upper limbs: 34%; lower limbs: 4%, and trunk: nil. IMMA: face: 48%; upper limbs: 38%; lower limbs: 14%, and trunk: nil. AL + IMMA: face: 78%; upper limbs: 15%; lower limbs: 6%, and trunk: 2%. Most participants had plaque‐type, papular, and nodular lesions. By groups: AL: plaque: 64%, papule: 29%, and nodule: 7%. IMMA: plaque: 71%, papule: 18%, and nodule: 11%. AL + IMMA: plaque: 77%, papule: 19%, and nodule: 4%.

Mean number of lesions: group 1, 1.5; group 2, 1.8; group 3, 1.4. Most had multiple lesions (≤ 7 lesions)

Mean duration of lesions: group 1, 8 months; group 2, 7.4 months; group 3, 8 months

Interventions

Type of interventions:

  • Group 1: AL orally 15 mg/kg/d for 3 weeks

  • Group 2: IMMA 30 mg/kg/d for 2 weeks

  • Group 3: AL + IMMA simultaneously

Duration of follow‐up: one month

Outcomes

Primary outcome: percentage of participants 'cured' at the end of treatment. The response to treatment was graded as excellent (reduction in the size of the lesion by at least 80% or complete clearance), good (reduction in the size of the lesion by 50%), and poor (minimal or no change in the lesion)

Secondary outcome: adverse effects

Time points reported: end of treatment and 2 and 4 weeks later

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "the patients were randomly divided into three groups."

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Quote: "the patients were randomly divided into three groups."

Comment: no further information provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: the treatment it is not possible to blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: no reference to the outcome assessment blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: no missing outcome data

Selective reporting (reporting bias)

High risk

Comment: no reference to adverse effects

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Faghihi 2003

Methods

Study design: randomised controlled trial

Setting/location: Isfahan, Iran

Study period: 15 months

Sample size calculation: yes

Participants

Type of Leishmania: L major is endemic in Isfahan

Inclusion criteria: clinical and parasitological diagnosis of CL

Exclusion criteria: pregnant or had > 3 lesions, ulcerative lesions, lesions with cartilage or lymphatic involvement or hypersensitivity to the drug

N randomised: 96 participants, 48 in each group

Withdrawals: 0

N assessed (lesions): 96 (190). 48 (95) in each group

Age (years): age range 1‐48 years (mean age of 16 years old and a median of 14.5 years)

Sex: male/female: 40/56

Baseline data: all lesions treated were papules or early nodules with mean diameter of about 4 mm. Mean number of lesions per person in each group: 1

Interventions

Type of interventions:

  • Group 1: 15% PR sulphate and 10% urea in Eucerin ointment, applied twice daily at 1 mm thickness over the total surface of the lesion(s)

  • Group 2: ILMA injections of 1.5 g/5 mL (maximum 12), weekly. The mean amount of solution required for each lesion was 0.2 mL to 0.8 mL.

Duration of intervention: treatment was continued in both groups for 3 months or until complete recovery (return to the normal tissue texture without any atrophic changes or scar formation)

Duration of follow‐up: one year

Outcomes

Primary outcome: percentage of participants 'cured' within 2 months after treatment. Complete recovery or cure was defined as re‐epithelialisation and return to normal tissue texture in less than 2 months, with no residual scar or relapse after follow‐up of up to 1 year

Secondary outcomes: duration of remission and percentage of people with treated lesions that recur within one year; prevention of scarring

Time points reported: completely recovered (cured) (< 2 months). Healed (in 2–3 months)

Notes

Study funding sources: Isfahan University of Medical Sciences (Pharmacy College and its Research Laboratory) and the Amin Leishmaniasis Research Group in Isfahan City

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "In the period of recruitment, […] they were randomised by fixed block random allocation".

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Treatment description do not appear as blinded

Blinding of outcome assessment (detection bias)
All outcomes

High risk

In statistical methods section, quote "There was no blinding method used."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No missing outcome data

Selective reporting (reporting bias)

Low risk

Relevant outcomes were reported

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Farajzadeh 2015

Methods

Study design: randomised controlled trial

Setting/location: Afzalipour Hospital, Kerman University of Medical Sciences, Iran

Study period: 1 year (2011‐12)

Sample size calculation: a sample size of 40 participants per treatment group was planned, a probability of a type I error at alpha = 0.05 and beta = 0.1 to determine a 20% difference between topical terbinafine compared with control group

Participants

Type of Leishmania: L tropica

Inclusion criteria: participants between 2‐60 years and without serious medical illness were included, not have received any other leishmanicidal treatment during last 3 months, duration of their lesions should be less than 6 months

Exclusion criteria: cardiac, hepatic, renal and other systemic diseases; pregnant and nursing women; hypersensitivity to trial drugs

N randomised: 80, 40 in each group

Withdrawals: 0

N assessed: 80, 40 in each group

Mean (SD) age: terbinafine: 16.3 years (14.8), IMMA: 20.74 years (19.4)

Sex (n, %): terbinafine: male 18 (45); female 18 (45); the sex of 4 (10) participants was unknown. IMMA: male: 16 (40); female: 18 (45); the sex of 6 (15) participants was unknown

Baseline data:

  • Mean duration of lesions (SD): terbinafine: 3.97 months (0.3). IMMA: 3.81 months (0.3)

  • Mean number of lesions (SD): terbinafine: 1.85 (1.3). IMMA: 1.93 (1.16)

  • Mean size of lesions (SD): terbinafine: 4.15 mm² (5.97). IMMA: 5.21 mm² (10.65)

  • The most frequent location of the lesions was on participants' face (terbinafine: n = 99, 45.8% vs IMMA: n = 93, 50%)

Interventions

Type of interventions:

  • Group 1: oral terbinafine, 125 mg/d (for less than 20 kg body weight), 250 mg/d (20‐40 kg body weight), 500 mg/d (for more than 40 kg body weight)

  • Group 2: 15 mg/kg/d IMMA (Glucantime; Haupt Pharma in France)

Co‐interventions: Both groups received cryotherapy every 2 weeks for 4 weeks

Duration of intervention: group 1: 4 weeks, group 2: 3 weeks

Duration of follow up: patients were followed monthly for 3 months after the treatment.

Outcomes

Clinical cure: clinical response was determined based on the following criteria:

  • Complete improvement (decrease in induration size > 75%)

  • Partial improvement (decrease in induration size between 25% and 75%)

  • No improvement (decrease in induration size < 25%)

Time to clinical cure: development of oartial and complete response to treatment in IMMA and terbinafine groups.

Time points reported: the improving rate determined by measuring indurations at baseline, in the middle (day 10 for IMMA group and day14th for terbinafine group), and at the end of the study (day 21 for IMMA group and day 28 for terbinafine group)

Notes

Study funding sources: Kerman University of Medical Sciences accepted the financial support of this study

Possible conflicts of interest: the authors declare that there is no conflict of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The randomisation sequence was obtained by the use of a randomisation table.

Allocation concealment (selection bias)

Low risk

A simple block randomisation list with a block size of 4 was accumulated by a team member who was not involved in the enlistment and follow‐up of the participants. The randomisation allocation concealment was carried out by sending the randomisation numbers in envelopes to a dermatologist who was responsible for giving the assigned treatment after each participant was en‐rolled.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Different administration of treatments: oral terbinafine and IMMA.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

This study was an assessor blind trial in which the outcome assessor was unaware of the drugs used by the participants.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

High risk

The study published does not report 2 primary outcomes: adverse effects and recurrence

Other bias

Low risk

Other items assessed correctly reported

Fekri 2015

Methods

Study design: randomised controlled trial

Setting/location: Afzalipour hospital affiliated to Kerman University of Medical Sciences and Dadbin Health Center, Kerman/Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: —

Inclusion criteria: age over 7 years, positive smear or biopsy, providing informed consent

Exclusion criteria: pregnant or lactating, lesions on face, more than 5 lesions, duration longer than one year, size larger than 3 cm, any treatment in past month, history of allergy to MA or dapsone, systemic diseases, taking immunosuppressive drugs in past 6 months, lupoid or sporotrichoid forms

N randomised: 73

Withdrawals: 5

N assessed (lesions): 68 (73). Group 1: 33 (35); group 2: 35 (38)

Mean (SD) age: 29.6 years (15.4) in group 1, 31.6 years (20.4) in group 2

Sex (male/female): intervention group: 17/16, group 2: 14/17

Baseline data:

  • Number of lesions: 38 lesions in group 1 and 35 in group 2

  • Mean±SD of duration of lesions (months): 4.6 ± 9.3 in group 1 and 3.7 ± 1.9 in group 2

  • Mean±IQR of the initial lesion size (mm2): 2 (1.4‐2.9) in group 1 and 1.5 (1.1‐3) in group 2

Interventions

Type of interventions:

  • Group 1: weekly ILMA + niosomal dapsone gel twice a day

  • Group 2: weekly ILMA + cryotherapy every 2 weeks

Duration of intervention: until complete healing or max 16 weeks

Outcomes

Healing response: complete healing (100% epithelialisation and loss of induration), moderate healing (50‐99% epithelialisation and loss of induration), no response (less than 50% epithelialisation and loss of induration)

Time points reported: 16 weeks after beginning intervention, 1 year later for recurrence

Notes

Study funding sources: Kerman University of Medical Sciences

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number table

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open study

Incomplete outcome data (attrition bias)
All outcomes

High risk

No reasons for missing outcome data

Selective reporting (reporting bias)

High risk

One outcome of interest in the review is not reported: adverse effects, so that it cannot be entered in a meta‐analysis

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Firooz 2005

Methods

Study design: randomised controlled trial

Setting/location: Isfahan, Iran

Study period: 6 months

Sample size calculation: not described

Participants

Type of Leishmania: L major endemic in the area

Inclusion criteria: the presence of parasitologically confirmed lesion(s) of CL, aged 12‐60 years, and otherwise healthy on the basis of medical history and physical examination

Exclusion criteria: women of childbearing age without adequate effective contraception; pregnant or breastfeeding; duration of lesions > 8 weeks; the presence of lesions on face, joints or near the mucous membranes; the presence of > 5 lesions or any lesion with a diameter > 5 cm; history of any antileishmanial therapy in the past 4 weeks

N participants randomised (lesions): 72 (106). 36 (53) treated with zinc sulphate (ZS) and 36 (53) treated with ILMA

Withdrawals participants (lesions): 37 (56). ZS group 23 (34): unresponsive: 12, lost to follow‐up: 5, non‐medical events: 2, protocol deviation: 2, complications: 1, patient request: 1. ILMA 14 (22): unresponsive: 2, lost to follow‐up: 10, non‐medical events: 1, patient request: 1

N assessed (lesions): 35 (50). 13 (19) treated with ZS and 22 (31) treated with ILMA

Mean (SD) age: 20.2. years (9.6). ZS group: 18.1 years (6.1); ILMA: 22.3 years (11.2)

Sex (male/female): 33/39. ZS group: 19/17; ILMA group: 14/22.

Baseline data:

  • Location of the lesions (n (%)): head and neck: ZS group: 7 (13.2); ILMA group: 1 (1.9); trunk: ZS group: 3 (5.7); ILMA group: 1 (1.9); upper extremity: ZS group: 28 (52.8); ILMA group: 32 (60.4); lower extremity: ZS group: 15 (28.3); ILMA group: 19 (35.8)

  • ZS group: MNL (SD): 1.4 (0.8). MSL (SD): 7.6 mm (5.5). MDLBT: 5.8 weeks (2.0)

  • ILMA group: MNL (SD): 1.5 (0.8). MSL (SD): 7.9 mm (7.5). MDLBT: 5.5 weeks (2.3)

Interventions

Type of interventions:

  • Group 1: IL 2% zinc sulphate

  • Group 2: ILMA

Duration of intervention: up to 6 weeks

Duration of follow‐up: 5 weeks

Outcomes

Healing rates: percentage of lesions 'cured' 5 weeks after treatment. Complete re‐epithelialisation of each ulcer with marked reduction in induration with or without scarring was considered as the main efficacy parameter.

Amount of injection into each lesion

Adverse effects: the pain experienced by the participant (using a verbal analogue scale on a 0‐10 scale) was recorded

Time points reported: 6 weeks

Notes

Study funding sources: this study was supported by a grant from Chancellery of Research, Tehran University of Medical Sciences.

Possible conflicts of interest: none declared

The plot and the figures were not matched accordingly

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Table of random numbers

Allocation concealment (selection bias)

Low risk

The randomisation sequence was concealed from the investigators until the data entry was completed and the data bank was locked.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Double‐blind"

Comment: the preparation and coding of drugs were done by a pharmacist outside of the research team and investigators were blinded to them

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

High rate of dropouts (37/72, 54%). Missing outcome data imbalanced in numbers and reasons across intervention groups. ZS group: 23. ILMA group: 14. 'As‐treated' analysis done with substantial departure of the intervention received from that assigned at randomisation

Selective reporting (reporting bias)

Low risk

All relevant outcomes reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Firooz 2006

Methods

Study design: randomised controlled trial

Setting/location: Mashad, Iran

Study period: 18 months

Sample size calculation: 45 participants per treatment group were needed to have 80% power to detect a significant difference in the expected cure rate of 50% in the vehicle group and the desired cure rate of 80% in the imiquimod‐treated group at week 8 with a type Ierror level of 0.05. To compensate for 20% estimated dropout, 54 participants would be required in each group.

Participants

Type of Leishmania: L tropica endemic in the area

Inclusion criteria: parasitologically proven cases of CL based on positive smear or culture, otherwise healthy participants

Exclusion criteria: pregnant or lactating women, duration of the lesions > 6 months, number of lesions > 5, any lesions > 5 cm, history of any standard course of treatment with antimonials, history of allergy to antimonials, serious systemic illnesses, participation in any drug trials in the last 60 days

N participants randomised (lesions): 119 (252). 59 (128) participants in the imiquimod group and 60 (124) in the vehicle group

Withdrawals: 30 participants. Imiquimod group (n = 17): 9 lost to follow‐up, 4 inadequate efficacy, 3 protocol deviation, 1 consent withdrawal. Vehicle group (n = 13): 9 lost to follow‐up, 2 inadequate efficacy, 2 protocol deviation

N assessed: 89. 42 participants in the imiquimod group and 47 in the vehicle group

Mean (SD) age: 27.0 years (SD 1, range 12‐60). Imiquimod group: 7.4 years (13); vehicle group: 6.5 years (12)

Sex: approximately 50%‐55% of the participants were female (35/59 in the imiquimod group, and 31/60 in the vehicle group)

Baseline data:

  • Lesions were mainly located on the upper extremities (66.3%) and around 15% to 17% on the face and lower extremities

  • Location of lesions (n (%)): face: imiquimod group: 20 (15.6), vehicle group: 19 (15.3); trunk: imiquimod group: 1 (0.8); vehicle group: 1 (0.8); upper extremity: imiquimod group: 85 (66.4); vehicle group: 82 (66.1); lower extremity: imiquimod group: 22 (17.2); vehicle group: 22 (17.7)

  • Imiquimod group: MNL: 2.2. MDLBT: 13 weeks. MSL: 174 mm²

  • Vehicle group: MNL: 2.0. MDLBT: 13.2 weeks. MSL: 237 mm²

Interventions

Type of interventions:

  • Group 1: imiquimod cream 5% + IMMA

  • Group 2: vehicle + IMMA

Duration of intervention: imiquimod and vehicle 3 times per week for 28 days, IMMA 20 mg/kg/d for 14 days

Duration of follow‐up: 20 weeks after initiation of treatment

Outcomes

Primary outcome: percentage of participants 'cured' 3.5 months after treatment. Clinical cure of the participants, defined as more than 75% reduction in the size of lesions compared with baseline

Secondary outcomes: the relapse rate (defined as a reappearance of lesions at the site or periphery of previously healed lesions or an increase in the size of lesions after initial improvement) was assessed 16 weeks after the end of treatment

Adverse effects

Time points reported: at the end of the treatment period (week 4) and 4 weeks later

Intention‐to‐treat analysis of rates of complete re‐epithelialisation, clinical cure, and clinical improvement at weeks 4, 8,and 20 after initiation of treatment

Notes

Study funding sources: this study was supported by the Small Grants Scheme for Operational Research in Tropical and Other Communicable Diseases from the Joint World Health Organization Eastern Mediterranean Region Division of Communicable Diseases and the Special Program for Research and Training in Tropical Diseases.

Possible conflicts of interest: the funding source was involved in the study design, in the writing of the manuscript, and in the decision to submit the manuscript for publication, but not in the collection, analysis, or interpretation of data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "randomly divided into 2 groups according to a list made by a simple randomisation block design"

Allocation concealment (selection bias)

Low risk

Quote: "The randomisation allocation concealment was performed by sending the randomisation numbers in envelopes to a pharmacist who was responsible for giving the assigned treatment after each eligible patient was enrolled."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Treatment unlikely to be blinded. This study was a single‐blind trial.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "To keep the trial blinded, the physicians who were responsible for evaluation of patients were uninvolved in the process of allocation and drug dispensing and were unaware of the drug used by the patients."

Incomplete outcome data (attrition bias)
All outcomes

High risk

Withdrawals: 30/119 (25.2%); no ITT analyses performed

Selective reporting (reporting bias)

Low risk

All relevant outcomes reported

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Gholami 2000

Methods

Study design: randomised controlled trial

Setting/location: Iran

Study period: 24 months

Sample size calculation: yes

Participants

Type of Leishmania:L major

Inclusion criteria: CL confirmed with direct smear, age 5‐50 years, maximum number of lesions 3, duration of disease < 100 days

Exclusion criteria: previous treatment for leishmaniasis, use of immunosuppressives, history of chronic systemic disease, lesions on face, pregnancy or lactating, age < 5 years, duration of disease > 100 days

N randomised: 197

Withdrawals: 26

N assessed: 171. 96 were treated with garlic 5% cream and 75 with vehicle

Mean age: garlic, 18.5 years; vehicle, 23.7 years

Sex (male/female): garlic, 51/45; placebo, 38/37

Baseline data: not reported

Interventions

Type of interventions:

  • Group 1: garlic cream 5%

  • Group 2: vehicle

Duration of intervention: both applied twice daily under occlusion with sterile gauze for 3 hours, for 20 days

Duration of follow‐up: 60 days (after the 3 week treatment, participants were followed for another period of 40 days)

Outcomes

Healing rates: percentage of participants 'cured' one month (40 days) after treatment

Time points reported: yes

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

We only have the abstract. The author (A Khamesipour) was contacted and kindly agreed to extract the data from the original paper written in Persian.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

List generated by a computer

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Low rates of dropouts (26/197; < 25%)

Selective reporting (reporting bias)

Unclear risk

All relevant outcomes reported

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Harms 1991

Methods

Study design: randomised controlled trial

Setting/location: Aleppo, Syria

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L tropica

Inclusion criteria: up to 3 lesions diagnosed clinically and parasitologically as CL; absence of chronic or intercurrent systemic disease

Exclusion criteria: pregnancy and no prior antimonial medication

N randomised and withdrawals: not reported

N participants assessed (lesions): 40 (75) ILMA: 20 (38); IFN‐γ: 20 (37)

Mean age (range): ILMA: 21 years (6‐60); IFN‐γ: 27 years (12‐57)

Sex (male/female): 17/23. ILMA: 9/11; IFN‐γ: 2/12

Baseline data:

  • Type of lesions (%). ILMA group: papular: 13; papular‐nodular: 21; nodular‐ulcerative: 61; plaque‐form: 5. IFN‐γ group: papular: 19; papular‐nodular: 19; nodular‐ulcerative: 62; plaque‐form: none

  • Lesion site (%). ILMA group: upper extremity: 63; lower extremity: 18; face: 18; trunk: none. IFN‐γ group: upper extremity: 59; lower extremity: 30; face: 8; trunk: 3

  • Median size (range) (mm) : ILMA group:15 x 14(4 x 4‐ 49 x 42);IFN‐γ group: 13 x 11(7 x 7‐ 45 x 40)

  • Mean duration of lesions before treatment (range) (months): ILMA group: 2.5(1‐6); IFN‐γ group:2.5(2‐8)

Interventions

Type of interventions:

  • Group 1: ILMA (1‐3 mL)

  • Group 2: IL Lyophilised recombinant IFN‐γ (25 mg)

Duration of intervention: once weekly for 5 weeks

Duration of follow‐up: 10 weeks

Outcomes

Primary outcome: percentage of lesions 'cured' one month after treatment

Secondary outcome: adverse effects

Tertiary outcomes: microbiological or histopathological cure of skin lesions

Time points reported: weeks 3, 6, 10

Notes

Study funding sources: none reported

Possible conflicts of interest: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random numbers table method used

Allocation concealment (selection bias)

Unclear risk

Comment: no information about allocation concealment

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Comment: no information about blinding

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: no information about blinding

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No losses to follow‐up were reported.

Selective reporting (reporting bias)

Low risk

Comment: all relevant outcomes provided

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Iraji 2004

Methods

Study design: randomised controlled trial

Setting/location: Department of Dermatology, Isfahan Medical University, Iran

Study period: 10 months

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania spp: due to a previous study in the area, it was likely that participants were infected with L major

Inclusion criteria: proven leishmaniasis based on typical lesions of ACL and a positive direct smear. Number of lesions < 4 and duration of lesion < 12 weeks

Exclusion criteria: cases of reinfection, pregnant or nursing women, those who had lesion on the face or joints and participants with sporotrichoid or erysipeloid lesions

N randomised: 104. ILMA: 55, zinc sulphate (ZS): 49

Withdrawals: 38. ILMA: 20, ZS: 18. 13 due to occurrence of new lesions: 7 from ILMA group and 6 from ZS group; 6 participants with sporotrichoid spread: 4 from the ILMA group and 2 from the ZS group; 19 participants lost to follow‐up: 9 from the ILMA group and 10 from the ZS group

N assessed: 66. ILMA: 35, ZS: 31

Mean age (range): ILMA: 11 years (2‐67); ZS: 12 years (3‐64)

Sex (male/female): 50/54. ILMA: 14/21; ZS: 17/14

Baseline imbalances: sex distribution in each group

Severity of illness: Mean duration of lesions (SD) (weeks): ILMA: 6.73 ±0.53 ;ZS: 7.64±0.12 .

Interventions

Type of interventions:

  • Group 1: ILMA injection of 50 mL

  • Group 2: IL ZS injection of 50 mL

In cases where there was a slight to mild improvement, another injection was given after 2 weeks

Duration of intervention: 2‐6 weeks

Duration of follow‐up: 6 weeks

Outcomes

Healing rates: percentage of participants 'cured' at the end of treatment. The results of treatment were graded according to the system of Sharquie 1997 slight = 1, mild = 2, moderate = 3, marked = 4, 5 = total clearance of the lesion and parasite not detected in the affected area by smear

Adverse effects

Time points reported: 2, 4, 6 weeks

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants were treated randomly with intralesional injection of any of 2 similar 50‐mL vials marked A (MA) or B (ZS) by an independent physician. ZS solution was prepared by dissolving 2 g ZS (ZNSO4W 7H2O) per 100 mL of bidistilled deionised water.

The blinding was unlikely to have been broken.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing outcome data balanced in numbers across intervention groups, with similar reasons for missing data across groups

Selective reporting (reporting bias)

Low risk

The study's pre‐specified (primary and secondary) outcomes that are of interest in the review have been reported in the pre‐specified way

Other bias

High risk

Sample size calculation, reporting of Leishmania spp involved and baseline comparability was not correctly reported

Iraji 2005

Methods

Study design: randomised controlled trial

Setting/location: Iran

Study period: not described

Sample size calculation: the required sample size, 40 in each treatment group, was estimated, assuming one in every 4 cases would not provide full data, using the formula n = [Z1 − (a/2) + Z1 − b] 2 × [P1(1 − P1) + P2(1 − P2)]/(P1 − P2)2, and setting P1 (the paromomycin efficacy) to 0.7, P2 (the vehicle efficacy) to 0.3, a to 0.05, b to 0.1, Z1 − (a/2) to 1.96, and Z(1 − b) to 1.64

Participants

Type of Leishmania: L tropica and L major are responsible for hyperendemic CL in rural areas and endemic CL in parts of many cities in Iran

Inclusion criteria: had the clinical signs of CL and all of their cutaneous lesions were found smear‐positive for amastigotes

Exclusion criteria: cases who had first noticed a skin lesion > 3 months previously, had lesions on their face, had lesions with diameter of > 3 cm, had received previous treatment, or who were lactating or pregnant

N randomised: 80, 40 in each group

Withdrawals: 15. Intervention group: 10; control group: 5

N assessed: 65. Intervention group: 30; control group: 35

Age: range 8‐55 years. Mean intervention group: 21.4 years; control group: 21.5 years

Sex (male/female): 33/32. Intervention group: 19/11; control group: 14/21

Baseline data: mean duration of lesions (years): intervention group: 1.65; control group: 1.75

Interventions

Type of interventions:

  • Group 1: PR sulphate 15% + 10% urea applied to a 1 mm‐thick layer twice daily

  • Group 2: vehicle control group (Eucerin containing 10% urea) applied topically twice daily

Duration of intervention: 30 days

Duration of follow‐up: for clinical and parasitological follow‐up on day 30 and for only parasitological follow‐up on day 60

Outcomes

Healing rates: percentage of participants 'cured' one month after treatment, clinical and parasitological cure

  • Complete healing was defined as a reduction in the size and induration of the lesion(s) by at least 75% and lesion smears that appeared amastigote‐free

  • Partial cure was defined as a reduction in the size and induration of the lesion(s) by > 25% but < 75%, and lesion smears that appeared amastigote‐free.

  • Failure was defined as a reduction in the size and induration of the lesion(s) by < 25% and/or an amastigote‐positive smear

Adverse effects

Time points reported: days 30 (clinical and parasitological follow‐up) and 60 (parasitological follow‐up)

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The cases enrolled were assigned to two treatment groups, placebo or paromomycin, using computer‐based randomisation."

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Study was described as "double blind" but no description about allocation method was given.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No blinding of outcome assessment described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No data about dropouts were given

Selective reporting (reporting bias)

Low risk

Relevant outcomes were reported

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Jaffar 2006

Methods

Study design: randomised controlled trial

Setting/location: Saudi Arabia (Bahrain)

Study period: 12 months

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: proved positive for Leishmania parasites (amastigotes) on microscopic examination or biopsy

Exclusion criteria: not described

N randomised: 62. Rifampicin: 46; placebo: 16.

Rifampicin: 2 groups were analysed; group 1a (children aged 3‐11 years ‐ mean age, 7.5 years), and group 1b (adults aged 12‐65 years ‐ mean age, 33 years). 32 participants enrolled in group 1a and 30 in group 1b. Out of these, 8 participants in each group (16 in total) served as control to receive the placebo.

Withdrawals: 21 participants lost to follow‐up. Rifampicin: 12; placebo: 9

N assessed: 41. Rifampicin: 34; placebo: 7. Number participants imbalanced between groups

Age: range 3‐65 years; mean 20 years

Sex (male/female): 43/19

Baseline data: the duration of the lesions varied between 1 and 12 months (mean 2.6 months). The lesions of CL were single or multiple and were mostly over the extremities (upper limbs 51% and lower limbs 38%) and to a lesser extent on the face 30%. Most of the lesions were active being nodular, nodule‐ulcerative, or ulcerative. 2 of the participants were members of the same family. However, the rest of the participants had a negative family history.

Interventions

Type of interventions:

  • Group 1: rifampicin orally in a dose of 10 mg/kg/d

  • Group 2: placebo

Duration of intervention: 2 equally divided doses during meals for 4‐6 weeks

Duration of follow‐up: 3 months

Outcomes

Primary outcome: percentage of participants 'cured' 3 months after treatment

Secondary outcomes: duration of remission and percentage of people with treated lesions that recur up to 3 months of follow‐up

Adverse effects

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Randomized"

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Comment: "Randomized" but no further information was provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "Each subject in the control group was given placebo, which was supplied, in capsules/suspension identical in shape and colour to that given in the rifampicin group."

Participants look blinded but no information about personnel were provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: "Double‐blinded" but no further information

Incomplete outcome data (attrition bias)
All outcomes

High risk

There is a 38.87% of follow‐up, the percentage is very different between the groups, and it isn't explained.

Selective reporting (reporting bias)

High risk

No adverse effects data available

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Jaffary 2010

Methods

Study design: randomised controlled trial

Setting/location: Skin Disease and Leishmaniasis Research Center of Isfahan (SDLRC), Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: not described

Exclusion criteria: not described

N randomised: 140 participants

Withdrawals: 64

N assessed: 76 participants (47 with ILMA and topical Cassia fistula fruit gel and 29 participants in vehicle group)

Age: not described

Sex: not described

Baseline data: not described

Interventions

Type of interventions:

  • Group 1: Cassia fistula fruit gel + ILMA

  • Group 2: vehicle gel + ILMA

Duration of intervention: not described

Outcomes

Cure: complete cure, partial cure and treatment failure

Adverse effects (itching and erythema)

Time points reported: 12 weeks

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

We only have the abstract

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "140 patients with cutaneous leishmaniasis referring to Skin Diseases and Leishmaniasis Research Center of Isfahan (SDLRC) were randomly allocated in two groups"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Comment: the vehicle came exactly in the same colour and shape as the drug and the application procedure was also the same. We think the outcome is unlikely to be influenced by lack of blinding of personnel.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There is no information about withdrawals.

Selective reporting (reporting bias)

Low risk

Our primary outcomes (cured and adverse effects) were described in the Methods and reported in the text.

Other bias

High risk

Sample size calculation, inclusion criteria, reporting of Leishmania spp involved and baseline comparability was not correctly reported

Jaffary 2014a

Methods

Study design: double‐blind, randomised controlled study

Setting/location: Isfahan, Iran

Study period: January 2009 to February 2010 (13 months)

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: met the histological criteria for presence of parasite, age > 5 years, acute leishmanial lesions smaller than 5 cm², fewer than 5 lesions, more than 3 months of disease duration

Exclusion criteria: pregnancy, women of childbearing age, history of administration of immune suppressive drugs in the last 6 months or anti‐leishmanial drugs in the last month

N randomised: 60 participants, 30 in each group

Withdrawals: 0

N assessed: 60, 30 in each group

Mean (SD) age: 25.29 years (4.01). Achillea millefolium 5%: 25.8 years (3.7); vehicle: 24.6 years (4.2).

Sex: 15 (25%) female; 45 (75%) male. A millefolium: 7 (23.3%) female, 23 (76.6%) male; vehicle: 8 (26.6%) female, 22 (73.3%) male)

Baseline data:

  • Mean number of lesions (SD): A millefolium: 1.56 (0.8); vehicle: 1.52 (0)

  • Type of lesions (%): A millefolium: papule 8/44 (18.1), ulcer 16/44 (36.3), plaque 10/44 (22.7), nodule 10/44 (22.7). vehicle: papule: 9/38 (23.6), ulcer: 16/38 (42.1), plaque: 11/38 (28.9), nodule: 12/38 (31.5)

  • Mean duration of lesions (SD): A millefolium: 3 months (0.5); vehicle: 2.7 months (0.7)

Interventions

Type of interventions:

  • Group 1: topical gel of 5% A millefolium (yarrow (containing 5% polyphenol)

  • Group 2: vehicle gel. The vehicle gel was prepared using the same material except for the plant extract and chlorophyll was used as colorings agent. Both gels were identical in terms of the colour and consistency

Duration of intervention: 4 weeks

Co‐interventions: all participants received 4 weekly an injections of MA (Glucantime, Paris, France) at a dose of 20 mg/kg

Outcomes

Definition: complete or partial cure of lesions

Time points reported: week 12 (visits months ‐ 3)

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "During this double‐blind randomised study, 60 patients were randomised into two treatment groups by using random allocation computer software."

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "During this double‐blind randomised study ..."

Quote: "Both gels were identical in terms of the colour and consistency."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No losses to follow‐up were reported, therefore ITT analyses performed

Selective reporting (reporting bias)

Unclear risk

Insufficient information to judge

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Jaffary 2014b

Methods

Study design: randomised parallel clinical trial

Setting/location: Skin Diseases and Leishmaniasis Research Center (SDLRC), Isfahan, Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: 6‐60 years old, with positive results of leishmaniasis smear test

Exclusion criteria: size of lesion > 3 cm, > 5 lesions, lesion duration > 12 weeks, lesion located on the eyelid or < 2 cm away from the eye, pregnant or lactating

N randomised: 165, 55 in each of 3 treatment groups

Withdrawals: 7; 3 (5.5%), participants were in the concentrated boiled extract group and 2 (3.6%) participants in the other groups withdrew from the study due to the allergic reaction to the medications

N assessed: 158. Concentrated boiled extract of Cassia fistula: 52; hydroalcoholic extract of C fistula: 53; ILMA: 53

Mean (SD) age: boiled extract of C fistula: 20.6 years (12.4); hydroalcoholic extract of C fistula: 19.8 years (11.5); ILMA: 22.9 years (13.6)

Sex (n,(%)): boiled extract of C fistula: male: 35 (63.6), female: 20 (36.4); hydroalcoholic extract of C fistula: male: 29 (52.7), female: 26 (47.3); ILMA: male: 25 (45.5), female: 30 (54.5)

Baseline data:

  • Location of lesions (n(%)):

    • Concentrated boiled extract of C fistula: foot 9 (16.4), hand 25 (45.5), trunk 5 (9.1), head and neck 8 (14.5), hand and foot 8 (14.5).

    • Hydroalcoholic extract of C fistula: foot 11 (20), hand 23 (41.8), trunk 6 (11), head and neck 10 (18.2), hand and foot 5 (9.1).

    • ILMA: foot 8 (14.5), hand 24 (43.6), trunk 5 (9.1), head and neck 11 (20), hand and foot 7 (12.7)

  • Mean number of lesions (SD):

    • Concentrated boiled extract of C fistula: 1.8 (1.13)

    • Hydroalcoholic extract of Cassia fistula: 1.72 (0.98)

    • ILMA: 2.03 (1.07)

  • Kind of lesions (n(%)):

    • Concentrated boiled extract of C fistula: nodule 23 (41.8), papule 17 (30.9), papule and nodule 15 (27.3)

    • Hydroalcoholic extract of C fistula: nodule 30 (54.5), papule 11 (20), papule and nodule 14 (25.5)

    • ILMA: nodule 39 (70.9), papule 5 (9.1), papule and nodule 11 (20)

Interventions

Type of interventions:

  • Group 1: concentrated boiled extract of C fistula. 500 g of C fistula was powdered and then mixed with water with ratio of 1:3, then boiled for 30 minutes and concentrated by distillation in vacuum condition.

  • Group 2: hydroalcoholic extract of Cassia fistula on the lesions. 500 g of C fistula was powdered and mixed with 70% ethanol with ratio of 1:3, then placed in a percolator. After 48 hours, extraction was performed. The extract was then concentrated using distillation in vacuum condition.

  • Group 3: ILMA. 0.5‐2 mL twice a week

Duration of intervention: until complete resolution of the lesion (complete epithelialisation) or for a maximum duration of 4 weeks.

Follow‐up: 3 months after completing the project, they were examined every month to assess the recurrence of the lesions

Outcomes

Clinical and parasitological cure: lesions were considered completely cured (improvement), if the participants achieved both clinical and parasitological resolutions (negative results of direct smear). Participants were considered resistant to the treatment, if no clinical changes were observed in the lesion or it was worsened.

Time to healing: comparison of complete cure time of the cutaneous Leishmaniasis lesions between 3 groups

Adverse effects: allergic reaction to the medications

Time points reported: week 1, 2, 3, 4, 16, complete cure

Notes

Study funding sources: the study was self‐funded

Possible conflicts of interest: there was no conflict of interest

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The participants were randomly allocated to 3 groups using table of random numbers.

Allocation concealment (selection bias)

Unclear risk

No information provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Different administration of treatments: 2 groups: apply the extract‐soaked gauze; third group: ILMA

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

The numbers and reasons (allergic reaction to the medications) of withdrawals were similar and small in 2 of the groups 3.5% (3/28) in concentrated boiled group vs 3.6% (2/28) in the other groups.

Selective reporting (reporting bias)

Low risk

The study published reports our primary outcomes: cured and adverse effects

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Jebran 2014

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Leishmania Clinic of the German Medical Service (GMS), Darwaze‐e‐Lahory, 1st district, Kabul, Afghanistan

Study period: 36 months (2004‐7)

Sample size calculation: sample size was calculated based on the primary outcome (i.e. complete wound healing). A number of 40 participants each was calculated to be necessary to obtain a power of 80% (significance level 5%). A presumed dropout rate of 20% led to a minimum total enrolment of 100 participants

Participants

Type of Leishmania: L tropica

Inclusion criteria: male or female participants of the GMS Clinic; 5 years of age or older; with non‐ulcerated (papule/nodule) or ulcerated lesions (ulcer with or without crust); presence of at least one parasitologically confirmed CL lesion and no prior history of leishmaniasis and/or anti‐parasitic treatment

Exclusion criteria: participants below age 5; with the presence of CL lesion(s) on or immediately adjacent to the nose, lips or eyes; pregnancy, or serious co morbidities

N randomised: 135. Group 1: 73; group 2: 62

Withdrawals: due to protocol breach: group 1, n = 14; group 2, n = 8. Lost to follow‐up: group 1, n = 21; group 2, n = 22

N assessed: 113 (83.7%): group 1: 59 (80.82%); group 2: 54 (87.09%).

Mean age (range): group 1: 18.62 years (5‐66); group 2: 18.16 years (6‐63)

Sex (male/female): 93/42; group 1: n = 51/22; group 2: n = 42/20.

Baseline data:

  • Lesion type: group 1: papule or nodule: 24, ulcer: 49; group 2: papule or nodule: 17, ulcer: 45

  • Location of the target lesion: group 1: 22 face, 2 shoulder/neck, 42 upper extremity, 7 lower extremity; group 2: 14 face, 0 shoulder/neck, 44 upper extremity, 4 lower extremity

  • MLDBT (range): group 1: 2.92 months (1‐12); group 2: 2.61 months (1‐9)

  • Giemsa smear: group 1: G0 (no amastigotes (AM)): 2, G 1 (1–10 AM): 49, G2 (11–100 AM): 7, G3 (> 100 AM): 15; group 2: G0 (no AM): 0, G 1 (1–10 AM): 40, G2 (11–100 AM): 11, G3 (> 100 AM): 11

  • Parasite load in the 1st biopsy (Leishmania per gram tissue): mean (SEM): group 2: 9.14 (2.85) × 107; group 2: 1.54 (0.83) × 108.

  • Wound surface after EC treatment: mean (SEM): group I: 347.9 mm³ (23.39); group 2: 391.9 mm³ (27.80)

Interventions

Type of interventions:

  • Group 1: superficial coagulation and removal of lesion tissue was performed using bipolar high‐frequency electrocauterisation (EC; also termed high‐frequency electrocoagulation) with the help of the electrosurgical Mini Cutter HMC 80 HF. After EC, a polyacrylate hydrogel with freshly prepared 0.045% (w/w) pharmaceutical sodium chlorite (sodium chlorosum, DAC N‐055) gel was applied to the lesions, daily

  • Group 2: superficial coagulation and removal of lesion tissue was performed using bipolar high‐frequency electrocauterisation (EC; also termed high‐frequency electrocoagulation) with the help of the electrosurgical Mini Cutter HMC 80 HF. After EC, a polyacrylate hydrogel without DAC N‐055

Duration of intervention: time to wound closure

Co‐interventions: systemic antibiotics were only given if bacteria were detected and the wound infection was severe. Neither local nor systemic antifungal therapy was allowed to exclude anti‐protozoal effects on the Leishmania infection

Outcomes

Primary outcome ‐ time to wound closure (days): the time needed for complete epithelialisation of the lesion wound was defined and photodocumented.

Secondary outcome ‐ microbiological cure: the parasite load (Leishmania/g) of the lesions was taken prior to electrocauterisation (1st time), after wound closure (2nd) and after 6 months (3rd)

Adverse effects: such as bacterial or fungal superinfections of the wounds, the formation or scars and the rate of re‐ulcerations were monitored during the treatment and follow‐up period

Notes

Possible conflicts of interest: not described

Study funding sources: this study was supported by the German‐Academic Exchange Service (travel grants to KWS, AFJ and FM), the Senior Expert Service (travel grants to KWS) and the Interdisciplinary Center of Clinical Research at the Universitätsklinikum Erlangen (IZKF, project A49 to US and CB). The founders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "subjects were randomly assigned with the help of a GMS computer at a ratio of 1:1"

Allocation concealment (selection bias)

Unclear risk

Quote: "RS and KWS were responsible for the enrolment of the patients and their assignment to the treatment groups."

Comment: not further information was provided regarding the method for allocation concealment.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "The participants, the medical personnel treating and evaluating the patients, the investigators analysing the clinical course of treated lesions, as well as the lab members performing the parasitological analyses were blinded. The preparation of the two different gels (marked as jelly A and B), which could not be visually distinguished, was performed by a technician who was not involved in the treatment of patients".

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The participants, the medical personnel treating and evaluating the patients, the investigators analysing the clinical course of treated lesions, as well as the lab members performing the parasitological analyses were blinded. The preparation of the two different gels (marked as jelly A and B), which could not be visually distinguished, was performed by a technician who was not involved in the treatment of patients".

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

14 participants of group 1 and 8 participants of group 2 were retrospectively excluded due to deviations from the study protocol.

For the follow‐up analysis, 6 months after wound healing, only 70 participants could be included, as 21 participants of group I and 22 participants of group II did not observe their appointments.

Selective reporting (reporting bias)

Low risk

Main outcomes were included. Trial was registered at clinicaltrial.gov: NCT00947362

Other bias

Low risk

Other items assessed correctly reported

Jowkar 2012

Methods

Study design: double‐blind, randomised placebo‐controlled clinical trial

Setting/location: south of Iran

Study period: October 2007 to January 2009 (15 months)

Sample size calculation: not described

Participants

Type of Leishmania: L major and L tropica

Inclusion criteria: men and women aged at least 12 years old, positive smear and/or polymerase chain reaction (PCR) for CL, cutaneous lesions of less than 4 months' duration

Exclusion criteria: pregnancy and lactation, facial lesions, lesions of more than 4 months' duration, complete or incomplete treatment with a systemic or topical medication within the previous month, drug sensitivity

N randomised: 100 participants

Withdrawals: 37 participants

N assessed: 63 participants (36 in treatment and 27 in control group)

Age: range 12‐62 years (mean 32.6 (SD 12) in the treatment group and 33.1 (14) in the vehicle group)

Sex (male/female): 35/28

Baseline data:

  • Location of lesion(s): treatment: arm 12, hand 5, wrist 5, leg 7, foot 5, trunk 1, neck 1; control: arm 5, hand 7, wrist 4, leg 5, foot 2, trunk 2, neck 2

  • Type of lesion: treatment: plaque 19 and nodule 17; control: plaque 14 and nodule 13

  • Origin of the lesion: treatment: rural 24 and urban 12; control: rural 17 and urban 10

Interventions

Type of interventions:

  • Group 1: 3% sodium nitrite in aqueous cream

  • Group 2: aqueous cream alone (vehicle)

Duration of intervention: 12 weeks

Co‐interventions: simultaneously, cryotherapy once a week and aqueous cream containing 3% salicylic acid (emulsifying ointment 30 g, phenoxyethanol 1 g and freshly boiled and cooled water 69 g)

Outcomes

Clinical response:

  • Complete clinical response: complete re‐epithelialisation of the ulcer and disappearance of induration

  • Partial clinical response: reduction of more than 50% of the ulcer and induration in relation to the last clinical evaluation

  • Absence of clinical response: increase or reduction of less than 50% of the ulcer and the indurated area in relation to the last clinical evaluation

Adverse effects

Time points reported: 12 weeks

Notes

Study funding sources: not described

Possible conflicts of interest: no conflicts of interest

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "This investigation was designed as a 12‐week, double blind, N randomised, placebo‐controlled study and its protocol was approved by the ethics committee of the vice chancellor of research affairs of Shiraz University."

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "The identically coded and packaged creams were applied two times daily."

Quote: "The study was blinded to both the patients and the assessing physician."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The study was blinded to both the patients and the assessing physician."

Incomplete outcome data (attrition bias)
All outcomes

High risk

100 participants were enrolled but only 63 completed the study. Reasons for withdrawal were not reported.

Selective reporting (reporting bias)

Low risk

Our primary outcomes (cure and adverse effects) were described in Methods and reported in Results

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Kashani 2010

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Center for Research & Training in Skin Diseases & Leprosy, Tehran University of Medical Sciences, Tehran, Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L tropica and L major

Inclusion criteria: the participants with CL who attended to Center for Research & Training in Skin Diseases & Leprosy, Tehran University of Medical Sciences, Tehran, Iran. The diagnosis was based on clinical findings and physical examination and confirmed by Giemsa‐stained direct smear of the lesion

Exclusion criteria: ore than 5 lesions; lesions greater than 5 cm in diameter; received anti‐CL treatment during the previous month; lesions on the face and around vital organs; diabetic individuals; pregnant, lactating female participants; subjects with active infectious diseases

N randomised: 85. Group 1: mesotherapy administration of meglumine antimonate: 41. Group 2: ILMA injection: 44

Withdrawals: 25. Group 1: 11 (7 lost to follow‐up, 3 inadequate efficacy, 1 allergic reaction). Group 2: 14 (10 lost to follow‐up, 2 inadequate efficacy, 2 allergic reaction)

N assessed: 60 (70.59%). Group 1: 30 (73.17%), group 2: 30 (68.18%)

Mean (SD) age: Group 1: 25.24 years (16), group 2: 24.32 years (15.26)

Sex (male/female): Group 1: 19/11, group 2: 17/13

Baseline data:

  • Group 1: type of lesions (n (%)): papule 2 (6.66), nodule 6 (20), plaque 22 (73.33). Location of lesions (n (%)): upper extremities 15 (50), lower extremities 9 (30), other 9 (20).Ulcer (mean(SD), cm) (mean(SD), cm) 1.80 (0.64). Erythema (mean(SD), cm) 2.25 (0.66). Induration 0.91 cm (0.41)

  • Group 2: type of lesions (n (%)): papule 3 (10), nodule 7 (23.33), plaque 20 (66.66). Location of lesions (n (%)): upper extremities 13 (43.33), lower extremities 10 (33.33), other 7 (23.33). Ulcer(mean(SD), cm) 1.86 (0.55). Erythema (mean(SD), cm) 2.34 (0.69). Induration (mean (SD)) 1.12 cm (0.682)

Interventions

Type of interventions:

  • Group 1: "Inderm Mesotherapy Gun (Innovative Med. Inc., Irvine, CA, USA) was set to inject 0.05 mL of MA in each shot in the depth of 2 mm using a 30 G needle. The shots were repeated in each 5 mm of the lesion."

  • Group 2: 0.1 mL of MA (Glucantime, Rhone‐Poulenc, France) was injected intradermally in the periphery of the lesions using a 30‐G needle. The shots were repeated in each 1 cm of lesion until the whole lesion was blanched.

Duration of intervention: 6 weeks or until complete cure

Outcomes

Clinical cure: 3 categories were defined for ulcer healing: complete cure: complete re‐epithelialisation of the ulcer with loss of induration; partial cure: 25 to 99% re‐epithelialisation of lesions; failure: less than 25% re‐epithelialisation of lesions. Improvement of the lesions: ulcer improvement (cm), induration improvement (cm), induration improvement (cm), speed of improvement (cm/week), speed of erythema improvement (cm/week), scare size (cm)

Microbiological cure: Leismanian body: negative, positive.

Pain severity during injection: visual analogue scale (VAS; 0 to 10).

Amount of drug used (mL/cm²) and number of treatment sessions

Adverse effects

Time points reported: 3 months after treatment. Adverse effects: after the each treatment session.

Notes

Study funding sources: this study was supported by a research grant from Center for Research & Training in Skin Diseases & Leprosy, Tehran University of Medical Sciences, Tehran, Iran

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Eligible patients were randomised according to a computer generated simple randomisation list to be treated with intralesional injection of MA using either the classic method or mesotherapy gun."

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

High risk

14/44 participants with the conventional technique and 11/41 with mesotherapy gun withdrawn the study (29.4%)

Selective reporting (reporting bias)

Unclear risk

Protocol not available; not registered in a trial registry; SD not provided for pain severity (VAS)

Insufficient information to permit judgment

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Khatami 2012

Methods

Study design: randomised controlled trial

Setting/location: Bam and Mashhad in Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: parasitologically proven cases of CL; otherwise healthy subjects on the basis of medical history, physical examination and results of blood tests; age 12–50 years; body weight >40 kg; willingness to participate in the trial and signing the informed consent

Exclusion criteria: not described

N randomised: 138 participants (75 in IMMA and 63 in miltefosine)

Withdrawals: 33 in IMMA and 32 in miltefosine

N assessed: 42 in IMMA arm and 31 in miltefosine

Age (years): not described

Sex: not described

Baseline data: not described

Interventions

Type of interventions:

Group 1: oral miltefosine 2.5/kg/d for 28 days

Group 2: IMMA 60 mg/kg/d for 14 days

Duration of intervention: not described

Co‐interventions: not described

Outcomes

Healing of their lesions and occurrence

Adverse effects

Time points reported: participants were evaluated weekly for 28 days and then 1 month later

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote:"This open N randomised controlled trial was carried out in Bam and Mashhad in Iran …"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote:"This open N randomised controlled trial was carried out in Bam and Mashhad in Iran …"

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote:"This open N randomised controlled trial was carried out in Bam and Mashhad in Iran…"

Incomplete outcome data (attrition bias)
All outcomes

High risk

Losses to follow‐up quite high (around 50%)

Intervention group: 32/63= 50.8%

Control group: 33/75= 44%

Selective reporting (reporting bias)

Unclear risk

It is an abstract. At least they reported complete cure of cases and adverse effects.

Other bias

High risk

Sample size calculation, reporting of Leishmania spp involved and baseline comparability was not correctly reported

Khatami 2013

Methods

Study design: randomised, assessor‐blind, controlled clinical trial

Setting/location: Kashan, Tehran, Iran

Study period: September 2008 to April 2010 (19 months)

Sample size calculation: 56 lesions per treatment group were needed to have 80% power to detect a significant difference in the expected cure rate of 60% in the ILMA alone group and the desired cure rate of 85% in the ILMA and either silver or non‐silver dressing‐treated group at week 10 with a type I error level of 0.05. Compensating for a 20% loss‐to‐follow‐up, recruiting 68 (round up to 70) lesions per treatment group looked reasonable.

Participants

Type of Leishmania: L major

Inclusion criteria: parasitologically confirmed cases of CL based on positive smear and/or culture; otherwise healthy subjects on the basis of medical history; age of 12‐60 years; willingness to participate in the study and signing the informed consent form (by the participant or his/her parent/guardian in cases younger than 18 years).

Exclusion criteria: pregnant or lactating women; duration of lesion more than 3 months; number of lesions more than 5; ulcer size greater than 5 cm in largest diameter; history of receiving full course standard treatment (antimonials); history of allergy to MA or silver; serious systemic illnesses (as judged by the physician); participation in any drug trials in the last 60 days; indication for systemic treatment with MA; presence of secondary bacterial infection of the lesion according to clinical appearance

N randomised: 83 participants (158 lesions) ILMA: 26 participants (45 lesions); ILMA + non‐silver dressing: 26 participants (53 lesions); ILMA + silver dressing: 31 participants (60 lesions)

Withdrawals: 10 participants (18 lesions)

N assessed: 73 (140 lesions)

Mean (SD) age: 28.81 years (14.45). ILMA: 32.88 years (12.92); ILMA + non‐silver dressing: 30.31 years (15.41); ILMA + silver dressing: 24.13 years (13.97)

Sex: male 39; ILMA: male 8; ILMA + non‐silver dressing: male 11; ILMA + silver dressing: male 20

Baseline data:

  • Mean duration of lesions (SD): 7.951 weeks (2.67); ILMA: 8.31 weeks (2.86); ILMA + non‐silver dressing: 8.46 weeks (2.15); ILMA + silver dressing: 7.23 weeks (2.82)

  • Mean number of lesions (SD): 1.90 (1.21); ILMA: 1.77 (1.34); ILMA + non‐silver dressing: 2.00 (1.47); ILMA + silver dressing: 1.94 (1.12)

  • Induration area median (IQR): 169 mm² (79‐433); ILMA: 270 mm² (120‐557); ILMA + non‐silver dressing: 156 mm² (67‐490); ILMA + silver dressing: 141 mm² (48‐346).

  • Ulceration area median (IQR): 6 mm² (1‐25); ILMA: 8 mm² (2‐37); ILMA + non‐silver dressing: 6 mm² (0‐15); ILMA + silver dressing: 6 mm² (0‐24)

Interventions

Type of interventions:

  • Group 1: eligible participants were randomly allocated into 2 groups and treated for 6 weeks with either:

    • Weekly injections of ILMA combined with application of a non‐silver dressing (AtraumanH, Hartmann, CMC Consumer Medical Care GmbH, Germany) on the lesions, or

    • Weekly injections of ILMA combined with application of a silver containing dressing (AtraumanH Ag, Hartmann, CMC Consumer Medical Care GmbH, Germany) on the lesions.

  • Group 2: weekly injections of ILMA (GlucantimeH; Rhodia Laboratories, Rhone‐Poulenc, France) alone

Duration of intervention: 6 weeks

Outcomes

Clinical cure (complete healing defined as more than 75% reduction, clinical improvement defined as 50%–75% reduction, and no response to treatment defined as less than 50% reduction in the size of the lesion compared with baseline)

Relapse: defined as a reappearance of lesions at the site or periphery of previously healed lesions or an increase in the size of lesions after initial improvement was assessed 5 months after the termination of treatment

Adverse effects: itching and burning, exudation, oedema, and dermatitis

Time points reported: oucomes were assessed at the end of the treatment period (end of week 6), then at 4 weeks and 5 months after the last treatment session

Notes

Study funding sources: the budget of the research project has been provided by the Vice‐Chancellery of Research of Tehran University of Medical Sciences.

Possible conflicts of interest: the authors have declared that no competing interests exist.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A random sequence generated by using the online software Random Sequence Generator, which is available at URL: www.random.org [22]. It was done by an investigator with no clinical involvement in the trial. R. Talaee was responsible for enrolment of the patients."

Allocation concealment (selection bias)

Low risk

Quote: "The method for randomisation concealment was to use sequentially numbered, opaque, sealed envelopes (SNOSE). The envelopes were kept in a safe box, which was only accessible to A. Khatami who was responsible for assigning the patients to the interventions."

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "An assessor who was blinded to the type of treatment visited the patients at weekly intervals during the treatment period and 1‐ month and 5 months after the last treatment session"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

An intent‐to‐treat analysis was performed at 2 time points (end of the treatment period (day 42) and one month later (day 72)). The numbers of and the reasons for withdrawals were not significantly different between the 3 groups: 11% ILMA group; 19% ILMA + non‐silver and 6.45% ILMA + silver

Selective reporting (reporting bias)

Low risk

Comment: all relevant outcomes were reported

Other bias

Low risk

Other items assessed correctly reported

Kochar 2000

Methods

Study design: randomised controlled trial

Setting/location: India

Study period: 5 months

Sample size calculation: not described

Participants

Type of Leishmania: L tropica

Inclusion criteria: participants with a confirmed diagnosis of L tropica. The diagnosis was confirmed by demonstrations of L tropica bodies (amastigote stage of L tropica under oil immersion).

Exclusion criteria: no mention about inclusion or exclusion criteria

N randomised: 50. Rifampicin 1200 mg/d orally: 25; placebo: 25

Withdrawals: 4. Rifampicin 1: 2 (lost to follow‐up); placebo: 2 (withdrew due to exacerbation of lesions)

N assessed: 46. Rifampicin: 23; placebo: 23

Mean (SD) age: rifampicin 29.54 years (17.34); placebo: 26.96 years (10.96)

Sex (male/female): rifampicin: 10/13; placebo: 9/14

Baseline data:

  • Type of lesions (% rifampicin versus placebo): ulcerative: 21.7 versus 17.4, nodular: 30.4 versus 30.4; nodule‐ulcerative: 17.4 versus 30.4, and erythematous plaques: 30.4 versus 21.7.

  • Regarding the distribution (%): face: 17.5 versus 20.9; neck; 3.5 versus 0; shoulder: 1.75 versus 0; arms: 17.5 versus 20.9; hand: 19.2 versus 18.6; legs: 28 versus 16.2; feet: 8.77 versus 20.9, and abdomen: 3.5 versus 2.32

  • Mean number of lesions (SD): rifampicin 2.92 (4.61); placebo 2.08 (0.78)

  • Mean duration of lesions (SD): rifampicin 3.58 months (6.14); placebo: 3.23 months (2.93)

  • Mean size of lesions (SD): rifampicin 31.91 mm² (15.99); placebo: 37.04 mm² (12.37)

Interventions

Type of interventions:

  • Group 1: rifampicin 1200 mg/d orally

  • Group 2: placebo capsules orally

Duration of intervention: 2 divided doses for 4 weeks

Duration of follow‐up: 4 weeks

Co‐interventions: the 2 withdrew due to exacerbation of lesions in placebo group and were treated by another therapeutic regimen

Outcomes

Healing rates: percentage of participants 'cured' at the end of treatment: a cure was defined as complete healing and disappearance of the lesion or reversible hypopigmentation at the site of lesion. Incomplete or partial healing was defined as a reduction in the size of a lesion and the absence of parasites on smear. A treatment failure or non‐healing was defined as the absence of any change in the lesion and persistence of parasites on smear

Adverse effects

Tertiary outcomes: microbiological or histopathological cure of skin lesions

Time points reported: at the end of the treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomized"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Comment: "Randomized" but no further information provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Comment: "Double‐blinded" but no further information provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: "Double‐blinded" but no further information provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing outcome data balanced in numbers across intervention groups

Selective reporting (reporting bias)

Low risk

Comment: all relevant outcomes were reported

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Kochar 2006

Methods

Study design: randomised controlled trial

Setting/location: hospital, Bikaner, India

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L tropica

Inclusion criteria: participants with a confirmed diagnosis of anthroponotic CL.

Exclusion criteria: —

N randomised: 50. Intervention group: 25, placebo group: 25

Withdrawals: 6 lost to follow‐up. Intervention group: 2, placebo group: 4

N assessed: 44. Intervention group: 23, placebo group: 21

Age: intervention group: n = 12, 16‐20 years; n = 5, 21‐30 years; n = 3, 31‐40 years; n = 2, 41‐50 years; n = 2, 51‐60 years; n = 0, 61‐70 years. Placebo group: n = 10, 16‐20 years; n = 4, 21‐30 years; n = 5, 31‐40 years; n = 3, 41‐50 years; n = 2, 51‐60 years; n = 1, 61‐70 years

Sex (male/female): 34/16. Intervention group: 17/8; placebo group: 17/8

Male predominance was probably due to the habit of sleeping in open spaces outside the house without mosquito nets and improper clothing during night when the sandflies are active.

Baseline data:

  • Type of lesion:

    • Intervention group: erythematous nodular: 11; erythematous plaque: 3; ulcerative: 3; erythematous ulcerative: 2; nodular: 4; nodular ulcerative: 2

    • Placebo group: erythematous nodular: 9; erythematous plaque: 5; ulcerative: 2; erythematous ulcerative: 5; nodular: 2; nodular ulcerative: 2

  • Ratio lesions:participant

    • Intervention group: 1:14; 2:5; 3:5; > 3:1

    • Placebo group: 1:14; 2:7; 3:4; > 3:0

  • Distribution of lesions:

    • Intervention group: face: 4; neck: 3; arms: 6; hand: 9; back: 2; abdomen: 1; buttock: 1; legs: 4; foot: 3

    • Placebo group: face: 11; neck: 2; arms: 6; hand: 9; back: 2; abdomen: 3; buttock: 1; legs: 3; foot: 4

  • Most cases were presented with 1 to 3 months of duration of the disease.

Interventions

Type of interventions:

  • Group 1: rifampicin (1200 mg/d) in 2 divided doses + omeprazole 20 mg

  • Group 2: placebo

Duration of intervention: 6 weeks

Duration of follow‐up: 6 weeks

Outcomes

Healing rates: percentage of participants 'cured' at the end of treatment: complete healing; partial response; no response

Time points reported: at the end of treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomized"

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Comment: "Randomized" but no further information provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Comment: "Double‐blinded" but no further information provided

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: "Double‐blinded" but no further information provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing outcome data balanced in numbers across intervention groups, with similar reasons for missing data across group

Selective reporting (reporting bias)

High risk

Comment: no adverse effects information reported

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Larbi 1995

Methods

Study design: randomised controlled trial

Setting/location: Saudi Arabia

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania:L major

Inclusion criteria: only participants in whom parasites (amastigotes) were demonstrated were enrolled

Exclusion criteria: pregnancy, chronic illness, immunocompromised or hyper allergic reaction to the trial drugs, treatment with regular medications such as antituberculous agents and steroids, which might have affected specific therapy, treatment with anti‐leishmanial drugs within the previous months, the presence of scars of previously treated lesions

N randomised: 58. 2% miconazole cream: 27; 1% clotrimazole cream: 31

Withdrawals: 4: 2% miconazole cream: 4 (lost to follow‐up); 1% clotrimazole cream: 0

N participants assessed (lesions): 54 (151). 2% miconazole cream:n23 (62); 1% clotrimazole cream: 31 (89)

Mean (SD) age: 2% miconazole cream: 19.5 years (15.4); 1% clotrimazole cream: 21.5 years (12.8)

Sex (male/female): 42/12. 2% miconazole cream: 17/6; 1% clotrimazole cream: 25/6

Baseline data:

  • Lesion type (%): 2% miconazole group: nodular: 21 (34%), nodule‐ulcerative: 37 (59%) and papular: 4 (7%). 1% clotrimazole group: nodular: 36 (41%), nodule‐ulcerative: 50 (56%) and papular: 4 (3%)

  • Location of lesions: 2% miconazole group(percentage): the lower limbs: 56%; upper limbs 39%; head, neck, upper chest: 5%. 1% clotrimazole group: lower limbs: 34%; upper limbs: 46%; head, neck, upper chest: 19%, abdomen and groin: 1%.

  • 2% miconazole group: MNL: Saudi: 2.2 and non‐Saudi 3.4. MSL (range): 1.02 cm (0.06‐5). Mean duration of lesions before treatment (SD): 2.1 weeks (1.3)

  • 1% clotrimazole group: MNL: Saudi: 2.3 and non‐Saudi 3.5. MSL (range): 1.38 cm (0.05‐6). Mean duration of lesions before treatment (SD): 2.3 weeks (1.2)

Interventions

Type of interventions:

  • Group 1: 2% miconazole cream

  • Group 2: 1% clotrimazole cream

Duration of intervention: twice daily for 30 consecutive days

Duration of follow‐up: 30 days

Outcomes

Healing rates: percentage of lesions 'cured' at the end of treatment. Response to treatment was based on results of repeated parasitologic evaluation and/or clinical changes using the same parameters as in the follow‐up. On this basis, response to treatment was graded into 4 categories:

  • Lesion fully healed (lesions were completely healed and parasitologically negative

  • Size reduced (lesions showed a reduction in infiltration, erythema, and size with positive or negative parasitology

  • No change (lesions did not show any change in infiltration, erythema, size, and parasitology

  • Lesions worsened or size increased (there was an increase in the size of the lesion and a worsening of infiltration and erythema and positive parasitology)

Adverse effects

Time points reported: at the end of treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomization was carried out by using a table of random numbers"

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Patients admitted into the trial were given unlabeled tubes containing either 1% clotrimazole cream or 2% miconazole"

Comment: we think that the outcome is unlikely to be influenced

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Of 27 participants treated with 2% miconazole, 4 defaulted and were lost to follow‐up; they were excluded from analysis" (4/27 = 14.28%)

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but he published reports probably include all expected outcomes.

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Layegh 2007

Methods

Study design: randomised, prospective

Setting/location: dermatology department of Qaem University Hospital, Mashhad, Iran

Study period: from October 2004 through November 2005 (13 months)

Sample size calculation: not described

Participants

Type of Leishmania: not reported (although they reported that CLL is endemic and L tropica and, less commonly, L major are the most prevalent species)

Inclusion criteria: aged > 2 years with CL of < 6‐months' duration; no treatment of the current infection during the last 3 months; and no history of a renal or liver disease

Exclusion criteria: pregnant women; history of intolerance or allergy to Glucantime or macrolides;

simultaneous use of antacids containing Al and Mg, theophyline, phenytoin, barbiturates, carbamazepine, and cyclosporin

N randomised: 49: azithromycin: 22 (35 lesions), IMMA: 27 (58 lesions).

Withdrawals: 2 (azithromycin group)

N assessed: 47 participants; 20 (29 lesions) in the azithomicin group and 27 (58 lesions) in the IMMA group.

Mean (SD) age: range 4‐70 years; azithromycin: 21.77 years (17.13) and IMMA: 22.78 years (13.17)

Sex (male/female): 24/25; azithromycin: 8/14; IMMA: 16/11

Baseline data:

  • Mean number of lesions (SD): azithromycin: 1.86 (1.52) and IMMA: 2.15 (1.38)

  • Mean duration of lesions (SD): azithromycin: 4.28 months (1.39) and IMMA: 3.22 months (2.03)

  • Type of lesions:

    • Papule and plaque: azithromycin: 27 (77.1%); IMMA: 49 (84.5%)

    • Nodular: azithromycin: 6 (17.1%); IMMA: 7 (12.1%)

    • Ulcerative: azithromycin: 2 (5.8%); IMMA: 2 (2.8%)

Interventions

Type of interventions:

  • Group 1: "daily oral dose of 500 mg of azithromycin for 5 days/month in adults and 10 mg/kg in children; treatment cycles were repeated monthly when no favourable response was achieved, up to a maximum duration of 4 months"

  • Group 2: 60 mg/kg IMMA for 20 days

Duration of intervention: up to 4 months (azithromicin) and 20 days (IMMA)

Co‐interventions: participants showing no favourable response after 4 months of azithromycin therapy would receive IMMA at the same dose as the other group, at the end of the study.

Outcomes

Clinical cure: clinical response was determined on the basis of the lesions' size, border, induration, and infiltration of papulonodular lesions; ulcerative lesions were evaluated on the basis of the ulcer depth, diameter, crusting, and the extent of re‐epithelialisation

  • Complete or significant improvement: decrease in the induration size > 75% or full re‐epithelialisation of the le‐scions and a negative direct skin smear (absence of parasites in the lesion)

  • No improvement: decrease in induration size < 30%

  • Partial improvement: any changes between above criteria

Recurrence

Adverse effects

Time points reported: participants in the azithromycin group were evaluated monthly up to a maximum of 4 months, but participants in the IMMA group were evaluated both at the end of the treatment period (day 20) and 45 days later. Recurrence was reported after the 16‐week follow‐up period.

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The 49 patients who met our inclusion criteria were randomly divided into two groups."

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

48 of 50 participants randomised completed the study. 2/20 participants of the azithromycin group withdrew because of adverse effects. Applications of study drugs were conducted according to the protocol

Selective reporting (reporting bias)

Unclear risk

Protocol not available

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Layegh 2009

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Dermatology Clinic of Qaem Hospital in Mashad, Iran

Study period: 10 months

Sample size calculation: to detect a 22% difference in the cure rate between cryotherapy and ILMA groups and assuming a 57% cure rate in the cryotherapy group, with a 65% power and a 10% 2‐sided type I error, 40 persons were needed in each group. The 79 participants who met our criteria were randomly divided into 2 groups: 40 participants received cryotherapy and 39 participants received ILMA

Participants

Type of Leishmania: L tropica

Inclusion criteria: positive direct skin smears for CL, were ≤ 13 years of age, had visited the dermatology clinic from September 2006 through June 2007, and had lesions with a duration of < 12 weeks to exclude any natural self‐healing during follow‐up

Exclusion criteria: > 13 years of age, had a lesion history > 3 months, allergic to antimonial drugs, simultaneously using any other therapeutic method

N randomised: 79. Cryotherapy: 40, ILMA: 39

Withdrawals: 7: cryotherapy group: 4, ILMA: 3

N assessed: 72 (91.14%): cryotherapy group: 36 (90%), ILMA: 36 (92.31%)

Mean (SD) age: 6.8 years (3.4) in cryotherapy group and 6.2 years (3.4) in ILMA group

Sex (male/female): cryotherapy: 18/22; ILMA: 20/19

Baseline data:

  • Mean number of lesions (SD): cryotherapy: 1.9 (1.02); ILMA: 1.4 (0.76)

  • Location of lesions by site:

    • Head and neck: cryotherapy: 24, ILMA: 31

    • Upper limb (hand): cryotherapy: 10, ILMA: 13

    • Lower limbs (foot): cryotherapy: 4, ILMA: 3

  • Type of lesion:

    • Papule/plaque: cryotherapy: 35, ILMA: 37

    • Nodule: cryotherapy: 2, ILMA: 3

    • Ulcer: cryotherapy: 1, ILMA: 2

  • Mean duration of lesions (SD): cryotherapy: 11.2 weeks (11.8); ILMA: 12.1 weeks (18.4)

Interventions

Type of interventions:

  • Group 1: cryotherapy group: liquid nitrogen (−195°C) was applied twice to the lesion. Each cycle was 10–15 s of freezing time with a thawing interval of 20 s. Care was taken to ensure that freezing reached up to a few millimetres within the healthy skin surrounding the lesion

  • Group 2: ILMA (Glucantime; Specia, Paris, France) in a volume of 0.5–2 cm³ was injected into each lesion until the lesion was completely infiltrated (blanched)

Duration of intervention: 6 weeks

Outcomes

Clinical cure: complete cure was defined as full re‐epithelialisation; disappearance of oedema, induration, and other signs of inflammation; and a negative direct skin smear result. Score:

  1. Complete improvement (full re‐epithelialisation of the lesions and a negative direct skin smear result),

  2. Significant improvement (decrease in induration size > 75%),

  3. Partial improvement (decrease in induration size between 50% and 75%,

  4. Slight improvement (decrease in induration size between 25% and 50%

  5. No improvement (decrease in induration size < 25%)

Time to healing

Adverse effects

Time points reported: at the end of treatment and 6 months (clinical cure); at 4, 5 and 6 weeks (time to healing); at the end of treatment, after 6 weeks (adverse effects)

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Open trial; lesions of all participants were photographed.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

4 participants in the cryotherapy group and 3 participants in the ILMA group were excluded because they received 2 medications simultaneously, did not complete the treatment course, did not visit the clinic for follow‐up 6 months later, or changed their address and were lost to follow‐up

Selective reporting (reporting bias)

Low risk

Protocol not available; not registered; however, pre‐specified outcomes of the review were reported

Other bias

Low risk

Other items assessed correctly reported

Layegh 2011

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Dermatology Clinic of Ghaem Hospital, Mashhad, Iran

Study period: March 2008 to September 2010 (30 months)

Sample size calculation: to detect a 35% difference in the cure rate between the 2 groups of liposomal AmB and ILMA, assuming a 90% cure rate in liposomal AmB group with a 95% power and a 5% 2‐sided type I error, 36 subjects were required in each group. Also regarding a loss to follow‐up of 20%, this number was raised to 50 participants for each group

Participants

Type of Leishmania: L tropica, L major

Inclusion criteria: cutaneous leishmaniasis approved by either a direct smear stained with Giemsa or a positive skin biopsy of lesions with less than 6‐month duration, in cases with a previous history of anti‐leishmaniasis therapy, a 3‐month treatment‐free interval from the last treatment course

Exclusion criteria: pregnancy, breastfeeding, taking any other specific treatment while participating in the study, past medical history of any local or systemic disease during the last 2 months, a significant underlying disease such as cardiac, renal, or liver dysfunction

N randomised: 110. Group 1: 50; group 2: 60

Withdrawals: 34 did not adhere. Group 1: 11; group 2: 23

N assessed: group 1: 39 (78%); group 2: 37 (61.7%)

Mean (SD) age: group 1: 20.54 years (18.72); group 2: 25.30 years (15.70)

Sex (male/female): group 1: 23/27; group 2: 21/39

Baseline data:

  • Mean number of lesions (SD): group 1: 1.91 (1.02); group 2: 1.4 (0.76)

  • Location of the target lesion:

    • Group 1: head and neck, 26; hand, 18; leg and trunk, 6

    • Group 2: head and neck, 22; hand 32; leg and trunk, 6

  • Type of lesions:

    • Group 1: papule/plaque, 33; nodule, 8; ulcer, 9

    • Group 2: papule/plaque, 53; nodule, 5; ulcer, 2

Interventions

Type of interventions:

  • Group 1: liposomal AmB was administered as 3–7 drops twice daily according to the lesion size

  • Group 2: ILMA (Glucantime; Specia, Paris, France) was injected into each lesion once a week, to the point when the lesion's surface became fully infiltrated and up to a maximum dose of 2 mL

Duration of intervention: 8 weeks

Outcomes

Clinical response: determined on the basis of the lesion induration size of and the extent of re‐epithelialisation in ulcerative ones. Scales: clinical response:

  • Slight improvement: decrease in induration size up to 25%

  • Mild improvement: decrease in induration size between 25 and 50%

  • Moderate improvement: decrease in induration size between 50 and 75%

  • Marked improvement: decrease in induration size more than 75%

Adverse effects

Time points reported: at the end of treatment, and 6 months after termination. Adverse effects were reported at the end of treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The 110 patients who met our inclusion criteria were randomly divided into two groups."

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT analyses performed

Losses to follow‐up per group:

Intervention group: 11/50 (22%)

Control group: 23/60 (38.3%)

Selective reporting (reporting bias)

Unclear risk

Number of lesions higher in the liposomal ampB group (P < 0.05) whereas number of papule and plaque lesions where higher in the ILMA group (P = 0.011)

Outcomes described in the methods were reported in the results section. However, only one time point was reported

Protocol not available; not registered in a clinical trial registry

Other bias

Low risk

Other items assessed correctly reported

Lynen 1992

Methods

Study design: randomised controlled trial

Setting/location: school children, Sudan

Study period: 80 days

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania spp not specified but L major was presumed to be the causative parasite.

Inclusion criteria: all positive smears

Exclusion criteria: children who had received diminazene aceturate or previous treatment in hospital

N randomised: 70. 35 in each group

Withdrawals: 8. Berelin: 3; Savlon: 5. 2 children in Berelin group were not enrolled because they received diminazene aceturate before. 2 in Savlon group were excluded because they were treated for CL in the hospital

N assessed: 62. Berelin: 32; Savlon: 30

Mean (SD) age: Berelin: 9.3 years (4.6); Savlon: 10.4 years (2.8)

Sex (male/female): Berelin: 18/15; Savlon: 20/13

Baseline imbalances: comparability with regard to sex, age, number, size, duration and location of ulcer, and previous treatment taken

  • Size of lesion (% (n/N)): small lesions (< 2 cm): Berelin: 58% (19/33); Savlon: 61% (20/33); big lesions (≥ 2 cm): Berelin: 42% (14/33); Savlon: 39% (13/33)

  • Duration (% (n/N)): ≤ 2 months: Berelin: 52% (17/33); Savlon: 64% (21/33). >2 months: Berelin: 48% (16/33); Savlon: 36% (12/33)

Interventions

Type of interventions:

  • Group 1: Berelin (1.05 g diminazene aceturate in 2.36 g of granulate, dissolved in 12.5 cc of distilled water) daily except on Fridays for 50 days

  • Group 2: Savlon (cetrimide 15% + chlorhexidine 1.5% in a 2% solution) for 50 days

Duration of treatment: 50 days

Duration of follow‐up: one month

Outcomes

Primary outcome: percentage of participants 'cured' at the end of treatment. Cure was defined when the skin lesion was completely closed and covered with scar tissue, without the possibility to evoke secretions on pressure and if this apparent cure was sustained for at least 2 weeks

Secondary outcomes: duration of remission and percentage of people with treated lesions that recur 20 to 35 days after cure

Adverse effects

Time points reported: at the end of treatment and at the end of follow‐up.

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

A double‐blind study design was attempted, but could not be sustained as Savlon is a well‐known product and soapy on application

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Low rates of dropouts (8/70, < 2.5%)

Selective reporting (reporting bias)

Low risk

The study protocol is not available but it is probably that the published reports include all expected outcomes

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Maleki 2012

Methods

Study design: randomised controlled trial

Setting/location: Imam Reza Hospital in Mashhad, Iran

Study period: March to September 2004 (7 months)

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: confirmation of cutaneous leishmaniasis based on direct smear, ≤ 3 lesions, duration of the disease < 12 weeks, aged 7‐60 years, completion of informed consent form by participant or parents of minor participants, dry cutaneous leishmaniasis

Exclusion criteria: pregnant or breastfeeding women; children < 7 years, lesions on ear, nose, joints, and near the eye; > 3 lesions; application of any kind of treatment for cutaneous leishmaniasis, duration of the disease longer than 12 weeks (to omit spontaneous healing cases during the follow‐up period), recurrent infection, wet cutaneous leishmaniasis

N randomised: 45 participants

Withdrawals: 11

N assessed: 34 participants (24, group 1; 10, group 2)

Age: not described

Sex: not described

Baseline data: mean size of lesions (SD): group 1, 1.27 cm (0.81); group 2, 0.98 cm (0.61)

Interventions

Type of interventions:

  • Group 1: 2 bouts of intralesional 2% zinc sulphate was performed twice within 2 weeks interval

  • Group 2: 6 weekly bouts of ILMA

Duration of intervention: group 1, 2 weeks; group 2, 6 weeks

Outcomes

Clinical response

  • Slight: partial reduction of erythema and oedema

  • Mild: reduction of lesion size up to 30%

  • Moderate: reduction of lesion size between 30% and 60%

  • Marked: reduction of lesion size more than 60% or negative smear

  • Total tolerance: complete healing of the lesion with negative smear

Time points reported: 8 weeks

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "This N randomised controlled trial was performed from March to September 2004 on patients with cutaneous leishmaniasis admitted to Imam Reza Hospital in Mashhad"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

45 participants were randomised into 2 groups. 11 were lost to follow‐up (24%).

No ITT analyses were performed

High imbalance between groups

Selective reporting (reporting bias)

High risk

No baseline data. Side effects not clearly reported (no statistical analyses performed either)

Other bias

High risk

Sample size calculation, reporting of Leishmania spp involved and baseline comparability was not correctly reported

Mapar 2010

Methods

Study design: randomised controlled trial

Setting/location: Ahvaz city, southern Iran

Study period: 12 months (2002)

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: clinically and parasitologically positive (Leishman bodies in Giemsa‐stained direct smear of the lesion scrapings), duration of lesions < 3 months, aged 15‐40 years, no evidence of secondary bacterial infections, no mucosal involvement, no history of previous treatment of cutaneous leishmaniasis, or of allergic reactions to MA or metronidazole

Exclusion criteria: history of underlying disease (such as cardiac, renal, or pulmonary); pregnant or breastfeeding; cutaneous leishmaniasis localised on or near the joints

N randomised: 36. Group 1: 18, group 2: 18

Withdrawals: 8. Group 1: 2, group 2: 6

N assessed: 28 (77.78%). Group 1: 16, group 2: 12

Age: mean 28.8 years (range 15‐40)

Sex (male/female): 21/15

Baseline data:

  • Total lesions: group 1: 29, group 2: 27

  • Group 1: lesions were of dry types (without any discharge), the longest diameters of the lesions were 0.5‐2.5 cm

  • Group 2: clinically dry type skin lesions. The longest diameters of the lesions were 0.5‐2.5 cm

Interventions

Type of interventions:

  • Group 1: weekly ILMA injections (150‐600 mg = 0.5‐2 mL of MA ampoule for each skin lesion). Intralesional injections were administered intradermally enough to blanch the lesions surface.

  • Group 2: weekly IL metronidazole (500 mg/100 mL vials were made by Fresenius, Bad Homburg), injected intradermally into each skin lesion enough to blanch the lesions surface (0.5‐2 mL for each lesion)

Duration of intervention: 8 weeks

Outcomes

  • Clinical cure of the lesions: clinical criteria for cure were: complete re‐epithelialisation, disappearance of oedema, induration, and other signs of inflammation, flattening of the lesions and change of colour from erythematous to blue or dark grey

  • Adverse effects: local or systemic adverse effect. Pain of intralesional injection. Local inflammatory reactions with oedema and induration. In both groups the participants complained of severe pain at the site of injections but group 2 had unbearable terrible pain

Time points reported: clinical cure: after 8 injections. Adverse effects: after the treatment.

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The method of randomisation was selecting a card among 36 cards with odd or even numbers

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

High risk

28/36 participants completed the study (77.8%)

Selective reporting (reporting bias)

High risk

No protocol available. Adverse effects not reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Mashood 2001

Methods

Study design: randomised controlled trial

Setting/location: skin department of PNS Shifa, a naval hospital in Karachi. Endemic areas of Balochistan province, Pakistan

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania spp not mentioned

Inclusion criteria: never received any treatment for their skin disease

Exclusion criteria: very young and very old people, those who had received some treatment for the disease, people suffering from diffuse CL or leishmaniasis recidivans, people with some known cardiac, renal or hepatic disease

N randomised: 40. 20 in each group

Withdrawals: 0

Participants assessed: 40 (100%). 20 in each group

Age: range 20‐40

Sex: 100% male

Baseline data. Nodular, ulcerative and crusted forms were the most common morphological patterns seen. Most lesions were on hands and feet.

  • Group 1: MSL: 570.7 mm². MDLBT (range): 8.6 weeks (4‐20)

  • Group 2: MSL: 960.3 mm². MDLBT: 12.6 weeks (8‐24)

Interventions

Type of interventions:

  • Group 1: oral AL 20 mg/kg/d in 3‐4 divided doses tablet

  • Group 2: IVSSG 20 mg/kg/d

Duration of intervention: 15 days

Duration of follow‐up: 3 months

Outcomes

Healing rates: percentage of participants 'cured' at the end of treatment

Adverse effects

Time points reported: at the end of treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Low risk

The study protocol is not available but it is probably that the published reports include all expected outcomes

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Mohebali 2007

Methods

Study design: randomised controlled trial

Setting/location: Golestan province, in northeastern Iran

Study period: 20 November 2005 to 20 July 2006 (8 months)

Sample size calculation: not described

Participants

Type of Leishmania: L major

Inclusion criteria: observation of Leishman bodies (amastigotes) in dermal lesions, no previous use of anti‐leishmanial drugs, no previously confirmed leishmaniasis (by scar or clinically compatible history), no pregnant or lactating women, no acute or chronic medical condition and no history of allergy, female participants of childbearing age were included after giving consent for effective contraception during therapy and until 3 months thereafter

Exclusion criteria: not explicitly reported, but can be inferred from the above inclusion criteria

N randomised: 63. Group 1: 32; group 2: 31

Withdrawals: 5. Group 1: 4 (lost after first week because lack of gastrointestinal tolerance); group 2: 1 (lost to follow‐up after 3 months)

N assessed: 63 (100%). Group 1: 28; group 2: 30

Mean age: group 1: 20.2 years; group 2: 16.8 years

Sex (male/female): 35/28. Group 1: 19/13; group 2: 16/15

Baseline data:

  • Location of the target lesion (%): < 20% of lesions were located on the face in each group: group 1: 12.5%; group 2: 16.1%

  • Mean number of lesions (range): group 1: 2.7 (1‐15); group 2: 2.5 (1‐10)

  • Mean size of lesions (range): group 1: 27.1 mm² (0.9‐80.0); group 2: 19.3 mm² (3‐67.3)

  • Mean duration of lesions: group 1: 45.5 days; group 2: 43.6 days

Interventions

Type of interventions:

  • Group 1: miltefosine orally 2.5 mg/kg daily for 28 days. Miltefosine capsules were administered as follows: 9‐14 kg, 3 capsules of 10 mg; 15‐29 kg, 1 capsule of 50 mg; 30‐45 kg, 2 capsules of 50 mg; 46‐84 kg, 3 capsules of 50 mg

  • Group 2: IMMA at 20 mg SbV⁵/kg body weight daily for 14 days

Duration of intervention: miltefosine: 28 days. IMMA: 14 days

Duration of follow‐up: 6 months

Outcomes

Healing rates: percentage of participants 'cured' 3 months after treatment

Secondary outcomes: duration of remission and percentage of people with treated lesions that recur within 6 months

Adverse effects

Time points reported: 2 weeks, 3 months, 6 months

Notes

Study funding sources: this study was financially supported by Medical Sciences/University of Tehran, Iran (from grant for full professor)

Possible conflicts of interest: not described

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "For randomisation of patients into two groups, we used balanced block method"

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "This study was an open‐label, N randomised comparison of miltefosine to meglumine antimonate in 63 patients"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Imbalance in numbers and reasons for missing data across intervention groups (4/32 = 12.5% miltefosine group; 1/31=3.22% IMMA group)

Comment: the results for the dropouts were not excluded from the statistical analysis.

We think missing outcome data likely to be related to true outcome.

Selective reporting (reporting bias)

Low risk

The study protocol is not available but it is probably that the published reports include all expected outcomes

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Momeni 1996

Methods

Study design: randomised controlled trial

Setting/location: Iran

Study period: not described

Sample size calculation: assuming a 30% spontaneous cure rate with placebo and the minimum usefulness of a 60% cure rate with itraconazole, and a 10% dropout rate, the sample size was calculated as 70 individuals in each group

Participants

Type of Leishmania: L major

Inclusion criteria: age > 12 years, diagnosis of CL confirmed by laboratory test results, no previous treatment for leishmaniasis, no serious concomitant medical problems

Exclusion criteria: age < 12 years, pregnant and nursing women, patients who had lesions on the face, lesions lasting > 4 months

N randomised: 140. Itraconazole: 70; placebo: 70

Withdrawals: itraconazole: 5

N assessed (lesions): 131. Itraconazole: 65 (219); placebo: 66 (262)

Age: mean 26.3 years (range 12‐46)

Sex (male/female): 90/41. Itraconazole: 44/21; placebo: 46/20

Baseline data:

  • Number of lesions: itraconazole: 219; placebo: 262

  • Mean duration of lesions (SD): itraconazole: 38 days (1.45 months); placebo: 45 days (1.5 months)

Interventions

Type of interventions:

  • Group 1: itraconazole orally 7 mg/kg/d (max. 400 mg per day)

  • Group 2: placebo capsules

Duration of intervention: 3 weeks

Duration of follow‐up: 51 days

Outcomes

Healing rates: percentage of participants 'cured' 51 days after treatment

Adverse effects

Time points reported: at the end of treatment (day 21); on day 51 (1 month after the end of treatment)

Notes

Study funding sources: this work was supported by the World Health Organization Special Program for Research and Training in Tropical Diseases and the research programme of Isfahan University of the Medical Sciences.

Possible conflicts of interest: not described

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Carried out by the WHO Special Programme for Research and Training in Tropical Diseases group in Geneva, Switzerland"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Identical capsules containing 100 mg of itraconazole or placebo were used in the study. The 2 treatment groups received either itraconazole as coded capsules for 21 days or placebo as coded capsules with the same dosages.

Review authors judge that the outcome is not likely to be influenced by lack of blinding

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reasons for missing outcome data unlikely to be related to true outcome

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes, including those that were pre‐specified. The study published reports our primary outcomes: 'cured' and adverse effects

Other bias

Low risk

Other items assessed correctly reported

Momeni 2002

Methods

Study design: randomised controlled trial

Setting/location: Skin Research Centre in Isfahan city. Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L major

Inclusion criteria: age > 5 years, diagnosis of CL confirmed by laboratory test results, no previous treatment for leishmaniasis, no serious concomitant medical problems, signed written informed consent

Exclusion criteria: pregnant and nursing women, age < 5 years, lesions lasting for > 4 months

N randomised: 72. Group 1: 36; group 2: 36

Withdrawals: 6, lost to follow‐up. Group 1: 5; group 2: 1

N participants assessed (lesions): 66 (196). Group 1: 31 (91); group 2: 35 (105)

Mean age (male/female): 21.9 years/17.6 years (range 5‐48 years). Group 1: 23.6 years/17.4 years; group 2: 20.2 years /18.1 years

Sex (male/female): 50 (75.7%)/16 (24.3%). Group 1: 26/5; group 2: 24/11

Baseline data:

  • Number of lesions: group 1: 91; group 2: 105

  • Mean duration of lesions (SD): group 1: 46.7 days (1.67 months); group 2: 45 days (1.58 months)

Interventions

Type of interventions:

  • Group 1: AL 20 mg/kg per day + low‐dose IMMA 30 mg/kg/d for 20 days

  • Group 2: IMMA 60 mg/kg/d for 20 days

Duration of intervention: 20 days

Duration of follow‐up: 51 days (1 month after the end of treatment)

Outcomes

Healing rates: percentage of participants 'cured' 51 days after treatment. On day 21 (end of therapy), the clinical response to treatment was coded subjectively as: apparent cure, partial response, and failure. After 1 month of follow‐up (day 51), the participants were described as showing definitive cure if all the lesions had healed and direct smears of the lesions were negative

Adverse effects

Tertiary outcomes: microbiological or histopathological cure of skin lesions

Time points reported: day 21, and 1 month after the end of treatment: day 51

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Seventy‐two patients were assigned randomly to two groups."

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Imbalance in numbers across intervention groups: 5/36 (13.89%), AL + IMMA group; 1/36 (2.78%), IMMAgroup.

We think missing outcome data likely to be related to true outcome.

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Momeni 2003

Methods

Study design: randomised controlled trial

Setting/location: endemic areas in the north of Isfahan city, Iran

Study period: not described

Sample size calculation: sample size calculation was based on 90% significance and 90% power to detect a difference between vehicle and ketoconazole. Assuming a 15% spontaneous cure with placebo, a minimum usefulness of a 60% cure with ketoconazole cream and about a 10% dropout, the sample size was calculated as 45 individuals in each group.

Participants

Type of Leishmania: Leishmania spp: L major and L tropica

Inclusion criteria: age > 5 years, laboratory‐confirmed diagnosis of CL, no previous treatment for leishmaniasis, no serious concomitant medical problems

Exclusion criteria: pregnant and nursing women, lesions on the face, lesions of > 4 months duration

N randomised: 90

Withdrawals: 17

N assessed: 73

Age: mean 19.9 years (range 6‐60)

Sex (male/female): 45/28

Baseline data:

  • Total number of lesions: 59 (group 1) and 43 (group 2)

  • Mean duration of lesions: 38 days (group 1) and 42 days (group 2)

Interventions

Type of interventions:

  • Group 1: ketoconazole cream

  • Group 2: vehicle

Duration of intervention: twice daily for 21 days.

Outcomes

Healing rates: clinical response to treatment was coded subjectively as: apparent cure, partial response or failure. Definitive cure was defined if at day 51, the lesions were healed and direct smears of the lesions were negative.

Adverse effects

Time points reported: the clinical response of the lesions was reported on days 21 (end of therapy) and 51

Notes

Study funding sources: supported by a research programme of Isfahan University of Medical Sciences

Possible conflicts of interest: not described

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The study was designed as a double‐blind, N randomised, placebo‐controlled trial"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Placebo and ketoconazole creams were identical cream containing either 25 ketoconazole 2% or placebo were used in the study. The two treatment groups received either the ketoconazole cream or placebo twice daily for 21 days as coded tubes with the same dosage"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes

Other bias

Low risk

Other items assessed correctly reported

Mostafavi 2013

Methods

Study design: randomised controlled trial

Setting/location: skin and CL clinic in Gaz in Isfahan, Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: size of ulcer less than 5 cm in diameter, ≤ 5 lesions, duration of disease < 3 months

Exclusion criteria: patients who had lesions on the face near the eyes or on the nose or ears,

pregnancy, lactation, lupoid or sporotrichoid lesions, use of immunosuppressor drugs in the past 6 months and severe adverse effects, exacerbation of the disease

N randomised: 40 participants (19 in prepared gel group and 21 vehicle gel mask)

Withdrawals: 1 participant in prepared gel group

N assessed: 39. 21 (100%) in vehicle gel group, and 18 in prepared gel group

Mean age: 28.2 years

Sex (male/female): 25/15

Baseline data: not described

Interventions

Type of interventions:

  • Group 1: MA gel mask. Prepared by using polyvinyl alcohol (PVA), sodium carboxymethylcellulose, and hydroxypropyl methylcellulose as the base polymer and glycerin as the plasticizer, applied twice a day

  • Group 2: vehicle gel mask, applied twice a day

Duration of intervention: 6 weeks

Co‐interventions: all of the participants received the standard treatment including ILMA weekly and cryotherapy every 2 weeks for a maximum of 6 weeks

Outcomes

Treatment outcome, defined as:

  • Complete healing: complete disappearance of the lesion

  • Moderate improvement: reduction in the size of the lesion more than 50%

  • Mild improvements: reduction in the size of the lesion less than 50%

  • No change: no considerable change in the size of the lesion

  • Worsening: increase in the size of lesion

  • Relapse: recurrence of lesion after complete healing

Time points reported: 2 months after the end of treatment for relapse

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The patients were randomly divided into two groups"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Open trials. Photography was done from all the lesions both at the first visit and all the follow‐up visits

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

High risk

Protocol not available; not registered; in clinical trial registry; adverse effects not reported

Other bias

High risk

Sample size calculation, reporting of Leishmania spp involved and baseline comparability was not correctly reported

Mujtaba 1999

Methods

Study design: randomised controlled trial

Setting/location: Pakistan

Study period: around 4‐5 months

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania spp not mentioned

Inclusion criteria: clinical diagnosis of CL and positive Giemsa‐stained smear of parasite

Exclusion criteria: > 5 lesions and pregnant women

N participants randomised (lesions): 104 (215). Group 1: n = 49 (111); group 2: n = 55 (104)

Withdrawals: 8

N assessed: 96. group 1: n = 49; group 2, n = 47

Age:

  • 0‐10 years: group 1, 11; group 2, 12

  • 11‐20 years: group 1, 19; group 2, 17

  • 21‐30 years: group 1, 10; group 2, 11

  • > 30 years: group 1, 9; group 2, 7

Sex (male/female): 58/38

Baseline imbalances: comparability regarding age, sex, number, and site of lesions and duration of the disease

  • Duration of the disease (months)

    • 0–2: group 1, 12; group 2, 11

    • 3‐4: group 1, 13; group 2, 12

    • 5–6: group 1, 11; group 2, 13

    • >6: group 1, 13; group 2, 11

  • No. of lesions in individual participants

    • 1: group 1, 21; group 2, 19

    • 2: group 1, 12 ; group 2, 11

    • 3: group 1, 11; group 2, 10

    • > 3: group 1, 5; group 2, 7

  • Total number of lesions at various anatomic sites in all participants

    • Upper limbs: group 1, 48; group 2, 45

    • Face: group 1, 33; group 2, 31

    • Lower limbs: group 1, 29; group 2, 28

    • Trunk: group 1, 1; group 2, 0

Interventions

Type of interventions:

  • Group 1: ILMA injections weekly until complete cure or up to 8 weeks

  • Group 2: ILMA injections fortnightly until complete cure or up to 8 weeks

Duration of intervention: until complete cure or up to 8 weeks

Outcomes

Healing: complete cure (100%); partial (> 50%; < 50%); failure (nil)

Speed of healing (time taken to be 'cured')

Duration of remission and percentage of people with treated lesions that recur within 6 months and 1, 2, and3 years:time assessment not specified

Prevention of scarring

Adverse effects

Time points reported: at 2, 4, 6 and 8 weeks of treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The patients were randomly divided into two treatment groups"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "Patients in group A were treated with weekly and those in group B with fortnightly intralesional injections"

Comment: we think the outcome is likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 8/104: 7.6% lost to follow‐up

Comment: the results for the defaulters were excluded from the statistical analysis. We think missing outcome data unlikely to be related to true outcome.

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes.

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Nassiri‐Kashani 2005

Methods

Study design: randomised controlled trial

Setting/location: Iran

Study period: October 2000 to February 2001 (5 months)

Sample size calculation: to detect a clinically relevant difference of at least 25% between placebo (35% estimated response rate) and itraconazole (60% estimated response rate) in the primary outcome parameter with a power of 90% and a 2‐sided type I error of 5%, a total of 164 subjects (82 in each group) were required. To compensate for dropouts, 20% more participants were enrolled in each group (in total 200 participants).

Participants

Type of Leishmania: L major

Inclusion criteria: aged 12‐60 years; presence of parasitologically confirmed lesion(s) of CL; healthiness on the basis of physical examination, medical history, and the results of blood biochemistry and haematology, carried out < 2 weeks before the start of the trial

Exclusion criteria: women of child‐bearing age without adequate effective contraception, pregnant or breastfeeding, duration of lesions > 45 days, presence of lesions on the face or near the mucous membranes, > 5 lesions, any lesion with a diameter > 3 cm, history of any systemic antileishmanial therapy, known hypersensitivity/allergy to itraconazole, receiving any drug with known interaction with itraconazole

N randomised: 200

Withdrawals: 42 in total. 17 in the Itraconozale group and 25 in the placebo group

N assessed: 158

Age: not described

Baseline imbalances: comparable with regard to age, sex, and duration of the lesions

  • Group 1: mean number of lesions: 2.5, mean size of lesions: 7.76 mm

  • Group 2: mean number of lesions: 2.2, mean size of lesions: 8.58 mm

  • Duration of < 45 days

Interventions

Type of interventions:

  • Group 1: itraconazole 200 mg once daily

  • Group 2: placebo

Duration of intervention: 8 weeks

Outcomes

Healing rates: for clinical assessment a 5‐point scoring system was employed: cure (complete re‐epithelialisation of all of the lesions), moderate improvement (more than 50% reduction in the size of the lesions), mild improvement (less than 50% reduction in the size of the lesions), no change, and worsening

Adverse effects: at each visit, all of the participants were asked about any adverse effects.

Time points reported: after completion of treatment (8 weeks), and at the end of the 3 months follow‐up (healing rates)

Notes

Study funding sources: supported by a grant from Janssen‐Cilag Pharmaceuticals Inc.

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The randomisation sequence was generated from a random number table"

Allocation concealment (selection bias)

Low risk

Quote: "Concealed from the investigator until the data entry was completed and the data bank was locked"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Patients were randomly allocated to receive either itraconazole in the form of two 100‐mg capsules or identical placebo capsules once daily for 8 weeks."

Comment: we think the outcome is unlikely to be influenced by lack of blinding

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Imbalance in numbers and reasons for missing data across intervention groups (17% itraconozale group; 25% placebo group).

Comment: the results for the defaulters were not excluded from the statistical analysis. We think missing outcome data likely to be related to true outcome.

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes.

Other bias

Low risk

Other items assessed correctly reported

Nilforoushzadeh 2004

Methods

Study design: randomised controlled trial

Setting/location: Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: not mentioned

Inclusion criteria: CL confirmed with direct smear, aged 1‐20 years, maximum number of lesions 3, maximum size of lesion 5 cm, duration of disease less than 8 weeks

Exclusion criteria: history of allergy to MA, pregnancy or lactating

N randomised: 210

Withdrawals: 53: 29/105 in group 1 and 24/105 in group 2

N assessed: 157. Group 1: 76, group 2: 81

Age: not described

Baseline data: not described

Interventions

Type of interventions:

  • Group 1: combination triple therapy consisting of: 15% PR + 10% urea applied twice daily for 4 weeks, cryotherapy with liquid nitrogen repeated every 2 weeks till complete healing or for a maximum of 3 sessions, ILMA twice every week till complete healing or for a maximum of 6 weeks (in sessions that both cryotherapy and injections were used, cryo was done before injections)

  • Group 2: ILMA twice every week till complete healing or for a maximum of 6 weeks

Duration of intervention: 6 weeks

Outcomes

Healing rates: percentage of participants 'cured' 2 weeks after treatment

Adverse effects

Time points reported: not described

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

High rates of dropouts: 53/210 (25.2%)

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes

Other bias

High risk

Sample size calculation, reporting of Leishmania spp involved and baseline comparability were not correctly reported

Nilforoushzadeh 2006

Methods

Study design: randomised controlled trial

Setting/location: Skin Disease and Leishmaniasis Research Center (SDLRC), Isfahan, Iran

Study period: around 5 months

Sample size calculation: not described

Participants

Type of Leishmania: not mentioned

Inclusion criteria: CL confirmed with direct smear

Exclusion criteria: pregnant or nursing mothers, treated previously for CL, any known intercurrent illness or a history of allergy to MA, palpebral lesions, having > 5 lesions, lesion > 3 cm, duration of lesions was > 12 weeks

N randomised: 80: 40 in each group

Withdrawals: 7

N assessed: 73: 38 in group 1 and 35 in group 2

Age: range 5‐75 years

Baseline data: the most common clinical type of the lesions in both groups was papule (50% in TCA and 44% in the ILMA group) and nodule (25% in TCA and 24% in the ILMA group).

Mean number of lesions: TCA: 1.2; ILMA: 1.4

Interventions

Type of interventions:

  • Group 1: TCA 50% (wt/vol) was applied on the lesions every 2 weeks up to 3 times

  • Group 2: ILMA weekly up to 6 weeks

Duration of intervention: up to 6 weeks

Outcomes

Healing: complete cure was defined as complete healing and re‐epithelialisation of the lesion(s); partial care as partial clinical improvement with reduction in erythema, induration and size of the lesions; treatment failure as the absence of any change or worsening of the lesion(s)

Duration of remission and percentage of people with treated lesions that recur after 3 months of follow‐up

Adverse effects

Microbiological or histopathological cure of skin lesions

Time points reported: after the last treatment session, 1 and 3 months post‐treatment. Complete cure was reported at 4 and 6 weeks of the treatment period. Recurrence was assessed at 3 months follow‐up

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random digit table

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "Unblindness is a limitation of this study"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "For 80 patients, which participated in the study, 73 cases completed the study"

Comment: the results for the dropouts were excluded from the statistical analysis.

We think missing outcome data likely to be related to true outcome.

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes.

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Nilforoushzadeh 2007

Methods

Study design: randomised controlled trial

Setting/location: Skin Disease and Leishmaniasis Research Center (SDLRC), Isfahan, Iran

Study period: 4 months

Sample size calculation: not described

Participants

Type of Leishmania: not mentioned

Inclusion criteria: CL confirmed with direct smear, no history of systemic or topical therapy for CL, absence of the malnutrition or severe predisposing disease such as cardiac, renal or hepatic disease and other contraindication for MA

Exclusion criteria: pregnant or lactating mothers, lesions > 3 months old and treated with the drugs that had interaction with MA

N randomised: 100, 50 in each group

Withdrawals: 23 (13 in the honey group and 10 in the ILMA group). However, in the abstract authors stated that 10 participants left the study.

N assessed: 90 (45 in each group)

Age: range 7‐70 years

Baseline data: the most common clinical type of the lesions in both groups was plaque (60% in honey + ILMA and 55.6% in the ILMA‐alone group). Mean number of lesions: honey + ILMA: 1.3; ILMA: 1.7

Interventions

Type of interventions:

  • Group 1: topical honey soaked gauze twice daily + ILMA

  • Group 2: ILMA

Duration of intervention: once weekly until complete healing of the ulcer or for maximum of 6 weeks

Outcomes

Healing: complete healing was defined as disappearance of the induration and complete re‐epithelialisation of the ulcer. Complete healing of the lesions was defined as complete clinical and parasitological healing (negative direct smear); partial healing of the lesions was defined as the decrease of the size and indurations of the lesions; non‐responsive was defined as no clinical change or progression of the lesions

Speed of healing (time taken to be 'cured'): expressed as mean healing time

Adverse effects

Time points reported: assessed weekly for 6 consecutive weeks and at the end of the 2nd, 3rd, and 4th month. Cure was assessed at the end of treatment and follow‐up

Notes

Study funding sources: not reported

Possible conflicts of interest: no competing interests

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Random allocation software (ver 1.0, May 2004; Saghaei)"

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

We think the outcome is likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Balance in numbers and reasons for missing data across intervention groups (13/45 (28.8%) in honey + ILMA group; 10/45 (22.2%) in ILMA‐alone group)

Comment: the results for the defaulters were excluded from the Kaplan‐Meyer analysis.

Selective reporting (reporting bias)

Low risk

Comment: relevant outcomes were reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Nilforoushzadeh 2008

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Skin Diseases and Leishmaniasis Research Center. Isfahan, Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: L tropica and L major

Inclusion criteria: all the participants had positive smear for Leishmania body and had not received any topical or systemic therapy for leishmaniasis, aged 7‐70 years, lesion not more than 3 months

Exclusion criteria: pregnant or lactating; history of cardiac, renal, hepatic diseases; any contraindication for the treatment

N randomised: 150. Group 1: 50; group 2: 50; group 3: 50

Withdrawals: 26. Group 1: 7; group 2: 14; group 3: 5

N assessed: 124 (82.66%). Group 1: 43 (86%); group 2: 36 (72%); group 3: 45 (90%)

Mean (SD) age: group 1: 33.1 years (17.5); group 2: 27.8 years (9.8); group 3: 29.1 years (14.7)

Sex (male/female): 88 (70.97%)/36 (29.03%)

  • Group 1: 32 (74.4%)/11 (25.6%)

  • Group 2: 23 (63.9%)/13 (36.1%)

  • Group 3: 33 (73.3%)/12 (26.66%)

Baseline data:

  • Type of lesion (no numbers): papule, nodule, plaque, ulcer, sporotrichid

  • Location of lesion (no numbers): face and neck, upper extremity, lower extremity, trunk

Interventions

Type of interventions:

  • Group 1: IMMA 60 mg/kg/d + placebo

  • Group 2: IMMA 30 mg/kg/d + 40 mg oral omeprazole for 3 weeks. Low dose of MA was prepared by adding normal saline to MA

  • Group 3: IMMA 30 mg/kg/d + oral placebo for 3 weeks

The oral placebo was identical in appearance to omeprazole capsules and was administered in the similar way

Duration of intervention: 3 weeks

Outcomes

Clinical cure: rate of complete response, 3 months (12 weeks) after starting treatment. Scale: complete healing of the lesions was regarded as complete clinical and parasitological healing (negative direct smear). Partial healing of the lesions was regarded as decrease of the size and indurations of the lesions, and no response was regarded as no clinical change or progression of the lesions.

Time to healing: proportion of complete improvement in the 3 groups during the course of treatment

Adverse effects: at the end of treatment

Time points reported: clinical cure: 12 weeks. Time to healing: 2, 4, 6, 8, 12 weeks. Adverse effects: at the end of treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: they declared no competing interests.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were selected and randomised by random allocation software into 3 groups.

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The oral placebo was completely similar to omeprazole capsules and was administered in the similar way; low dose of IMMA was prepared by addition of normal saline to IMMA

Both the investigating physicians and the participants were blinded to the type of treatment and drug codes were revealed only at the end of the study.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

High risk

Reason for missing outcome data likely to be related to true outcome, with either imbalance in numbers or reasons for missing data across intervention groups

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes.

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Nilforoushzadeh 2012

Methods

Study design: randomised controlled trial

Setting/location: Isfahan University of Medical Science, Isfahan, Iran

Study period: March 2008 to March 2009 (12 months)

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: unclear

Exclusion criteria: pregnant, children < 5 years, palpebral lesions (less than 20 mm from palpebral margin), > 5 lesions, > 12 weeks duration of leishmaniasis, history of any specific anti‐leishmaniasis therapy, significant underlying diseases

N randomised: 80 lesions

Withdrawals: 4 lesions

N assessed: 76 lesions (38 lesions in each group) and 60 patients (30 patients in each group)

Mean (SD) age: 5.11 years (13.3) (range 5‐50)

Sex: unclear

Baseline data:

  • Lesions according to sex:

    • Female: group 1, 16 (42.1%); group 2, 23 (60.5%)

    • Male: group 1, 22 (57.9%) and group 2, 15 (39.5%)

  • Mean size of lesions at the beginning of study was not different between groups (P = 0.58) and was 2.66 cm (SD 0.37) in group and 13.18 cm (SD 0.89) in group 2

  • Location of lesion:

    • 61/76 lesions in the upper extremities or head: 30/38 lesions (78.9%) in group 1 and 31/38 lesions (81.5%) in group 2

    • 15/76 lesions in the lower extremities: 8 lesions (21.05%) in group 1 and 7 lesions (18.4%) in group 2

Interventions

Type of interventions:

  • Group 1: after cleansing the lesion with alcohol, TCA 50% (Merck, Berlin, Germany) was applied onto the lesion using a cotton swab, until frosting the lesion, once a week and for up to 2 weeks. Afterwards, a controlled localised heating of the lesions was performed using an RF heat generator (4 MHz, maximum output 90 W; Ellman International Inc, NY, USA). The area was heated to 42°C surface temperature for 30 s once a week and for 4 consecutive weeks

  • Group 2: ILMA (Sanofi‐Aventis, France) administered twice a week and continued up to 8 weeks until absolute healing of lesions

Duration of intervention: non‐ablative radiofrequency + topical TCA 50%: 4 weeks; ILMA: 8 weeks

Outcomes

Treatment responses:

  • Complete cure: complete clinical healing was defined as complete re‐epithelialisation of the lesions, flattening of the lesions and lack of indurations along with negative direct smear

  • Partial cure: partial clinical improvement along with decreased size of indurations, erythematic areas, and lesions at the end of the treatment

  • Non‐cure or treatment failure: no clinical improvements along with unchanged or even increased size of lesions

Time points reported: 6 months of treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "This computer‐based randomised controlled trial was conducted from March 2008 to March 2009"

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The examinations and measurements were performed by the investigators who were blinded to the type of treatment."

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Originally 80 lesions were studied and randomly assigned to the 2 groups. However, due to missing data, only 76 were finally included in the analysis. No ITT analysis

Selective reporting (reporting bias)

High risk

Complete cure was described in Methods and Results, but partial cure and treatment failure was not reported in the Results section.

No adverse effects reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Nilforoushzadeh 2013

Methods

Study design: randomised controlled trial

Setting/location: Cutaneous Leishmaniasis and Skin Diseases Research Center, Isfahan, Iran

Study period: 2010 to 2011 (12 months)

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: age older than 5 years, diameter less than 3 cm, the duration of disease less than 12 weeks, lesions with less than 2 cm distance from eyelid, < 5 lesions

Exclusion criteria: pregnancy; lactation; colloid scar; lesions on cartilage, eyelids or joints; history of using other drugs against CL, treatment with immune system inhibitors

N randomised: 90 (30 people in each group)

Withdrawals: not described

N assessed: 90 (30 people in each group)

Mean (SD) age: 24.5years (14)

Sex (male/female):21/9, 23/7 and 21/9

Baseline data: Number of lesions:single lesion in 53 (58.9%), 2 lesions in 29 (32.2%) and 3 lesions in 8 patients (8.9%).

The type of lesion: plaque in 32 patients (35.6%), nodule in 44 (48.4%), plaque and nodule in 8 (8.9%), and papule in 6 patients (6.7%)

Distribution of lesions: hand 43, foot 15, face 8, trunk 5, hand and trunk 4, hand

and neck 4, neck 4, hand and foot 4, hand and face 2, ear 1

Interventions

Type of interventions:

  • Group 1: ILMA twice a week for a maximum of 8 subsequent weeks. The tip of the insulin syringe was entered from a point out side of the lesion at the periphery of lesion and the injection was continued until the whole area was blanched. This was repeated in all areas of the lesion until whole lesion was blanched.

  • Group 2: ILMA + TCA 50% solution. Simultaneously with the MA injection twice a week, the TCA solution 50% was applied with cotton applicator once a week for 8 consecutive weeks until the lesion was frosted. The participants were visited at the beginning of the treatment and after 3 and 6 months. Final evaluation was done in the 6th month.

  • Group 3: ILMA + fractional laser. In this group IL injection was done twice a week with fractional laser every 2 weeks. The laser used was Dosis M&M, Q ray FRX system with energy of 25, 1 pass, dots cycle 6 and pixel pitch of 1 mm. In each session, before treatment with laser, first lesion was cleaned with a mild cleanser and 70% then alcohol. The laser treatment was done by an experienced dermatologist every 2 weeks for 2 sessions after IL injection.

All patients in three groups were followed weekly for 3 months and then after 4 and 6 months to assess improvement of lesions and size of scar.

Outcomes

Clinical response, according to standard quartile grading:

  • 0 < 25% (mild improvement)

  • 25% < score 1 < 50% (moderate improvement)

  • 50% < score 2 < 75% (good improvement)

  • < 75% (very good improvement)

The quantitative 4‐point scale used included:

  • 0 = unchanged size

  • 1 = the scar size became a bit small

  • 2 = the scar size became very small

  • 3 = the scar is almost removed

Time points reported: 6 months

Notes

Study funding sources: Isfahan University of Medical Sciences

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Impossible for blinding

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote "Evaluation of patients were done by two physicians who were not aware of the treatment group. These two physicians did the evaluations separately. The reduction in this size of ulcer and the scar was compared with the same lesion"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not reported

Selective reporting (reporting bias)

Unclear risk

Protocol not available

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Nilforoushzadeh 2014a

Methods

Study design: prospective randomised controlled trial

Setting/location: Central Research Center of Skin Diseases and Leishmaniasis and Novin Laser Center; Isfahan, Iran

Study period: June 2010 to September 2011 (15 months)

Sample size calculation: sample size was calculated as 60 cases in each group with further rate of 10% dropouts and exclusions

Participants

Type of Leishmania: not described

Inclusion criteria: leishmaniasis scar diagnosed clinically on participants by a dermatologist

Exclusion criteria: pregnancy, lactation, colloid scar, treatment with immune system inhibitors,

use of isotretinoin or fillers for the past 6 months, use of dermabrasion or skin resurfacing for the past 12 months, skin types of IV to V1

N randomised: 120 (60 people in case group and 60 in control group)

Withdrawals: not described

N assessed: not described

Mean (SD) age: 27.21 years (11.52) (range 6‐45)

Sex (male/female): 51%/49%

Baseline imbalances: none

Severity of illness: mean (SD) scar size: laser CO₂ group: 2.3 cm (0.3) and fractional CO₂ laser: 3.4 cm (0.4)

Interventions

Type of interventions:

  • Group 1: ablative CO₂ laser after applying the topical anaesthetic on scar site with pulsed CO₂, duration: 10 ns, frequency: use of 20 kHz, and power: 25 Kw for one session. The participants were visited at the beginning of the treatment and after 3 and 6 months. Final evaluation was done in the 6th month.

  • Group 2: fractional laser in each session by energy: 25, one pulse, pass: 1, dot cycle: 6, with the system of Qray FRX, Dosis M and M

Outcomes

Clinical response, according to standard quartile grading:

  • 0 < 25% (mild improvement)

  • 25% < score 1 < 50% (moderate improvement)

  • 50% < score 2 < 75% (good improvement)

  • < 75% (very good improvement)

The quantitative 4‐point scale used included:

  • 0 = unchanged size

  • 1 = the scar size became a bit small

  • 2 = the scar size became very small

  • 3 = the scar is almost removed

Time points reported: 6 months

Notes

Study funding sources: Isfahan University of Medical Sciences

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quotes: "A photo was taken from patients before and after treatment with a digital camera ... Evaluation of participants were done by 2 physicians who were not aware of the treatment group. These 2 physicians did the evaluations separately. The reduction in this size of ulcer and the scar was compared with the same lesion"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not reported

Selective reporting (reporting bias)

Unclear risk

Protocol not available

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Nilforoushzadeh 2014b

Methods

Study design: randomised controlled trial

Setting/location: Skin Disease and Leishmaniasis Research Center, Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: participants with positive direct smear for CL; 6‐60 years of age; without any previous history of systemic or topical therapy for CL; lesions < 3 cm, < 12 week duration, and not located within 2 cm distance of palpebral margin

Exclusion criteria: pregnant and lactating women

N randomised: 200 participants

Withdrawals: not described

N assessed: 187; group 1: 91; group 2: 96

Mean (SD) age: 10.7 years (22.04); group 1: 10 years (20.6); group 2: 11.3 years (23.3)

Sex (male/female): 101/86

Baseline data: mean (SD) lesion area was 329 mm² (118.7) in group 1 and 359 mm² (55.6) in group 2

Interventions

Type of interventions:

  • Group 1: ILMA + 50% TCA was performed twice a week

  • Group 2: ILMA alone was performed twice a week

Duration of intervention: complete resolution of the lesions or end of 8 weeks

Outcomes

Clinical cure: complete re‐epithelialisation of the lesion and lack of induration. Clinical response was measured at the end of treatment and was defined as complete cure (negative direct smear and clinical healing), partial cure (partial clinical improvement with decreasing erythema, induration, and lesion size), non‐cure or treatment failure (no clinical change or worsening of the lesions)

Time points reported: 8 weeks

Notes

Study funding sources: the study is self‐funded

Possible conflicts of interest: there is no conflict of interest

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random allocation software (version 1.0, May 2004, Saghaei) was used for randomisation

Allocation concealment (selection bias)

Low risk

Participants with confirmed cutaneous leishmaniasis referred to the Skin Diseases and Leishmaniasis Research Center were selected and randomised by random allocation software into 3 groups

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The measurements were done before treatment and at the end of eighth week by the investigators who were blinded to the type of treatment."

Comment: Lesions were photographed before and after completion of the treatment course.

Not clear how blinding was preserved

Incomplete outcome data (attrition bias)
All outcomes

Low risk

From a total of 200 randomised participants, 96 participants in ILMA group and 91 participants in the combination group completed the study

Losses to follow‐up were below 25% and homogeneous between the groups

Selective reporting (reporting bias)

High risk

Protocol not available, not registered. Adverse effects not reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Ranawaka 2010

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Anuradhapura Teaching Hospital, Anuradhapura, Sri Lanka

Study period: 32 months. Participant selection: April 2006 to June 2007 (15 months)

Sample size calculation: not described

Participants

Type of Leishmania: L donovani

Inclusion criteria: participants with CL who attended to Anuradhapura Teaching Hospital, Anuradhapura, Sri Lanka from April 2006 to June 2007

Exclusion criteria: not described

N randomised: a total of 154 participants with 229 lesions were included in the study. IL 7% HSCS: 67; ILSSG: 87

Withdrawals: (4.5%: 7/154). HSCS: 3, ILSSG: 4

N assessed: 147 (95.45%) CL participants with 222 lesions completed treatments. HSCS: 64 (95.52%); ILSSG: 83 (95.40%).

Age: mean 32 years (range 16 months to 74 years)

Sex (male/female): 99/55, M:F ratio of 1.8:1

Baseline data:

  • N lesions: HSCS: 93, ILSSG: 136

  • Location of the target lesion: head and neck 41, upper limb 123, lower limb 43, trunk 14, buttock 1. 93.2% of lesions being on exposed parts of the body

  • Clinical presentation (n(%)): papules ≤ 1 cm diameter: 52 (23.6%); nodules > 1 cm diameter: 28 (12.7%); plaques: 14 (6.34%); central ulcer with erythematous induration around the lesion: 121 (55%); chronic non‐healing ulcers with undermined edges without surrounding induration; 5 (2.2%)

  • 19 lesions were wet lesions and all others (91.4%) were dry lesions

Interventions

Type of interventions:

  • Group 1: HSCS was prepared by dissolving 12.2 g of sodium chloride in 200 mL of 0.9% sodium chloride (normal saline) solutions. The solution was injected intralesionally and not subcutaneously

  • Group 2: ILSSG (Pentostam)

Duration of intervention: HSCS: 1‐29 weeks, depending on the size and the duration of the lesions (mean 8.78 weeks). SSG: 1‐13 weeks, depending on the size and the duration of the lesions (mean 5.11 weeks)

Participants were seen weekly for the first 3 injections; fortnightly for the fourth and fifth injections; then monthly until cure.

Outcomes

Clinical cure: the percentage of lesions that were cured with SSG and HSCS

Number of injections required per each lesion in total for cure (scales: 1‐10 injections)

Adverse effects: systemic side effects with SSG or HSCS. Local side effects: pain during injection. Scarring after healing, postinflammatory hyperpigmentation

Time points reported: at the end of treatment. Participants were followed‐up every 3 months after cure for 18 months to assess recurrences and evidence of visceralisation

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "double blind"

Insufficient information to permit judgment

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double blind", "At each visit the patients were examined by the primary investigator who was blind to the therapy"

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

4.5% (7/154) did not complete the study

Selective reporting (reporting bias)

Unclear risk

No protocol was available; not registered. Insufficient information to permit judgment

SD not provided for duration of treatment and number of injections

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Ranawaka 2015

Methods

Study design: randomised double‐blind clinical trial

Setting/location: Skin Clinic of Anuradhapura Teaching Hospital, Sri Lanka

Study period: January 2012 to February 2013 (14 months)

Sample size calculation: approximately 473 participants per group were required to enable the detection of a treatment difference of 5% with a lowest cure rate of one group of 90%

Participants

Type of Leishmania: L donovani

Inclusion criteria: the study recruited participants with suspected CL who consented to participation.

Exclusion criteria: pregnant or lactating women, subjects with a history of cardiac, renal, or hepatic disease

N participants randomised (lesions): 444 (643). 10% HSCS: 192 (297); 15% HSCS: 82 (101); ILSSG: 170 (245)

Withdrawals (lesions): 17 (20) lost to follow‐up. 10% HSCS: 8 (7); 15% HSCS: 3 (3); ILSSG: 6 (10)

N assessed (lesions): 427 (623); 10% HSCS: 79 (98); 15% HSCS: 184 (290); ILSSG: 164 (235)

Age: mean 32.7 years (range 14 months to 88 years). Most (60.8%) were aged 16–45 years

Sex (male/female): 286/158

Baseline data: the average delay in presentation was 8 months (range: 4 weeks to 5 years). 91% of lesions were located on exposed areas of the body. Most (69.8%) participants had a single lesion.

Interventions

Type of interventions:

  • Group 1: 10% HSCS, prepared by dissolving 18.2 g of medical sodium chloride in 200 mL of 0.9% sodium chloride (normal saline) solutions

  • Group 2: 15% HSCS, prepared by dissolving 28.2 g of medical sodium chloride in 200 mL of 0.9% sodium chloride (normal saline) solutions

  • Group 3: ILSSG

Duration of intervention: participants were seen weekly for the first 3 injections, fortnightly for the 4th and 5th injections, and then monthly until a cure was achieved.

Outcomes

Clinical and parasitological cure: responses were graded as slight (10% improvement from the initial status of the lesion), mild (20%–30% improvement), moderate (50% improvement), marked (80%–90% improvement), or as total if the lesion had cleared and parasites were not detected in the affected area by smear or culture. Both marked improvements and total clearance were considered to represent a cure. Reported at the end of treatment.

Duration of treatment: in weeks, mean (range)

Time required to achieve the cure of the lesion: mean and median times taken for cure for the different treatments groups. Every 5 weeks until achieve 30 weeks.

Injections per lesion, mean (range): number of injections for cure per lesion

Relapse: L recidivans

Adverse effects: local side effects: at the site of the lesion caused by the injections: pain during injections, postinflammatory hyperpigmentation, scarring, postinflammatory depigmentation, ulceration and necrosis, L recidivans. Systemic side effects: no systemic or significant local side effects with either SSG or 10% HSCS.

Time points reported: participants were followed up every 3 months after cure for more than 6 months to assess recurrences and evidence of visceralisation

Notes

Study funding sources: none reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The treatment options were documented separately and packed in opaque envelopes numbered consecutively according to the randomisation schedule at a ratio of 2: 2: 1

Allocation concealment (selection bias)

Low risk

The allocation sequence was concealed from the researcher who enrolled and assessed the participants by the use of sequentially numbered, opaque, sealed, and stapled envelopes that were impermeable even to intense light.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The participants and the investigator (who assessed the participants for their clinical response) were blind to the type of therapy allocated.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

At each visit, the participant was examined by the primary investigator, who was blinded to the therapy.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

The numbers of withdrawals were similar and small in the 3 groups: 4.1% (8/192) in group 1, 3.6% (3/82) in group 2, and 3.5% (6/170) in group 3.

Selective reporting (reporting bias)

Low risk

The study published reports our primary outcome and adverse effects.

Other bias

Low risk

Other items assessed correctly reported

Reithinger 2005

Methods

Study design: randomised controlled trial

Setting/location: Khair Khana Clinic; Kabul, Afghanistan

Study period: 100 days

Sample size calculation: to detect a 20% difference in the cure rate between the SSG and thermotherapy groups, assuming an 80% cure rate in the SSG groups, with a 90% power and a 5% 2‐sided type I error, 98 subjects were needed in each group. To compensate for anticipated loss to follow‐up, 40% more participants were enrolled in each group.

Participants

Type of Leishmania: L tropica

Inclusion criteria: age > 5 years; the presence of a single, parasitologically confirmed CL lesion;

no prior history of disease and/or antimonial treatment

Exclusion criteria: the presence of a CL lesion located on or immediately adjacent to the nose, lips, or eyes; pregnancy; breastfeeding; major surgery in the previous 3 months; presence of any uncontrolled medical condition; anticipated unavailability for follow‐up

N randomised: 431. Group 1: 148, group 2: 144, group 3: 139

Withdrawals: 172 in total: 30 decided to withdraw after allocation and 142 did not complete the 4 visits or the follow‐up

N assessed: 259. Group 1: 93, group 2: 58, group 3: 108

Age: range 10‐20 years

Sex (male/female): 200/201

Baseline data: the lesions were primarily located on the face (43.4% of participants), on the hands (38.2%), legs (15.9%) and arms (2.4%)

  • Group 1: MNL: 1. MSL: 12.75 mm. MDLBT: 6 months

  • Group 2: MNL: 1. MSL: 13.75 mm. MDLBT: 5.5 months

  • Group 3: MNL: 1. MSL: 10.25 mm. MDLBT: 6 months

Interventions

Type of interventions:

  • Group 1: ILSSG, 5 injections of 2‐5 mL every 5‐7 days depending on the lesion size for up to 29 days

  • Group 2: IMSSG (20 mg/kg) daily for 21 days

  • Group 3: thermotherapy using radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s depending on lesion size)

Duration of intervention: not described

Outcomes

Percentage of participants 'cured' at 100 days after treatment initiation and by time to cure: cure was defined as the complete re‐epithelialisation of the CL lesion, with no evidence of papules, inflammation, or induration

Speed of healing (time taken to be 'cured')

Adverse effects: the occurrence of adverse effects was evaluated blindly by means of participant interviews and physical examinations during follow‐up visits

Time points reported: trial endpoint was 100 days after start of therapy

Notes

Study funding sources: the HNI Malaria and Leishmaniasis Control Program is funded by the European Union. This study was supported by the United Nation Children's Fund/United Nation Development Program/WorldBank/World Health Organization Special Program for Research and Training in Tropical Diseases, the Afghan Research Evaluation Unit, and the Leveen Family Fund.

Possible conflicts of interest: since March 2004, R Reithinger has been a part‐time employee of Thermosurgery Technologies. All other authors: no conflicts

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients then proceeded to pick 1 of 3 identical cardboard pieces out of a hat (the cardboard had been labelled with different treatment codes on one of its sides, the codes being non‐visible to the patient). After patients were randomly assigned to receive a treatment, the cardboard piece picked was returned to the hat."

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: no blinding of participants and personnel seemed to be performed.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The occurrence of adverse effects was evaluated blindly by means of patient interviews and physical examinations during follow‐up visits", "Microscopic examination was performed blindly."

Comment: microscopic examination and adverse effects were performed blindly but no description about clinical examination was given.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

172/431 (39.9%) withdrew from the study. However, ITT analyses were performed.

Quote: "An intention‐to‐treat analysis of the data (... including the patients lost to follow‐up, who were considered to have had treatment failure) yielded similar results for the comparison of the odds of cure for the different treatments"

Selective reporting (reporting bias)

Low risk

Comment: relevant outcomes were reported

Other bias

Low risk

Other items assessed correctly reported

Sadeghian 2006a

Methods

Study design: randomised controlled trial

Setting/location: Iran

Study period: 3 months

Sample size calculation: not described

Participants

Type of Leishmania: L major

Inclusion criteria: CL in whom the diagnosis was confirmed by laboratory demonstration of the parasite in the lesions by direct smear

Exclusion criteria: allergic to antimonial drugs or pentoxifylline, lactating or pregnant, history of systemic illness

N randomised: 64, 32 in each group

Withdrawals: 1 participant withdrew from the MA + placebo group

N participants assessed (lesions): 63. Group 1: 32 (143), group 2: 31 (164)

Mean age: group 1: 27 years, group 2: 31 years

Sex (male/female): 29/34

Baseline data: duration of the disease approximately 1.3 months.

  • Group 1: 59.5% had plaque, 24.5% nodule and 16% papule type of lesions, and they were primarily located in the face (35%), upper limbs (24.5%), lower limbs (21%) and trunk (19.5%). MNL: 4, MDLBT: 1.2 months

  • Group 2: 42.7% had plaque, 33.6% nodule and 23.7% papule type of lesions, and they were primarily located in the face (33.5%), upper limbs (24.3%), lower limbs (22%) and trunk (20.2%). MNL: 5, MDLBT: 1.3 months

Interventions

Type of interventions:

  • Group 1: IMMA (20 mg pentavalent antimony/kg/d) + pentoxifylline (400 mg 3 times daily)

  • Group 2: IMMA (20 mg pentavalent antimony/kg/d) + placebo (3 tablets daily)

Duration of intervention: 20 days

Duration of follow‐up: 3 months

Outcomes

Healing rates:

  • Complete improvement defined as flattened lesions, no induration and appearance of epidermal creases

  • Partial improvement defined as reduction in the size of the lesions but without the appearance of epidermal creases

  • Poor response defined as no reduction in the size of the lesions

Adverse effects

Time points reported: after end of treatment and 3 months' follow‐up

Notes

Study funding sources: not described

Possible conflicts of interest: not described

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The patients were randomly divided into two groups"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Study was described as "double blind" but has no description about allocation method.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Study was described as "double blind" but has no description about the blinding method.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No blinding of outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

One patient withdrew in the placebo group. Reasons for withdrawal were reported. However, no ITT analyses were performed.

Selective reporting (reporting bias)

Low risk

Comment: relevant outcomes were reported

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Sadeghian 2006b

Methods

Study design: randomised controlled trial

Setting/location: Skin Diseases and Leishmaniasis Research Center in Isfahan, Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania:L major endemic in the area.

Inclusion criteria: participant with positive smear for Leishman bodies, of both sexes and > 5 years old who did not have indication for systemic therapy

Exclusion criteria: facial lesions or lesions on joints, sporotrichoid type, lupoid leishmaniasis, erysipeloid type and other atypical forms of CL, pregnant women, history of cardiovascular and renal diseases

N randomised: 72, 36 in each group

Withdrawals: 0

N assessed: 72

Mean age: HSCS, 20.52 years; ILMA group, 18.67 years

Baseline data: the most common site of lesions was on the extremities and the least common site was the trunk in both groups. The size range of lesions were from 0.5‐4 cm² (IL HSCL group) and from 0.5‐4 cm² (ILMA group)

Interventions

Type of interventions:

  • Group 1: IL HSCL (NaCl 5%; 0.5 to 1 mL)

  • Group 2: ILMA (0.5‐1 mL)

Duration of intervention: weekly 6‐10 weeks

Duration of follow‐up: 6 months

Outcomes

Healing: complete improvement is defined as completely clinical re‐epithelialisation with no signs of induration and inflammation with negative smear for Leishman bodies at the end of the treatment period. Partial improvement is defined as the re‐epithelialising lesion has become smaller but not cured. If the lesion has become larger or has not been differed it will be called as no response to treatment.

Time points reported: at 6 and 10 weeks of treatment and 6 months follow‐up

Notes

Study funding sources: not described

Possible conflicts of interest: not described

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "This N randomised controlled clinical trail study with simple sampling." "72 patients randomly divided in two equal groups".

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Low risk

Comment: relevant outcomes were reported

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Sadeghian 2007

Methods

Study design: randomised controlled trial

Setting/location: Skin Diseases and Leishmaniasis Research Center, Iran

Study period: 7 months

Sample size calculation: not reported

Participants

Type of Leishmania: not reported

Inclusion criteria: smear from a suspected CL lesion was confirmed positive for Leishmania

Exclusion criteria: pregnant women, children < 5 years of age, patients with facial lesions, those who had already received or were under other specific antileishmanial therapy, significant underlying diseases

N randomised: 117

Withdrawals: 0

N participants assessed (lesions): 117: 57 (83) lesions in group 1 and 60 (94) in group 2

Mean age: group 1, 25.12 years; group 2, 22.6 years

Sex (male/female): group 1, 37/20; group 2, 29/31

Baseline data: the shape of lesions was papule, nodule and plaque‐like. Lesions were located in the trunk and upper and lower limbs.

  • Group 1: mean number of lesions: 1.4. Mean duration of lesions: 4.42 weeks. Mean largest diameter: 15.6 mm

  • Group 2: mean number of lesions: 1.5. Mean duration of lesions: 3.85 weeks. Mean largest diameter: 14.7 mm

Interventions

Type of interventions:

  • Group 1: controlled localised heating using an RF heat generator (4 MHz, maximum output 90 W). The affected area was heated to 50ºC surface temperature for 30 s

  • Group 2: ILMA. The volume of the drug was 0.1‐4 mL (each mL contains 85 mg MA), depending on lesion size

Duration of intervention: once weekly for 4 consecutive weeks

Duration of follow‐up: 6 months

Outcomes

Percentage of participants 'cured':

  • Complete (lesions flattened, no induration, and epidermal creases appeared)

  • Partial (size of the lesion decreased but without the appearance of epidermal creases)

  • Poor (size of induration was unchanged or had increased)

Duration of remission and percentage of people with treated lesions that recur within 6 months

Prevention of scarring

Adverse effects

Time points reported: at the end of treatment, and 6 months after

Notes

Study funding sources: technical and financial support from the joint WHO Eastern Mediterranean Region (EMRO), Division of Communicable Diseases (DCD) and the WHO Special Program for Research and Training in Tropical Diseases (TDR): the EMRO/TDR Small Grant Scheme for Operational Research in Tropical and other Communicable Disease.

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were randomly allocated to one of two treatment groups using a random number list generated by Epi Info (a software package designed to provide easy form and database construction, data entry, and analysis with epidemiologist statistics, map sand graphs)"

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Group 1 was treated with controlled localised heating using an RF heat generator. Group 2 was treated with ILMA

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Follow‐up evaluation was made by clinical assessment of treated lesions by a second dermatologist who was blinded to the method of treatment."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No missing outcome data

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes.

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Safi 2012

Methods

Study design: randomised controlled trial

Setting/location: Leishmaniasis clinic of the National Malaria and Leishmaniasis Control Program in Darul‐Aman District, Kabul City, Afghanistan

Study period: not described

Sample size calculation: to detect an 8% difference in the cure rate between the current standard therapy (ILMA) and thermotherapy by radio frequency groups, assuming 90% cure rate in the conventional therapy with an 80% power and a 5% type I error, 174 subjects were needed in each group. To compensate for anticipated loss to follow‐up, sample size inflated by 10%. Hence, the final sample size, figuring research design, rounded to nearest 10 turned out to be 390 subjects, with 195 assigned to each treatment group.

Participants

Type of Leishmania: L tropica

Inclusion criteria: aged > 5 years; presence of a single, parasitologically confirmed CL lesion

Exclusion criteria: history of previous infections from anthroponotic CL that was treated with antimonial medications, lesions located within 2 cm of the eyelids or on the lips or nose

N randomised: 390 (195 assigned to each treatment group)

Withdrawals: 8 participants (6 in group 1; 2 in group 2).

N assessed: 382 (189 in group 1; 193 in group 2).

Median (interval) age: group 1, 14 years (5‐70); group 2, 13 years (5‐75)

Sex (male/female): group 1, 85/104; group 2, 92/101

Baseline data:

  • Mean (SD) systolic blood pressure: group 1, 116.2 mmHg (13.5); group 2, 113 mmHg (13.6)

  • Mean (SD) diastolic blood pressure: group 1, 72.4 mmHg (7.9); group 2, 72.2 mmHg(10.7)

  • Mean (SD) body weight: group 1, 42.5 kg (23.3); group 2, 39.01 kg (18.3)

  • Location of lesion (n (%))

    • Group 1: face/neck: 161 (50.8), upper extremity 26 (48.1), lower extremity 2 (25)

    • Group 2: face/neck: 156 (49.2), upper extremity 28 (51.9), lower extremity 6 (75), trunk 3 (100)

  • Type of lesion (n (%))

    • Group 1: nodule 69 (41.1), papule 120 (57.7)

    • Group 2: ulcer 6 (100), nodule 99 (58.9), and papule 88 (42.3)

  • Size of the lesion: group 1: median 1 cm² and size interval 1‐4; group 2, median 2 cm² and size interval 1 ‐4.

Interventions

Type of interventions:

  • Group 1: single application of thermotherapy by radio frequency producing localised heat of 50°C (122°F) for 30 s

  • Group 2: 5 injections of ILMA (2‐7 mL depending on the size of the lesion). The injections were given in 7‐day intervals for 5 weeks.

Duration of intervention: 5 weeks in ILMA group

Outcomes

Cure was defined as complete re‐epithelialisation of the lesion with no inflammation and resolution of the papule and/or nodule. Failure was defined as no improvement, with the lesion unchanged or worse compared with its status at the start of treatment. Lesions that showed some improvement but did not meet the criteria for cure were considered treatment failures

Adverse effects

Time points reported: 6 months after treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were asked to pick one of two identical cardboard pieces out of a box (the cardboard had been labelled with different treatment codes on one of its sides) for their group assignment. After patients were assigned to a treatment group, the cardboard piece picked was returned to the box."

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were losses to follow‐up and no ITT analyses were performed. Also, the distribution of the lesions and the size was uneven between the 2 arms.

Quote: "More papules (57.7%) were treated by thermotherapy compared to (42.3%) those which were treated by Glucantime. Nodules were mostly treated by Glucantime (58.9%), whereas all ulcers were treated by Glucantime. The lesion size interval was [1–4 cm²] with median of 1 cm² for thermotherapy by radio frequency and 2 cm² for Glucantime."

This is important because the authors found out the following significant associations:

Quote: "Type and size of lesion, were significantly associated with the outcome variable (cure/failure). Type of lesion was significantly associated with treatment outcome in this sample, taking papules as reference (Table V). Those who had nodules were 41% less likely and those with ulcers were 80% less likely to be cured compared with those with papules"

Selective reporting (reporting bias)

High risk

Adverse effects were described in Methods but not in Results. They mention them vaguely in the Discussion section.

Other bias

Low risk

Other items assessed correctly reported

Salmanpour 2001

Methods

Study design: randomised controlled trial

Setting/location: Iran

Study period: 18 months

Sample size calculation: not described

Participants

Type of Leishmania:Leishmania spp: L tropica and L major

Inclusion criteria: patient with CL confirmed parasitologically by direct skin smears or skin biopsies.

Exclusion criteria: children < 3 years of age, pregnant and lactating women, cases with concomitant renal, liver or heart disease, and any event of any laboratory abnormality prior to initiation of treatment

N randomised: 96

Withdrawals: 0

N assessed: 96 (100%): 64 in the ketoconazole group and 32 in the ILMA group

Age: range 3‐64 years

Sex (male/female): 44/52

Baseline data:

In the ketoconazole group:

  • Mean duration of lesions (SD): 2.6 months (1.4)

  • Mean number of lesions: 2.5

In the MA group:

  • Mean duration of lesions (SD): 3.1 months (1.4)

  • Mean number of lesions: 2.3

Interventions

Type of interventions:

  • Group 1: ketoconazole orally (adults: 600 mg/d for 30 days and children: 10 mg/kg/d for 30 days)

  • Group 2: ILMA 6 injections (some received up to 8)

Duration of intervention: 30 days in the ketoconazole group. There is no mention about frequency of injections in MA group.

Outcomes

Healing rates:

  • Cured if lesions had completely re‐epithelialised with little or no scarring at 6 weeks post‐treatment.

  • Failure if participants showed one of the following findings: < 25% reduction in the diameter of induration after 4 weeks of treatment; the persistence of lesions 6 weeks post‐treatment

  • Relapse of disease after initial healing at 6 weeks post‐treatment

Adverse effects

Time points reported: participants were followed every 2 weeks for the duration of the treatment and every 6 weeks after cessation of treatment up to 24 weeks

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomly allocated to two treatment groups using a simple randomisation method"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Group 1, which included 64 participants, was designated as the group treated with oral ketoconazole. Group 2, which included 32 participants, was designated as the group that received ILMA. The outcome is likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

High risk

Imbalance in numbers and reasons for missing data across intervention groups: 7/57, ketoconazole; 9/23, ILMA

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgment

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Salmanpour 2006

Methods

Study design: randomised controlled trial

Setting/location: Dermatology Clinic of Faghihi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran

Study period: unclear

Sample size calculation: not reported

Participants

Type of Leishmania: not reported

Inclusion criteria: participant with CL confirmed

Exclusion criteria: not described

N randomised: 60

Withdrawals: 0

N assessed: 60 participants distributed into the 3 groups of equal number (n = 20)

Age: not reported

Sex: not reported

Baseline data: lesions were located on the head, neck and lower and upper extremities. Lesion duration was divided in 2 groups: a duration of < 3 months and > 3 months. Regarding lesion size, the authors divided the sizes in 3 groups: < 1 cm, 1‐3 cm and > 3 cm.

Interventions

Type of interventions:

  • Group 1: cryotherapy. The freezing time was 10‐30 s with a thawing interval of 20 s

  • Group 2: cryotherapy + ILMA. The participants first received cryotherapy and after 5‐10 min were given ILMA

  • Group 3: ILMA. The solution was injected into each lesion (0.2 to 1.5 cm³ per session per week, depending on the size)

Duration of intervention: weekly for a total of 6‐8 times for each case

Outcomes

Healing rates: percentage of participants 'cured': timing not reported

Adverse effects

Time points reported: not reported

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Insufficient information to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The outcome is likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No blinding of outcome assessment, and the outcome measurement is likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

High risk

No explanation was provided for missing outcome data.

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports included all expected outcomes, including those that were pre‐specified.

Other bias

High risk

Sample size calculation, reporting of Leishmania spp involved and baseline comparability were not correctly reported

Shamsi Meymandi 2011

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: dermatology clinic and leishmaniasis research centre of Afzalipour hospital of Kerman, province of Iran

Study period: 21 months

Sample size calculation: not described

Participants

Type of Leishmania: L tropica

Inclusion criteria: aged 7–60 years, confirmed CL

Exclusion criteria:chronic systemic disease (such as renal failure, myocarditis, hepatitis and pancreatitis), immune suppression, breastfeeding, pregnancy, sporotrichoid and lupoid forms, maximum diameter of lesions > 3 cm, disease duration more than 1 year, more than 2 lesions present, history of sensitisation to MA,facial and joint lesions, receiving other specific anti Leishmania therapies

N randomised: 191. Group 1: 96, group 2: 95

Withdrawals: group 1: 16 (14: no follow‐up, 2 enrolled in other treatment), group 2: 15 (8 no follow‐up, 4 enrolled in other treatment, 3 did not adhere to treatment schedule)

N assessed: 160 (83.77%). Group 1: participants: 80 (83.33%), group 2: participants: 80 (84.21%)

Age: range 7‐60 years

Sex (male/female): Randomised patients: group 1: 50(52%)/46 (48%), group 2 39 (41%)/56 (59%). Assessed patients: group 1: 42 (50%)/38 (46%); group 2: 31 (39%)/49 (61%)

Baseline data (N lesions): group 1: 80, GB: 80

Locationof the target lesion:

  • Group 1: head and neck 1 (1.3%), upper limb 68 (85%), lower limb 7 (8.8%), trunk 4 (5%)

  • Group 2: head and neck 5 (6.3%), upper limb 64 (80%), lower limb 10 (12.5%), trunk 1 (1.3%)

Type of lesions:

  • Group 1: plaque or ulcerated plaque 33 (40.5%), nodule or ulcerated nodule 47 (59.5)

  • Group 2: plaque or ulcerated plaque 25 (31.3%), nodule or ulcerated nodule 55 (68.7%)

Case type:

New, group 1: 67 (83.3%); group 2: 69 (86.3%)

Recurrence, group 1: 0 (0%); group 2: 1 (1.3%)

Failure of previous treatment, group 1: 8 (10%); group 2: 0 (0%)

Missed previous treatment, group 1: 5 (6.3%); group 2: 10 (12.5%)

Interventions

Type of interventions:

  • Group 1: CO₂ laser. Lesions were locally anaesthetised by injection of 2% lidocaine. The CO₂ laser (6–8 W continuous wave) was applied to the lesions and an area 2–3 mm² around it. The procedure was repeated until the ulcer bed turned brown (maximum 3–5 times)

  • Group 2: participants were treated with combined cryotherapy (biweekly) and ILMA (weekly). Cryotherapy with liquid nitrogen was performed using dipstick technique. Then ILMA (Amp 1.5 gr in 5 Ml solution) was injected in lesions

Duration of intervention: 16 weeks

Outcomes

Clinical cure of the lesions: defined as complete re‐epithelialisation of 100% (± scar), complete flattening of induration

Laboratory cure of the lesions: negative smear of the lesions compared with baseline

Cure rate based on weeks of follow‐up

Adverse effects of 2 types of treatments

Time points reported: weeks 2, 6, 12 and 16

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The randomisation sequence was generated by the use of a randomisation Table. A simple block randomisation list with block size of 4 was prepared by a member of the study.

Allocation concealment (selection bias)

Unclear risk

Quote: "The randomisation allocation concealment was performed by sending the randomisation number in envelopes to a member who was responsible for giving the assigned treatment after each eligible patient was enrolled. The recruited patients were referred to this member to receive their assigned treatments."

Comment: unclear because no mention of opaque and numbered envelopes

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were 31 withdrawals (16/96 (16.7%) in the ILMA + cryotherapy and 15/95 (15.8%) in the CO₂ laser group)

Comment: no ITT analyses were performed. However, withdrawals accounted for < 20% and were homogeneous among the treatment groups.

Selective reporting (reporting bias)

Unclear risk

Protocol not available. Not registered. Some pre‐specified outcomes were reported. Data on individuals not presented. Conflicting data is presented between the abstract and the tables

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Shanehsaz 2015

Methods

Study design: randomised double‐blind placebo‐controlled clinical trial

Setting/location: Aleppo University Hospital Clinic, Aleppo, Syria

Study period: 12 months (from July 2009 to July 2010)

Sample size calculation: not described

Participants

Type of Leishmania: not described

Inclusion criteria: parasitological confirmation; aged 5–65 years; normal values of liver, kidney, and pancreas function tests and electrocardiogram before treatment

Exclusion criteria: pregnant and lactating women; history of cardiac, renal, and hepatic diseases; treatment with other drugs during the month before commencement of the study

N randomised: 90, 30 in each group

Withdrawals: not described, but it seems that there were no dropouts

N assessed: 90, 30 in each group

Sex (male/female): group 1, 21 (70%)/9 (30%); group 2, 17 (56.7%)/13 (43.4%); group 3, 20 (66.7%)/10 (33.3%)

Mean (SD) age: group 1, 26.3 years (11.2); group 2, 24.2 years (12.5); group 3, 25.1 years (12.4)

Baseline data:

Mean number of lesions (SD): group 1, 3.0 (1.92); group 2, 3.0 (1.92); group 3, 3.0 (1.92)

Interventions

Type of interventions:

  • Group 1: IMMA 60 mg/kg/d (Glucantime; Aventis, Paris Cedex, France) + oral placebo. The drug was available in 5 mL vials.

  • Group 2: IMMA 30 mg/kg/d + oral cimetidine 1200 mg/d

  • Group 3: IMMA 30 mg/kg/d and oral placebo

Duration of intervention: 3 weeks for 3 groups

Outcomes

Clinical and parasitological cure: the effectiveness of the treatment was classified in 3 levels: complete response of the lesions was regarded as complete clinical and parasitological healing (negative direct smear). Partial response of the lesions was regarded as decrease of the size and indurations of the lesions, and no response was regarded as no clinical change or progression of the lesions

Time to healing: proportion of complete response in 3 groups during the course of treatment

Adverse effects: complications

Time points reported: clinical and parasitological cure: 12 weeks. Adverse effects: 6, 8, 12 weeks

Notes

Study funding sources: this project was funded by the University of Aleppo, Aleppo, Syria

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were recruited and randomised by random allocation software into 3 treatment groups of 30 subjects each.

Allocation concealment (selection bias)

Unclear risk

No information provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

No blinding, but the review authors judge that the outcome is not likely to be influenced by lack of blinding; the oral placebo was completely identical in appearance to the cimetidine tablet, they were the same colour, and drug codes were revealed only at the end of the study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information provided

Selective reporting (reporting bias)

High risk

The study report fails to include results for a key outcome that would be expected to have been reported for such a study. The adverse effects are not properly described, although questionnaires regarding response to treatment and drug side effects were completed and in Table 2 there are data, numbers (%).

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Sharquie 1997

Methods

Study design: randomised controlled trial

Setting/location: Baghdad, Iraq

Study period: October 1994 to November 1995 (8 months)

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania spp: L major and L tropica endemic in the area

Inclusion criteria: confirmed cases of CL by smear or culture, or both; acute CL with a history of 12 weeks or less

Exclusion criteria: cases of reinfection

N randomised: 85

Withdrawals: 22 participants (participants who did not show up after the first or second injection were excluded). Losses were not reported by group

N assessed: 63. Group 1, 19; group 2, 17; group 3, 18; group 4, 9

Age: range 3 months to 65 years

Sex (male/female): 28/37

Baseline data:

  • Group 1: MNL: 2.0. MDLBT: 6.89 weeks

  • Group 2: MNL: 2.35. MDLBT: 7.65 weeks

  • Group 3: MNL: 1.94. MDLBT: 7.00 weeks

  • Group 4: MNL: 4.22. MDLBT: 8.89 weeks

Interventions

Type of interventions:

  • Group 1: IL ZS with a solution of 2%

  • Group 2: IL 7% HSCS

  • Group 3: ILSSG 100 mg/mL

  • Group 4: a few lesions on unimportant and unexposed parts of the body were left as controls

Duration of intervention: not described

Outcomes

Healing: responses were graded according to scale (Sharquie 1988):

  • Slight: decreased erythema and oedema of the lesion; mild: reduction in the size of the lesion of up to 30%

  • Moderate: reduction in the size of the lesion of 30%‐60%

  • Marked: reduction in the size of the lesion by ≥ 60% and parasite not detected in the lesion by smear and/or culture

  • Total clearance of the lesion: with parasites not detected in the affected area by smear and/or culture

Speed of healing (time taken to be 'cured'); expressed in days

Prevention of scarring

Adverse effects

Time points reported: participants were followed up every 10‐15 days for a period of 45 days. At the end of the 6 weeks follow‐up period, the lesions were reassessed and parasitological proof of cure or otherwise was sought by smear and/or culture

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Insufficient information about the sequence generation process to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

No blinding or incomplete blinding, but the review authors judge that the outcome is not likely to be influenced by lack of blinding

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

High risk

Of the 85 participants initially recruited, 63 participants were followed‐up for the full observation period. Participants who did not show up after the first or second injection were excluded. We do not know numbers of missing data across intervention groups

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes, including those that were pre‐specified.

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported.

Sharquie 2001

Methods

Study design: randomised controlled trial

Setting/location: Iraq (outpatient department)

Study period: October 1994 to May 1995 and November 1995 to May 1996 (15 months)

Sample size calculation: not described

Participants

Type of Leishmania: L major and L tropica

Inclusion criteria: CL confirmed by smear/culture; acute CL of ≤ 12 weeks duration (to exclude the possibility of self healing); those for whom systemic treatment was indicated including those for whom having multiple lesions ( > 5) or large lesions ( > 4 cm²), that could not be injected, or who had lesions near to critical areas such as the eye; very young children for whom no local injection was attempted; participants who refused local treatment (2%)

Exclusion criteria: received antileishmanial treatment, either local or systemic, cases of reinfection

N randomised: 130. Group 1, 39; group 2, 37; group 3, 39; group 4, 15

Withdrawals: 27. Group 1, 8; group 2, 8; group 3, 7; group 4, 3

N assessed: 104. Group 1, 31; group 2, 29; group 3, 32; group 4, 12

Mean (SEM) age: group 1, 20.57 years (3.20); group 2, 22.55 years (2.72); group 3, 13.83 years (2.45); group 4, 25.74 years (4.45)

Sex (male/female): 62/68. Group 1, 13/18; group 2, 19/10; group 3, 11/21; group 4, 6/6

Baseline data:

  • Lesions (Total): group 1, 161; group 2, 145; group 3, 149; group 4, 42

  • Mean number of lesions (SEM): group 1, 5.19 (0.56); group 2, 5.00 (0.73); group 3, 4.65 (0.7); group 4, 3.50 (0.88)

  • Site of lesion (groups 1, 2, 3, and 4, respectively)

    • Head and neck: 45; 19; 50; 3

    • Lower limb: 21; 57; 52; 24

    • Upper limb: 90; 69; 44; 14

    • Trunk: 5;2;3;1

  • ‐Type of lesion:

    • Wet: group 1, 41; group 2, 32; group 3,: 20; group 4, 7.

    • Dry: group 1, 120; group 2, 113; group 3, 129; group 4, 36.

  • Mean duration of lesions (SEM): group 1, 8.1 weeks (0.56); group 2, 7.95 weeks (0.49); group 3, 8.50 weeks (0.54); group 4, 8.89 weeks (1.11)

Interventions

Type of interventions:

  • Group 1: ZS orally 2.5 mg/kg

  • Group 2: ZS orally 5 mg/kg

  • Group 3: ZS orally 10 mg/kg

  • Group 4: without treatment

Duration of intervention: ZS groups one capsule every 8 hours

Duration of follow‐up: 6 weeks

Outcomes

Healing: responses were graded according to scale (Sharquie 1988):

  • Slight: decreased erythema and oedema of the lesion; mild: reduction in the size of the lesion of up to 30%

  • Moderate: reduction in the size of the lesion of 30%‐60%

  • Marked: reduction in the size of the lesion by ≥ 60% and parasite not detected in the lesion by smear and/or culture

  • Total clearance of the lesion: with parasites not detected in the affected area by smear and/or culture

Speed of healing (time taken to be 'cured'); expressed in days

Prevention of scarring

Adverse effects

Time points reported: participants were followed up every 10‐15 days for a period of 45 days. At the end of the 6 weeks follow‐up period, the lesions were reassessed and parasitological proof of cure or otherwise was sought by smear and/or culture

Notes

Study funding sources: none declared

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not stated

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 26/130 (25%)

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes.

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Shazad 2005

Methods

Study design: randomised controlled trial

Setting/location: Military Base Clinic, Iran

Study period: January to October 2001 (10 months)

Sample size calculation: not described

Participants

Type of Leishmania: L major

Inclusion criteria: proven cases of CL, healthy apart from CL, lesions not in close proximity to a vital organ or joint, number of lesions 1 to 3, ulcer size < 5 cm in diameter, onset of the lesions < 3 months, no previous standard anti‐Leishmania treatment, no history of allergy to the paromomycin family

Exclusion criteria: not reported

N randomised: 60, 30 in each group

Withdrawals: 4. Group 1: 1; group 2: 3,

N assessed: 36. Group 1: 29; group 2: 27

Age: Group 1: approx 20.6 years; group 2: approx 21.7 years

Sex: all men

Baseline data:

  • Group 1: 76.7% had ulcerative, 8.3% nodular, and 15% papular type of lesions, and they were primarily located in the head and neck (25%), upper extremities (41.7%), lower extremities (31.7%) and trunk (1.7%). MNL: 2. MSL 21.7 mm. MDLBT: 37.8 days

  • Group 2: 69.7% had ulcerative, 11.8% nodule, and 18.4% papule type of lesions, and they were primarily located in the head and neck (13.2%), upper extremities (47.4%), lower extremities (36.8%) and trunk (2.6%). MNL: 2.4. MSL: 25 mm. MDLBT: 39 days

Interventions

Type of interventions:

  • Group 1: 15% PR sulphate and 10% urea. Dose: 0.5 mg/mm²/d

  • Group 2: intradermal MA every other day

Duration of intervention: 20 days

Outcomes

Healing rates:

  • Complete cure was defined as complete re‐epithelialisation of all lesions at one week after cessation of treatment. Participants with multiple lesions were considered to be cured if all the lesions were healed.

  • Partial cure (parasitological cure) was defined when clinical cure had not occurred in any lesion in a participant but no L amastigote was found in a direct smear of the lesions(s), 3 slides were prepared, and stained from each active ulcer, and 1000 fields were checked (approximately 3 slides).

  • Failure of treatment was defined as no clinical cure and no parasitological cure at one week after cessation of treatment; relapse was defined as a reappearance of the lesion after complete cure.

Adverse effects

Time points reported: follow‐up and clinical evaluation were performed at week 1 and 6 after completion of treatment. Participants were visited once at 6 months after treatment was completed.

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Study was described as open and no blinding was done

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Study was described as open and no blinding was done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: relevant outcomes were reported

Selective reporting (reporting bias)

Low risk

Comment: relevant outcomes were reported

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Stahl 2014

Methods

Study design: single‐centre, 3‐armed, open label, randomised, controlled, phase IIb trial

Setting/location: Leishmania and Malaria Centre (LMC) of the Provincial Balkh Civil Hospital Mazar‐e‐Sharif (Afghanistan)

Study period: November 2010 to March 2011 (15 months)

Sample size calculation: the sample size calculation was based on the per protocol (PP) analysis, defined as all participants evaluable with respect to the primary endpoint

Participants

Type of Leishmania: in 44/69 participants analysed, 28 (64%) lesions were infected with L tropica and 16 (36%) with L major

Inclusion criteria: CL lesions with Leishmania‐positive Giemsa smears without prior CL treatment

Exclusion criteria: age < 12 years; more than one skin lesion (to exclude intra‐individual variations in this phase IIb analysis); lesion age > 3 months; lesions located on eye lids, lips or nose; drug addiction; coinfection with Mycobacterium tuberculosis, HIV, or diabetes

N randomised: 87 participants: group 1, 24 (27.5%), group 2, 32 (36.8%), and group 3, 31 (35.6%)

Withdrawals: lost after registration: group 2, 3; group 3, 3. Not followed up for the full treatment period: SSG, 1; group 2, 6; group 3, 3. Mixed treatment: group 3, 2

N assessed: 81 (93.10%). group 1: 24; group 2: 29; group 3: 28

Mean age: 29 years (95% CI 25–33)

Sex (male/female): 32 (46%)/37 (54%)

Baseline data: comparable in all 3 regimens

Interventions

Type of interventions:

  • Group 1: intradermal injections of 0.6 mL SSG according to a protocol used by Zeglin 2009

  • Group 2: aseptic MWT with 0.045% DAC N‐055 following a single initial superficial wound debridement with HF‐EC, which was performed under local anaesthesia after wound cleansing and disinfection with gauzes soaked in physiological saline solution containing 320 ppm chlorine dioxide (pH 5.5 acidified 0.09% DAC N‐055) for 15 min

  • Group 3: MWT with 0.045% DAC N‐055 alone

Duration of intervention: for all groups: daily treatments (with the exception of Fridays) during the first week, followed by topical treatments at the centre twice a week until the end of week 4 and thereafter once a week until wound closure

In group 1, the SSG treatment was discontinued after week 4. In groups 2 and 3, participants dressed their lesion themselves after week 4 with the topical NaClO₂‐basic‐crème between their visits at the centre until lesion closure

Outcomes

Primary outcome was the ratio of closed versus open wounds at day 75 (D75) in the PP analysis for each regimen

Speed of healing: not reported in Methods

Adverse effects: not reported in Methods

6 visits were scheduled in the first week, 2 visits per week from weeks 2 to 4, and 1 visit per week thereafter until complete wound closure. Follow‐up visits were required once a month until day 180 after treatment start

Notes

The funders had no role in study design, data collection, data analysis, and interpretation, decision to publish, or preparation of the manuscript. KWS and HCS are members of the Board and CB is a member of the non‐profit NGO Waisenmedizin PACEM e.V. promoting access to essential medicine

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Each patient was randomly assigned to one of the 3 regimens by the random allocation generator in the computer‐based Leishmedoc system."

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Study was described as open and no blinding was done

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: "the present trial could not be conducted as either a double or a single blinded trial due to the physical nature of the applied interventions"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "The intention‐to‐treat analysis (ITT) is solely added as additional information. Patients that could not be evaluated were patients that were lost immediately after registration before treatment started. 81 out of 87 patients (93.1%) were suitable for the ITT and 69 (79.3%) for the PP analysis"

Selective reporting (reporting bias)

Low risk

Clinicaltrials.gov ID: NCT00996463. Registered: 15 October 2009

Other bias

Low risk

Other items assessed correctly reported

Yazdanpanah 2011

Methods

Study design: randomised, prospective, double‐blind trial

Setting/location: Dermatology Department, Ghaem Hospital, Mashhad University of Medical Sciences, Iran

Study period: October 2006 to May 2008 (19 months)

Sample size calculation: not described

Participants

Type of Leishmania: L tropica

Inclusion criteria: proven acute CL by a positive direct smear were selected for this study. Duration of lesions less than 6 months and no anti‐leishmaniasis treatment received during 2 months ago

Exclusion criteria: pregnant or nursing women, participants with hepatic, renal, and heart diseases

N randomised: 115. Group 1: 30, group 2: 85

Withdrawals: excluded because of side effects: group 2, 6. Not followed up for the full treatment period: group 1, 4; group 2, 5

N assessed: 100 (86.96%). Group 1: 26 (86.67%); group 2: 74 (87.06%)

Mean (SD) age: group 1, 32 years (23), group 2: 25 years (19)

Sex (male/female): group 1, 11/15, group 2: 39/35

Baseline data:

  • Total lesions: group 1, 43; group 2, 127

  • Location of the target lesion:

    • Group 1: head and neck 19 (44.3%), upper limb 18 (44.9%), lower limb 5 (11.6%), trunk 1 (2.3%)

    • Group 2: head and neck 56 (44.1%), upper limb 56 (44.1%), lower limb 14 (11%), trunk 1 (0.8%)

  • Mean duration of lesions (SD): group 1, 4.5 months (1.5); group 2, 4 months (1.5)

Interventions

Type of interventions:

  • Group 1: received oral zinc sulphate (ZS) in a dose of 10 mg/kg/d during 45‐day period before meal in 3 divided times

  • Group 2: IMMA (Glucantime; MA, Specia, Paris, France) 20 mg/kg/d for 20 days with a maximum of 3 vials

Duration of intervention: group 1: 45 days, group 2: 20 days

Outcomes

Decreased indurations of lesion (lesions were measured by palpation and ruler): in aspect of response to treatment in comparison with first visit participants graded as 4 improvement rate groups:

  1. Slight (up to 25% decreased indurations of lesion)

  2. Mild (25%–50% decreased indurations of lesion)

  3. Moderate (more than 50% and less than 75% decreased indurations of lesion)

  4. Total clearance (75% or more improvement or complete re‐epithelialisation without any indurations)

Decreased indurations of lesion after 45 days from completing treatment period: the same scale

Adverse effects: severe muscular pain and topical reaction of inoculation site (severe erythema and pruritus)

Time points reported: at the end of clinical treatment. After 45 days from completing treatment period.

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Using simple randomisation".

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open trial

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Not done. At each visit all lesions were reexamined by the same dermatologist. The size and induration of lesions were measured by palpation and ruler. No photographs were taken

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

100 out of 115 participants completed the study. 6 participants who received systemic MA were excluded because of side effects that included severe muscular pain and topical reaction of inoculation site (severe erythema and pruritus)

Selective reporting (reporting bias)

High risk

Protocol not available. Not registered. Alhtough most pre‐specified outcomes were reported, adverse effects were not clearly reported (only for those excluded from the analysis)

Other bias

Unclear risk

There was not enough information in the publication to assess if there were other biases present.

Zerehsaz 1999

Methods

Study design: randomised controlled trial

Setting/location: Fars province, Iran

Study period: not described

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania spp: L major and L tropica in the area

Inclusion criteria: diagnosed as having CL based on positive smears from lesions, and in some cases, cultures and histopathologic studies were also performed.

Exclusion criteria: pregnant, nursing, or serious concomitant diseases.

N randomised: 171

Withdrawals: 0

N assessed: 171 participants: 86 participants in the herbal extract Z‐HE group and 85 participants in the IMMA (15 to 20 mg/kg/d)

Age: range 10 months to 69 years

Sex (male/female): 84/87

Baseline data: the duration of the disease was < 4 months. Most participants had papular and papulonodular lesion(s), although other clinical forms including ulcerative, eczematoid, hyperkeratotic, and erysipeloid types were also present. Most participants had multiple lesions, and the most common sites were the face and the extremities.

Interventions

Type of interventions:

  • Group 1: herbal extract Z‐HE as a black paste applied to the lesions and covered by a dressing for 5 consecutive days + placebo (saline) injected (0.5 mL) for 20 consecutive days

  • Group 2: IMMA (15‐20 mg/kg/d) for 20 consecutive days + vehicle (petrolatum and charcoal powder) applied on the lesions as a black paste covered by a dressing for 5 consecutive days

Duration of intervention: 20 consecutive days

Outcomes

Healing rates: complete cure was defined as clinical improvement with complete healing and re‐epithelialisation of the lesion(s), partial cure as partial clinical improvement with reduction in infiltration erythema, and size of the lesion(s), and failure as the absence of any changes in the lesion(s) or progression and worsening of the lesion(s)

Adverse effects

Time points reported: 6 weeks post‐treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Insufficient information about the sequence generation process to permit judgment

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured, and unlikely that the blinding could have been broken.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No missing outcome data.

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgment

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

Özgöztasi 1997

Methods

Study design: randomised controlled trial

Setting/location: Turkey

Study period: around 8 weeks

Sample size calculation: not described

Participants

Type of Leishmania: Leishmania spp: L tropica in the area

Inclusion criteria: confirmed diagnosis of CL

Exclusion criteria: pregnant, nursing, or serious concomitant diseases

N randomised: 72

Withdrawals: 0

N assessed: 72 (100%): 40 participants in group 1 and 32 participants in group 2

Age: most were children aged 10 years or younger

Sex: both sexes

Baseline data: the most common site of the lesion was the face and most of the participants had one papulonodular lesion, with duration of the lesions varying from 1 to 12 months

  • Group 1: MNL: 1. MDLBT: 4.3 months

  • Group 2: MNL: 1. MDLBT: 4.3 months

Interventions

Type of interventions:

  • Group 1: 15% PR sulphate + 12% MBCL twice daily for 15 days

  • Group 2: ketoconazole orally 400 mg per day for 30 days (reduced to 200 mg if participants < 12 years old)

Duration of intervention: 4 weeks

Outcomes

Healing rates: percentage of participants 'cured' one month after treatment

Adverse effects

Time points reported: at the end of treatment and 4 weeks post‐treatment

Notes

Study funding sources: not reported

Possible conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were divided randomly into two treatment groups"

Comment: insufficient detail was reported about the method used to generate the allocation sequence.

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "An open‐labeled, N randomised controlled trial to evaluate the efficacy of paromomycin ointment as compared with ketoconazole"

Comment: we think the outcome is likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "No subjects withdrew because of this adverse effect"

Comment: no dropouts, so ITT analyses were performed.

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that the published reports include all expected outcomes.

Other bias

High risk

Sample size calculation and reporting of Leishmania spp involved was not correctly reported

ACL: anthroponotic cutaneous leishmaniasis; AL: allopurinol; CCR: complete clinical response; CL: cutaneous leishmaniasis; HF‐EC: high frequency electrocauterisation; HSCS: hypertonic sodium chloride solution; IL: intralesional; ILMA: intralesional meglumine antimoniate; ILSSG: intralesional sodium stibogluconate; IM: intramuscular; IMMA: intramuscular meglumine antimoniate; IQR: interquartile range; IV: intravenous; MA: meglumine antimoniate; MBCL: methyl benzethonium chloride; MDLBT: median duration of lesions before therapy; MNL: median number of lesions; MSL: median size of lesions; MWT: moist wound treatment; PDT: topical photodynamic therapy; PR: paromomycin; RF: radiofrequency; RFHT: radiofrequency heat therapy; SD: standard deviation, SEM: standard error of the mean; SSG: sodium stibogluconate; TCA: trichloroacetic acid; TM: ThermoMed; ZS: zinc sulphate;

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Alavi‐Naini 2012

Review

Banihashemi 2015

Inclusion criteria of the patients: patients with lupoid leishmaniasis, chronic recurrent leishmaniasis, that typically follows an acute form of cutaneus leishmaniasis

Bumb 2010

Not an RCT. "Selected patients were categorised into two groups (groups A and B) of 110 persons each. Alternate patients were included in group A and B, respectively"

Dogra 1986

They stated that participants were randomly selected for the study, but not randomly assigned to the treatment groups.

Dogra 1994

They stated that participants were randomly selected for the study, but not randomly assigned to the treatment groups.

Dorlo 2012

Review

El On 1992

Cross‐over study

Frankenburg 1993

It measured immunity parameters but did not show any clinical results.

Kim 2009

This was a meta‐analysis.

Moosavi 2005

The method of generation of randomisation was inappropriate. The author was contacted and stated that after enrolling the participants, they did consecutively allocate ILMA for odd number participants and used topical paromomycin for even number participants.

Nilforoushzadeh 2010

They stated that participants were randomly selected for the study, but not randomly assigned to the treatment groups.

Nilforoushzadeh 2011

Included immunocompromised and chronically ill patients

Siavash 2013

Only studied electrocardiogram and biochemical adverse effects of meglumine antimoniate

Singh 1995

They stated that participants in both groups were randomly selected. However, they did not have an initial population eligible for the study that was randomly divided in the 2 treatment groups. But rather one group of people that were randomly selected for one group and another group of participants that were also randomly selected to form part of the other group. Besides, the duration of treatments was not the same and the follow‐up was also different for both treatment groups.

Trau 1987

No clear data is available for multiple lesions. Only lesions from participants with multiple lesions were randomised. The lesions from participants with single lesions a cross‐over study was performed.

RCT: randomised controlled trial.

Characteristics of studies awaiting assessment [ordered by study ID]

Farajzadeh 2016a

Methods

Double‐blind randomised controlled trial

This study was carried out in some clinics of Kerman University of Medical Sciences (Iran), from October 2008 to December 2010.

Sample size calculation: a sample size of 40 participants per treatment group was planned, a probability of a type I error at alpha = 0.05 and beta = 0.1 to determine a 20% difference between intralesional injection of ZS 2 % solution with ILMA (Glucantime)

Participants

Type of Leishmania: L tropica

Inclusion criteria: the presence of parasitological confirmed lesion(s) of CL and aged 5–60 years

Exclusion criteria: disease duration more than 6 months, those with a history of hypersensitivity to MA (Glucantime) or ZS, pregnant or nursing women, those with more than 5 lesions, those with lesions with size of 5 cm or more, and a history of any anti‐leishmanial therapy during last 4 weeks

N randomised: 80, 40 in each group

Withdrawals: 34, 18 in the ZS‐treated group and 16 patients in the MA (Glucantime)‐treated group (lost to follow‐up in all of the participants except in one in the ZS‐treated group, who had severe necrosis)

N assessed: 46; 22 patients in the ZS‐treated group and 24 patients in the MA (Glucantime)‐treated group

There was no significant difference between compared groups' baseline characteristics

Interventions

Group 1: intralesional injection of ZS 2% vials once a week for 10 weeks or sooner in case of complete resolution of the lesions

Group 2: ILMA once a week for 10 weeks or sooner in case of complete resolution of the lesions

Co‐intervention: in both groups cryotherapy was performed once every other week for 10 weeks

Outcomes

Clinical response: no response to treatment was defined if the area of a lesion decreased less than 75%, partial treatment if decreased 75% to 99%, and complete treatment if decreased 100% compared to its baseline area

Time to healing (partial/complete treatment)

Adverse effects: there were no major side effects in either group. Pain was observed in all participants of both groups. In ZS group 4 participants developed necrosis of the site of the injection while this was not observed in MA (Glucantime) group at all

Time points reported: on average, each participant was followed for 4.1 weeks (SD 2.6).

Notes

Study funding sources: none declared

Possible conflicts of interest: there is no conflict of interest

Farajzadeh 2016b

Methods

Triple‐blind randomised controlled trial

Performed in Shahid Dadbin Clinic of Leishmaniasis Research Center (Iran), at Kerman University of Medical Sciences, between 2008 and 2010

Sample size calculation: a sample size of 44 participants per treatment group was planned with a probability of a type I error at alpha = 0.05 and beta = 0.1 to determine a 20% difference between topical terbinafine and vehicle in outcome.

Participants

Type of Leishmania: L major and L tropica in the area

Inclusion criteria: < 2 years of age; parasitological diagnosis of cutaneous leishmaniasis from all lesions with direct smear; ulcerative lesions with duration of less than one month that were not on exposed areas or joints; the size of lesions has to be < 3 cm; ≤ 3 lesions; no anti‐leishmanial therapy during last 2 weeks, and with no history of hypersensitivity to MA

Exclusion criteria: pregnant or nursing women, patients with hepatic, renal, or heart diseases

N randomised: 88, 44 in each group

Withdrawals: 36 in topical terbinafine + MA; 29 in vehicle + MA

N assessed (3 months): 9 in topical terbinafine + MA; 15 in vehicle + MA

There are significant differences between participants' sex in intervention and control groups

Interventions

Group 1: IMMA 20 mg/kg/d + topical terbinafine for 20 days

3 months follow‐up after completing of the treatment phase

Group 2: IMMA 20 mg/kg/d + vehicle (Mahan Vaseline) for 20 days

Visited at days: 0, 14, 30 and 44

Outcomes

Clinical cure, defined as:

  • Complete improvement: full re‐epithelialisation for ulcerative lesions and decrease in induration size(75%, with or without a negative direct smear result)

  • Partially improved: decrease in the indurations size between 25% and 75%

  • No change: decrease in the indurations size of 25%

Time to healing (partial/complete treatment)

Notes

Study funding sources: none declared

Possible conflicts of interest: there is no conflict of interest.

Hanif 2016

Methods

Randomised controlled study

Department of Dermatology, Sheikh Zayed Medical College/Hospital, Rahim Yar Khan, Pakistan, from November 2013 to June 2014

Sample size: was calculated by using a formula for 2 proportions; expected proportion for duration of treatment in one group was taken as 55, and in second group 51 days; power of study was taken as 80, and standard error of 5%

Participants

Type of Leishmania: not reported

Inclusion criteria: patients of all age groups except neonates, with single or multiple parasitologically confirmed lesions of cutaneous leishmaniasis with no past history of disease and not previously treated with SSG or MA

Exclusion criteria: retinal abnormality or visual field defects, pregnancy, hepatic dysfunction, known hypersensitivity to chloroquine and those having visceral leishmaniasis

N randomised: 86; 48 intralesional chloroquine, 38 oral chloroquine

Withdrawals: 0

N assessed: 86; 48 intralesional chloroquine, 38 oral chloroquine

Interventions

Intralesional and oral chloroquine administration

Co‐interventions: lesions with secondary bacterial infection before, during, or after treatment were treated with topical antibiotics. If such infections required systemic treatment, an antibiotic that has no activity against Leishmania (e.g. erythromycin) was given.

Outcomes

Cure was defined as the complete re‐epithelialisation of the ulcerated lesions, with no evidence of papules, inflammation, or induration

Quantitative variables like, duration, cost and total dose of treatment were calculated

Adverse effects

Time points reported: < 4 weeks, 5‐8 weeks, ≥ 9 weeks

Notes

Study funding sources: none declared

Possible conflicts of interest: none declared

Jaffary 2016

Methods

Interventional randomised controlled study was conducted in the Skin Disease and Leishmaniasis Research Center of Isfahan University of Medical Sciences, Isfahan, Iran, between April 2010 and May 2011

Sample size calculation: not described

Participants

Leishmania type: not described

Inclusion criteria: patients older than 5 years with biopsy‐confirmed cutaneous leishmaniasis were eligible if they had the following criteria: lesion diameter < 3 cm, disease duration < 12 weeks, lesion‐to‐eyelid distance > 2 cm, and no history of systemic or topical therapy for cutaneous leishmaniasis

Exclusion criteria: pregnancy, lactation, immunosuppressive therapy, and serious side effects of medication

N randomised: 90. 30 in each group (ILMA (Glucantime) alone, ILMA + TCA 50%, or fractional CO₂ laser)

Withdrawals: 14

N assessed: 76 participants (84.5%) completed treatment and were followed up for 60 days

Interventions

ILMA alone, ILMA + TCA 50% or fractional CO₂ laser for up to 8 weeks

Participants in all 3 groups were followed up weekly for the next 6 months.

Outcomes

The clinical response was rated as complete improvement (complete re‐epithelialisation of the lesion with negative direct skin smear result), partial improvement (50%–75% improvement of the lesion size), and no change in the lesion appearance.

Time to healing

Adverse effects

The improvement of scar was also evaluated according to the participant's satisfaction, the morphology of the lesion, the level of induration, and the level of atrophy and scored as follows: score of 1 (less than 5% improvement), score of 2 (25% to 50% improvement), score of 3 (51% to 75% improvement), and score of 4 (76% to 100% improvement)

Notes

Study funding sources: none declared

Possible conflicts of interest: there is no conflict of interest.

Na‐Bangchang 2016

Methods

Exploratory phase II, randomised, vehicle‐controlled, single‐centre (Ankober Health Center, North Shewa zone of Amhara region, Ethiopia) study

Sample size calculation: not described

Participants

Leishamania type: not described

Inclusion criteria: patients with localised cutaneous leishmaniasis (LCL) aged 18–65 years with a new, uncomplicated, localised, single lesion on the face or arms and positive parasite smear by microscopy

Exclusion criteria: cutaneous leishmaniasis with secondary infection, concomitant diseases (mucocutaneous or visceral leishmaniasis), history of anti‐leishmanial treatment within the past 6 months, abnormality of biochemical and/or haematological laboratory tests, known hypersensitivity to any of the Shiunko components, or pregnancy (positive urine HCG test), breastfeeding, or possibility of becoming pregnant during the study

N randomised: 40 were randomised to receive vehicle and Shiunko ointment (20 participants for each group)

Withdrawals: 2 participants did not receive complete treatment, and 9 were not completely followed up. In the vehicle group, 2 and 5 cases did not have complete treatment and follow‐up, respectively.

N assessed: 38 participants had complete treatment and 31, complete follow‐up. In the vehicle group, 18 and 15 cases had complete treatment and follow‐up, respectively.

Interventions

Group 1: the composition of Shiunko was as follows: 2.04 g shikon, 1.02 g tohki, 16.92 g sesame oil, and 6.76 g honeycomb wax

Group 2: the vehicle contained 20 g wax

Co‐interventions: cryotherapy was to be offered to all participants who were withdrawn or discontinued from the study for safety reasons

Vehicle and Shiunko ointment applied on the lesion twice a day for 4 weeks

Follow‐up: 16 weeks

Outcomes

Cure: complete cure was defined as complete wound closure and re‐epithelialisation without inflammation or infiltration and absence of parasite (amastigotes) within 12 weeks after the end of treatment. Partial response was defined as improvement of Leishmania signs (skin oedema, erythema, and/or hardening) and/or reduction in size but not total disappearance of the lesion and absence of parasite (amastigotes) within 12 weeks after the end of treatment. Treatment failure was defined as failure of the lesion size to decrease and/or lack of lesion sign improvement or re‐epithelialisation and/or presence of leishmanial amastigotes in the lesion 12 weeks after end of treatment (week 16)

Adverse effects

Clinical and parasitological assessments were performed before treatment, weekly for 4 weeks, and then 4, 8, and 12 weeks after the end of treatment

Notes

Study funding sources: the study was supported by the Institute of Tropical Medicine, Nagasaki University (Japan), ArmauerHansen Research Institute (Ethiopia), and Okusa Co., Ltd. (Japan).

Possible conflicts of interest: there is no conflict of interest.

Rajabi 2016

Methods

Clinical randomised trial conducted from March 2008 through 2009 in the dermatology department of Quaem University Hospital, Mashhad, Iran

Sample size calculation: not described

Participants

Leishamania type: not described

Inclusion criteria: positive smear for leishmaniasis with disease duration of less than 12 weeks, no treatment for the current condition before and volunteer to participate in the study

Exclusion criteria: pregnancy, breastfeeding, history of simultaneous treatment with other methods before or during the trial, history of local cutaneous or systemic diseases in the past 2 months, history of intolerance or allergy to macrolides, severe underlying diseases such as cardiovascular, renal, or hepatic diseases

N randomised: 96; 26 (43 lesions) azithromycin, 40 (54 lesions) MA (Glucantime)

Withdrawals: 2 azithromycin group

N assessed: 94; 24 (40 lesions) azithromycin, 40 (54 lesions) MA

Interventions

Group 1: topical liposomal form of azithromycin was administered for the first group twice daily.
Group 2: the other group was treated by weekly ILMA with a volume of 0.5–2 cm³ into each lesion till complete blanching of the lesion occurred

Clinical evaluations were performed every week during the treatment course (8 weeks) by a single dermatologist in both groups.

The participants were followed up for recurrence or complications 6 and 12 months after the end of the treatment course.

Outcomes

Complete cure was defined as full re‐epithelialisation, disappearance of oedema, induration, and other signs of inflammation, and a negative direct skin smear result

Improvement rate (%): complete improvement (full re‐epithelialisation of the lesions for ulcerative ones or disappearance of induration and erythema; significant improvement (decrease in induration size > 60%); moderate improvement (decrease in induration size between 30% and 60%; slight improvement (decrease in induration size of < 30%).

Adverse effects

Notes

Study funding sources: none declared

Possible conflicts of interest: none declared

Refai 2016

Methods

Interventional randomised study

Setting: Sri Lanka

Sample size calculation: not described

Participants

Leishmania type:L donovani

Inclusion criteria: Laboratory‐confirmed CL patients with single lesions

Exclusion criteria: —

N randomised: test group (n = 98); control group (n = 115)

Withdrawals: —

N assessed: —

Interventions

Group 1: single session of radiofrequency induced heat therapy (RFHT) at 50ºC for 30 s

Group 2: received weekly ILSSG until cure or 10 doses

Participants were followed up fortnightly for 12 weeks to assess clinical response and adverse effects.

Outcomes

Cure rate

Adverse effects

Time points reported: 8, 10, and 12 weeks

Notes

Poster

Sattar 2012

Methods

Randomised controlled trial

Participants

40 participants (no more data described in abstract)

Interventions

Topical ointment prepared from the stem extract of Morinda citrifolia (no more data described in abstract)

Outcomes

Improvement (no more data described in abstract)

Notes

Article request to authors

CL: cutaneous leishmaniasis; HCG: human chorionic gonadotropin; ILMA: intralesional meglumine antimoniate; IM: intramuscular; MA: meglumine antimoniate; SSG: sodium stibogluconate; ZS: zinc sulphate.

Characteristics of ongoing studies [ordered by study ID]

ACTRN12614001288617

Trial name or title

A clinical trial to assess the safety and effect of heat therapy in comparison to standard intra‐lesional sodium stibogluconate for cutaneous leishmaniasis

Methods

Randomised controlled trial

Participants

Inclusion criteria: suggestive skin lesions (papules, nodules, plaques, ulcers and nodule‐ulcers) who were clinically diagnosed by a consultant dermatologist and parasitologically confirmed as CL

Exclusion criteria: non‐localised leishmaniasis (VL, MCL, leishmaniasis recidivans, diffuse cutaneous leishmaniasis and post‐kala‐azar dermal leishmaniasis); use of prior concomitant treatment for leishmaniasis including any traditional medicines; lesions close to nasal, oral, urogenital, anal areas (mucosae) and the eyes; multiple lesions; any chronic or concomitant illnesses and people on pace makers and any metallic devices; pregnancy and breastfeeding mothers; children < 12 years of age; immunocompromised states including HIV/AIDS and use of immunosuppressants like steroids; alcohol abuse; not capable of understanding and complying with the study protocol; known hypersensitivity or allergy to treatment

Age: 12‐90 years

Interventions

The device used was the ThermoMed 1.8, ThermoSurgery Technologies portable battery operated device. The generator has received clearance by the US Food and Drug Administration for the treatment of CL. It delivers precisely controlled localised radiofrequency waves to selectively destroy diseased tissue. Heat therapy was administered as a single session. The lesion and surrounding normal skin was cleaned with normal saline. Then the area was anaesthetised with 2% lignocaine using a 1 cc syringe. The device was used in according to manufacturers instructions (ThermoMed 1.8; Thermosurgery). It generates a 6.78 mHz frequency applied with a hand set that is attached to 2 applicator electrodes which were placed on the diseased skin. A temperature of 50ºC was applied for 30 s to cover an area about 49‐73 mm², according to the size of electrodes (3 sizes are provided). Then the applicator is moved to another area of the lesion until the entire area is treated. Once treatment begins the temperature is measured by a thermistor embedded in the applicator which ensures that the applied temperature remains constant. After treatment the lesions were covered with gauze to prevent secondary infections and participants were given an antibiotic cream (soframycin 1% per 30 g depending on the size of lesion to cover the lesion) to be applied twice a day for 3 days.

Outcomes

Recurrence

Treatment efficacy was measured by the percentage of participants clinically cured by 8 weeks, 10 weeks, 12 weeks and 16 weeks after initiation of treatment in both groups.

  • Cure was defined as complete re‐epithelialisation of the CL lesion with no evidence of papules, active inflammation or induration.

  • Partially cured if healing was 50%‐100%

  • Non‐responders or treatment failures were those who had lesions that were less than 50% healed at the end of 3 months or if new lesions occur while on treatment

Any adverse effects like secondary bacterial infections, allergy to treatment or hypersensitivity and worsening of the lesion clinically was recorded during or after treatment with either therapy

Starting date

December 2014

Contact information

Dr Fathima Wardha Refa

PGIM address: No. 160, Professor Nandadasa Kodagoda Mawatha (Norris Canal Road), 00700, Colombo 7. 2.Department of Parasitology, Faculty of Medicine Colombo University of Colombo address: No 25, Kynsey Road, Colombo 8 0080, Sri Lanka. 7 MACLEOD ROAD COLOMBO 4, Sri Lanka

+94 727800003 or +94 11 2580093 or+94 11 2699284

[email protected]

Notes

IRCT138904091159N7

Trial name or title

Comparison between the efficacy of intralesional placebo and nitric oxide releasing patch versus placebo patch and Glucantime in the treatment of cutaneous leishmaniasis

Methods

Randomised, placebo‐controlled trial

Participants

Inclusion criteria: diagnosis of acute zoonotic cutaneous leishmaniasis, proven parasitology by direct smear; aged 18‐50 years; lesion diameter ≤ 3 cm; 1 lesion, not located on the face near the eyes, joints, cartilage, on the nose and ears; < 3 months passed from diseases times

Exclusion criteria: pregnancy and lactation; risk of presence or incidence of sporotrichoid or satellite lesions; history of hepatitis, heart, or kidney disorder; taking immunosuppressive medications (over the last 6 months); local or systemic treatment against leishmaniasis in the last 3 months; history of heart attack or high blood pressure; liver disease; anaemia; history of surgery for head and brain haemorrhage; taking blood pressure medications, antidepressants, or drugs used for angina

Interventions

Group 1: nitroglycerin patch (the pharmacy of Nour and Ali Asghar hospital in Isfahan) once a day covering 5.08 to 7.62 cm of lesion + intralesional injection of placebo (injection of distilled water) once a week (15‐30 mg) until whitening of base of the ulcer for 8 weeks or until complete cure

Group 2: ILMA (Meglusan) from Avenue de Scheut company in Belium, once a week (15‐30 mg) until whitening of base of the ulcer + placebo patch (the pharmacy of Nour and Ali Asghar hospital in Isfahan) once a day covering 5.08 to 7.62 cm of lesion for 8 weeks or until complete cure

Outcomes

Improvement rate. Timepoint: 1, 2, 3, 4, 7, 8 weeks during treatment and 12, 16, 20 weeks after end of treatment. Method of measurement: observation and examination

Induration. Timepoint: 1, 2, 3, 4, 7, 8, 12, 16, 20 weeks after end of treatment. Method of measurement: physical examination

Lesion area. Timepoint: 8, 12, 20 weeks after end of treatment

Starting date

August 2013

Contact information

Fariba Jaffary

Skin Diseases and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Sedigheh Tahereh (AS) Research Centers Complex, Khorram Ave., Isfahan, Iran Isfahan Iran, Islamic Republic Of Iran.

00983113373736

[email protected]

Notes

IRCT2013092414746N1

Trial name or title

The effect of MJ1 (topical dairy extract) versus routine care for treatment of cutaneous leishmaniasis (rural) in Isfahan Iran:a randomised controlled clinical trial (RCTs)

Methods

Randomised controlled trial

Participants

Inclusion criteria: cutaneous leishmaniasis of parasitology confirmed that they have not been previously treated; any other medical problem (such as heart disease, renal, hepatic, or haematological); available for follow‐up; informed consent to participate in the study

Exclusion criteria: secondary infections, history of allergic reactions

Interventions

Group 1: in this group patients apply MJ1 skin cream 3 times a day, they are trained to use it for a month with a diameter of 3 mm on the rub of the lesion without dressing

Group 2: skin cream MJ1 ‐ 3 times a day ‐ for a month

Outcomes

Lesion size, lesion diameters measured

Timepoint: 1, 2, 3, 4 and 8 weeks after starting treatment, participants are evaluated.

Method of measurement: at each visit, the 2 dimensions are measured and the area calculated

Starting date

December 2013

Contact information

Dr Mohsen Janghorbani

Hezar jarib Street, School of Public Health, Isfahan University of Medical Sciences Isfahan Iran, Islamic Republic Of

00983117922774

[email protected]

Notes

NCT00840359

Trial name or title

Phase 2 study of the efficacy of daylight activated photodynamic therapy in the treatment of cutaneous leishmaniasis

Methods

Randomised, parallel assignment, open label

Participants

Age: ≥ 18 years

Interventions

Group 1: photodynamic therapy Application of Metvix 16% cream followed by exposure to daylight for 2.5 h

Group 2: cryotherapy for 2 times 20 s

Outcomes

Eradiation of amastigotes

Starting date

September 2009

Contact information

Prof Claes D. Enk, Hadassah Medical Organization

Notes

NCT01050777

Trial name or title

Pilot study of efficacy of topical nano‐liposomal meglumine antimoniate (Glucantime) or paromomycin in combination with systemic Glucantime for the treatment of anthroponotic cutaneous leishmaniasis (ACL) caused by Leishmania tropica

Methods

Randomised, parallel assignment, double‐blind

Participants

Inclusion criteria: aged 12‐60 years; parasitologically proven CL due toL tropica; history of failure to at least one full course of systemic meglumine antimoniate (Glucantime); general good health based on history and physical examination; ≤ 4 lesions; lesion size < 3 cm; signed informed consent voluntarily and knowingly; guardian's signature for volunteer less than 18 years old

Exclusion criteria: pregnant or lactating women and those who are planning to be pregnant in next 60 days; use of other types of treatment for CL; involvement in any other drug or vaccine trial during the study period; known heart, kidney, liver diseases based on history and physical exam; abnormal ECG

Age: 12‐60 years

Interventions

Group 1: liposomal paromomycin (liposomes containing 10% paromomycin
) + liposomal MA

Group 2: liposomal MA (Glucantime)

Group 3: placebo + liposomal MA

Outcomes

Complete cure equal to complete re‐epithelialisation of all lesions

Starting date

March 2011

Contact information

Ali Khamesipour, Tehran University of Medical Sciences

Notes

SLCTR/2014/028

Trial name or title

Randomised, double‐blind, controlled study on efficacy and safety of intralesional metronidazole vs intralesional sodium stibogluconate in L donovani cutaneous leishmaniasis

Methods

Randomised controlled trial

Participants

Inclusion criteria: > 12 years of age, positive slit skin smear and/or skin biopsy for Leishmania parasites

Exclusion criteria: pregnancy, breastfeeding, known renal impairment, known liver impairment, congestive cardiac failure

Interventions

Group 1: intralesional metronidazole 0.2‐4 mL per lesion depending on the size of the lesion, weekly until cure or maximum of 10 injections

Group 2: intralesional stibogluconate 0.2‐4 mL per lesion depending on the size of the lesion, weekly until cure or maximum of 10 injections

Outcomes

Rate of clinical cure

Adverse effects – anticipated local side effects are pain, ulceration, scarring, postinflammatory hyperpigmentation or depigmentation. Systemic side effects are not anticipated as the drug is given intralesionally.

Starting date

November 2014

Contact information

Ranthilaka R Ranawaka

0112855200

[email protected]

Notes

CL: cutaneous leishmaniasis; ECG: electrocardiogram; ILMA: intralesional meglumine antimoniate.

Data and analyses

Open in table viewer
Comparison 1. ILMA weekly versus ILMA fortnightly for up to 8 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 1.1

Comparison 1 ILMA weekly versus ILMA fortnightly for up to 8 weeks, Outcome 1 Lesions cured.

Comparison 1 ILMA weekly versus ILMA fortnightly for up to 8 weeks, Outcome 1 Lesions cured.

Open in table viewer
Comparison 2. ILMA (every other day) versus IMMA (6 d/week) for up to 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 2.1

Comparison 2 ILMA (every other day) versus IMMA (6 d/week) for up to 4 weeks, Outcome 1 Lesions cured.

Comparison 2 ILMA (every other day) versus IMMA (6 d/week) for up to 4 weeks, Outcome 1 Lesions cured.

Open in table viewer
Comparison 3. IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (30 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 3.1

Comparison 3 IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (30 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.

Comparison 3 IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (30 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.

Open in table viewer
Comparison 4. IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.1

Comparison 4 IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.

Comparison 4 IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.

Open in table viewer
Comparison 5. IMMA (60 mg/kg/d for 3 weeks) + placebo versus IMMA (30 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 5.1

Comparison 5 IMMA (60 mg/kg/d for 3 weeks) + placebo versus IMMA (30 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.

Comparison 5 IMMA (60 mg/kg/d for 3 weeks) + placebo versus IMMA (30 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.

Open in table viewer
Comparison 6. IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

2

148

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.71, 0.96]

Analysis 6.1

Comparison 6 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Comparison 6 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 6.2

Comparison 6 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 2 Adverse effects.

Comparison 6 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 2 Adverse effects.

2.1 Serious adverse effects

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Skin reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Cardiac toxicity 'QT prolongation'

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 7. IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 7.1

Comparison 7 IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Comparison 7 IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 8. IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 8.1

Comparison 8 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Comparison 8 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 9. IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 9.1

Comparison 9 IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Comparison 9 IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 10. ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 10.1

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 1 Lesions cured.

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 1 Lesions cured.

2 Adverse effects (itching and burning) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 10.2

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 2 Adverse effects (itching and burning).

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 2 Adverse effects (itching and burning).

3 Adverse effects (oedema) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 10.3

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 3 Adverse effects (oedema).

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 3 Adverse effects (oedema).

Open in table viewer
Comparison 11. ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 11.1

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 1 Lesions cured.

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 1 Lesions cured.

2 Adverse effects (itching and burning) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 11.2

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 2 Adverse effects (itching and burning).

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 2 Adverse effects (itching and burning).

3 Adverse effects (oedema) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 11.3

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 3 Adverse effects (oedema).

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 3 Adverse effects (oedema).

Open in table viewer
Comparison 12. ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 12.1

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 1 Lesions cured.

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 1 Lesions cured.

2 Adverse effects (itching and burning) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 12.2

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 2 Adverse effects (itching and burning).

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 2 Adverse effects (itching and burning).

3 Adverse effects (oedema) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 12.3

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 3 Adverse effects (oedema).

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 3 Adverse effects (oedema).

Open in table viewer
Comparison 13. ILMA (weekly injections for 6 weeks) + gel mask twice a day versus ILMA (weekly injections for 6 weeks) + vehicle

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 13.1

Comparison 13 ILMA (weekly injections for 6 weeks) + gel mask twice a day versus ILMA (weekly injections for 6 weeks) + vehicle, Outcome 1 Participants complete cure.

Comparison 13 ILMA (weekly injections for 6 weeks) + gel mask twice a day versus ILMA (weekly injections for 6 weeks) + vehicle, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 14. ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5) versus ILSSG (1000 mg/mL days 1, 3, 5)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 14.1

Comparison 14 ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5) versus ILSSG (1000 mg/mL days 1, 3, 5), Outcome 1 Lesions cured.

Comparison 14 ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5) versus ILSSG (1000 mg/mL days 1, 3, 5), Outcome 1 Lesions cured.

Open in table viewer
Comparison 15. ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 15.1

Comparison 15 ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 1 Participants complete cure.

Comparison 15 ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 1 Participants complete cure.

2 Adverse effects (mild heart symptoms) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 15.2

Comparison 15 ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 2 Adverse effects (mild heart symptoms).

Comparison 15 ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 2 Adverse effects (mild heart symptoms).

Open in table viewer
Comparison 16. Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 16.1

Comparison 16 Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks, Outcome 1 Participants complete cure.

Comparison 16 Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks, Outcome 1 Participants complete cure.

2 Adverse effects (nausea and vomiting) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 16.2

Comparison 16 Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks, Outcome 2 Adverse effects (nausea and vomiting).

Comparison 16 Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks, Outcome 2 Adverse effects (nausea and vomiting).

Open in table viewer
Comparison 17. Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 17.1

Comparison 17 Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections), Outcome 1 Participants cured.

Comparison 17 Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections), Outcome 1 Participants cured.

2 Adverse effect (liver enzymes increase) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 17.2

Comparison 17 Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections), Outcome 2 Adverse effect (liver enzymes increase).

Comparison 17 Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections), Outcome 2 Adverse effect (liver enzymes increase).

Open in table viewer
Comparison 18. ILSSG (100 mg/mL days 1, 3, 5) + oral ketoconazole (600 mg/d for 4 weeks) versus ILSSG (100 mg/mL days 1, 3, 5)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 18.1

Comparison 18 ILSSG (100 mg/mL days 1, 3, 5) + oral ketoconazole (600 mg/d for 4 weeks) versus ILSSG (100 mg/mL days 1, 3, 5), Outcome 1 Lesions cured.

Comparison 18 ILSSG (100 mg/mL days 1, 3, 5) + oral ketoconazole (600 mg/d for 4 weeks) versus ILSSG (100 mg/mL days 1, 3, 5), Outcome 1 Lesions cured.

Open in table viewer
Comparison 19. ILSSG (100 mg/mL days 1, 3, 5) + ketoconazole (600 mg/d for 4 weeks) versus ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 19.1

Comparison 19 ILSSG (100 mg/mL days 1, 3, 5) + ketoconazole (600 mg/d for 4 weeks) versus ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5), Outcome 1 Lesions cured.

Comparison 19 ILSSG (100 mg/mL days 1, 3, 5) + ketoconazole (600 mg/d for 4 weeks) versus ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5), Outcome 1 Lesions cured.

Open in table viewer
Comparison 20. Itraconazole (200 mg for 6 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 20.1

Comparison 20 Itraconazole (200 mg for 6 weeks) versus placebo, Outcome 1 Participants complete cure.

Comparison 20 Itraconazole (200 mg for 6 weeks) versus placebo, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 21. Itraconazole (200 mg for 3 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 21.1

Comparison 21 Itraconazole (200 mg for 3 weeks) versus placebo, Outcome 1 Participants complete cure.

Comparison 21 Itraconazole (200 mg for 3 weeks) versus placebo, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 22. Itraconazole (200 mg for 6 to 8 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

3

244

Risk Ratio (M‐H, Random, 99% CI)

3.70 [0.35, 38.99]

Analysis 22.1

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 1 Participants complete cure.

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 22.2

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 2 Adverse effects.

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 2 Adverse effects.

2.1 Mild abdominal pain and nausea

3

204

Risk Ratio (M‐H, Random, 95% CI)

2.36 [0.74, 7.47]

2.2 Mild abnormal liver function

3

84

Risk Ratio (M‐H, Random, 95% CI)

3.08 [0.53, 17.98]

2.3 Headache and dizziness

1

20

Risk Ratio (M‐H, Random, 95% CI)

2.63 [0.16, 43.63]

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 22.3

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 3 Microbiological cure of skin lesions.

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 3 Microbiological cure of skin lesions.

Open in table viewer
Comparison 23. Itraconazole (200 mg for 6 to 8 weeks) versus no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 23.1

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 1 Participants complete cure.

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 1 Participants complete cure.

2 Adverse effects (headache and dizziness) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 23.2

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 2 Adverse effects (headache and dizziness).

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 2 Adverse effects (headache and dizziness).

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 23.3

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 3 Microbiological cure of skin lesions.

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 3 Microbiological cure of skin lesions.

Open in table viewer
Comparison 24. Fluconazole (200 mg for 6 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 24.1

Comparison 24 Fluconazole (200 mg for 6 weeks) versus placebo, Outcome 1 Lesions cured.

Comparison 24 Fluconazole (200 mg for 6 weeks) versus placebo, Outcome 1 Lesions cured.

2 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 24.2

Comparison 24 Fluconazole (200 mg for 6 weeks) versus placebo, Outcome 2 Participants complete cure.

Comparison 24 Fluconazole (200 mg for 6 weeks) versus placebo, Outcome 2 Participants complete cure.

Open in table viewer
Comparison 25. Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 25.1

Comparison 25 Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks), Outcome 1 Participants complete cure.

Comparison 25 Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks), Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 25.2

Comparison 25 Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks), Outcome 2 Adverse effects.

Comparison 25 Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks), Outcome 2 Adverse effects.

2.1 Rise creatinine and liver enzymes

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Cheilitis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Nausea

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 26. Oral dapsone (200 mg/d for 6 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete Cure Show forest plot

2

160

Risk Ratio (M‐H, Random, 95% CI)

24.08 [1.44, 403.43]

Analysis 26.1

Comparison 26 Oral dapsone (200 mg/d for 6 weeks) versus placebo, Outcome 1 Participants complete Cure.

Comparison 26 Oral dapsone (200 mg/d for 6 weeks) versus placebo, Outcome 1 Participants complete Cure.

2 Adverse effects Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 26.2

Comparison 26 Oral dapsone (200 mg/d for 6 weeks) versus placebo, Outcome 2 Adverse effects.

Comparison 26 Oral dapsone (200 mg/d for 6 weeks) versus placebo, Outcome 2 Adverse effects.

2.1 Nausea

2

160

Risk Ratio (M‐H, Random, 95% CI)

21.86 [3.04, 157.29]

Open in table viewer
Comparison 27. Allopurinol (15 mg/kg/d for 3 weeks) + IMMA (20 mg/kg/d for 2 weeks) versus allopurinol (15 mg/kg/d for 3 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

3.83 [1.71, 8.60]

Analysis 27.1

Comparison 27 Allopurinol (15 mg/kg/d for 3 weeks) + IMMA (20 mg/kg/d for 2 weeks) versus allopurinol (15 mg/kg/d for 3 weeks), Outcome 1 Lesions cured.

Comparison 27 Allopurinol (15 mg/kg/d for 3 weeks) + IMMA (20 mg/kg/d for 2 weeks) versus allopurinol (15 mg/kg/d for 3 weeks), Outcome 1 Lesions cured.

Open in table viewer
Comparison 28. Allopurinol (15mg/kg/d for 3 weeks)+ IMMA (20 mg/kg/d for 2 weeks) versus IMMA (20 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

1.92 [1.08, 3.41]

Analysis 28.1

Comparison 28 Allopurinol (15mg/kg/d for 3 weeks)+ IMMA (20 mg/kg/d for 2 weeks) versus IMMA (20 mg/kg/d for 2 weeks), Outcome 1 Lesions cured.

Comparison 28 Allopurinol (15mg/kg/d for 3 weeks)+ IMMA (20 mg/kg/d for 2 weeks) versus IMMA (20 mg/kg/d for 2 weeks), Outcome 1 Lesions cured.

Open in table viewer
Comparison 29. Allopurinol (15 mg/kg/d for 3 weeks) versus IMMA (20 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions Cured Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.35, 1.62]

Analysis 29.1

Comparison 29 Allopurinol (15 mg/kg/d for 3 weeks) versus IMMA (20 mg/kg/d for 2 weeks), Outcome 1 Lesions Cured.

Comparison 29 Allopurinol (15 mg/kg/d for 3 weeks) versus IMMA (20 mg/kg/d for 2 weeks), Outcome 1 Lesions Cured.

Open in table viewer
Comparison 30. Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 30.1

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 1 Lesions cured.

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 30.2

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 2 Adverse effects.

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 2 Adverse effects.

2.1 Mild abdominal pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Skin eruption

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Muscle pain and weakness

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Microbiological cure of skin lesions Show forest plot

1

72

Risk Ratio (M‐H, Random, 95% CI)

1.11 [0.88, 1.41]

Analysis 30.3

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 3 Microbiological cure of skin lesions.

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 3 Microbiological cure of skin lesions.

Open in table viewer
Comparison 31. Allopurinol (20 mg/kg/d for 3 weeks)+ IMMA (10 mg/kg/d for 20 d) versus IMMA (20 mg/kg/d for 28 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 31.1

Comparison 31 Allopurinol (20 mg/kg/d for 3 weeks)+ IMMA (10 mg/kg/d for 20 d) versus IMMA (20 mg/kg/d for 28 d), Outcome 1 Adverse effects.

Comparison 31 Allopurinol (20 mg/kg/d for 3 weeks)+ IMMA (10 mg/kg/d for 20 d) versus IMMA (20 mg/kg/d for 28 d), Outcome 1 Adverse effects.

1.1 Secondary infection

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Myalgia

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.3 ECG changes

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.4 Chest pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.5 Pain injection site

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.6 Abscess

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 32. Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 32.1

Comparison 32 Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d), Outcome 1 Participants complete cured.

Comparison 32 Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d), Outcome 1 Participants complete cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 32.2

Comparison 32 Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d), Outcome 2 Adverse effects.

Comparison 32 Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d), Outcome 2 Adverse effects.

2.1 Abdominal symptoms

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Liver abnormalities

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Myalgia

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.4 Rash

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 33. Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 33.1

Comparison 33 Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo, Outcome 1 Participants complete cure.

Comparison 33 Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo, Outcome 1 Participants complete cure.

1.1 Evaluated 3 months after treatment

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Evaluated after treatment

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 33.2

Comparison 33 Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo, Outcome 2 Microbiological cure of skin lesions.

Comparison 33 Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo, Outcome 2 Microbiological cure of skin lesions.

Open in table viewer
Comparison 34. Oral rifampicin (10 mg/kg/d) + omeprazole (20 mg/d) for 6 weeks versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 34.1

Comparison 34 Oral rifampicin (10 mg/kg/d) + omeprazole (20 mg/d) for 6 weeks versus placebo, Outcome 1 Participants complete cure.

Comparison 34 Oral rifampicin (10 mg/kg/d) + omeprazole (20 mg/d) for 6 weeks versus placebo, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 35. Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 35.1

Comparison 35 Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d), Outcome 1 Lesions cured.

Comparison 35 Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d), Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 35.2

Comparison 35 Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d), Outcome 2 Adverse effects.

Comparison 35 Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d), Outcome 2 Adverse effects.

2.1 Nausea and vomiting

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 36. Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 36.1

Comparison 36 Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d), Outcome 1 Participants complete cure.

Comparison 36 Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d), Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 36.2

Comparison 36 Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d), Outcome 2 Adverse effects.

Comparison 36 Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d), Outcome 2 Adverse effects.

2.1 Gastrointestinal complaints and headache

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Gastrointestinal complications

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Myalgia

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 37. Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 37.1

Comparison 37 Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d, Outcome 1 Participants complete cure.

Comparison 37 Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 37.2

Comparison 37 Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d, Outcome 2 Adverse effects.

Comparison 37 Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d, Outcome 2 Adverse effects.

2.1 Allergic macule‐papular

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 38. Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 38.1

Comparison 38 Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks), Outcome 1 Participants complete cure.

Comparison 38 Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks), Outcome 1 Participants complete cure.

Open in table viewer
Comparison 39. Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 39.1

Comparison 39 Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks), Outcome 1 Participants complete cure.

Comparison 39 Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks), Outcome 1 Participants complete cure.

Open in table viewer
Comparison 40. Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 40.1

Comparison 40 Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d, Outcome 1 Participants complete cure.

Comparison 40 Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 40.2

Comparison 40 Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d, Outcome 2 Adverse effects.

Comparison 40 Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d, Outcome 2 Adverse effects.

2.1 Nausea and vomiting

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 41. Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 41.1

Comparison 41 Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 1 Participants complete cure.

Comparison 41 Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 41.2

Comparison 41 Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 2 Adverse effects.

Comparison 41 Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 2 Adverse effects.

2.1 Nausea and vomiting

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 42. Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 42.1

Comparison 42 Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 1 Participants complete cure.

Comparison 42 Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 42.2

Comparison 42 Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 2 Adverse effects.

Comparison 42 Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 2 Adverse effects.

2.1 Nausea and vomiting

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 43. Oral zinc sulphate (10 mg/kg/d for 45 d) versus IMMA (20 mg/kg/d for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 43.1

Comparison 43 Oral zinc sulphate (10 mg/kg/d for 45 d) versus IMMA (20 mg/kg/d for 20 d), Outcome 1 Participants complete cure.

Comparison 43 Oral zinc sulphate (10 mg/kg/d for 45 d) versus IMMA (20 mg/kg/d for 20 d), Outcome 1 Participants complete cure.

Open in table viewer
Comparison 44. Artesunate 400 mg + sulphamethoxypyrazine/pyrimethamine 1000 mg/50 mg 4 times daily for 4 d versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 44.1

Comparison 44 Artesunate 400 mg + sulphamethoxypyrazine/pyrimethamine 1000 mg/50 mg 4 times daily for 4 d versus placebo, Outcome 1 Participants complete cure.

Comparison 44 Artesunate 400 mg + sulphamethoxypyrazine/pyrimethamine 1000 mg/50 mg 4 times daily for 4 d versus placebo, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 45. Topical 2% miconazole (twice a day) versus topical 1% clotrimazole (twice a day) for 30 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 45.1

Comparison 45 Topical 2% miconazole (twice a day) versus topical 1% clotrimazole (twice a day) for 30 d, Outcome 1 Lesions cured.

Comparison 45 Topical 2% miconazole (twice a day) versus topical 1% clotrimazole (twice a day) for 30 d, Outcome 1 Lesions cured.

Open in table viewer
Comparison 46. Topical ketoconazole (twice a day) versus vehicle (twice a day) for 30 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 46.1

Comparison 46 Topical ketoconazole (twice a day) versus vehicle (twice a day) for 30 d, Outcome 1 Participants complete cure.

Comparison 46 Topical ketoconazole (twice a day) versus vehicle (twice a day) for 30 d, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 47. Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure (ITT) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 47.1

Comparison 47 Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks, Outcome 1 Participants complete cure (ITT).

Comparison 47 Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks, Outcome 1 Participants complete cure (ITT).

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 47.2

Comparison 47 Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks, Outcome 2 Adverse effects.

Comparison 47 Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks, Outcome 2 Adverse effects.

2.1 Hypersensitivity

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Mild pruritus

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Erithema and oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 48. Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 48.1

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 1 Lesions cured.

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 1 Lesions cured.

2 Scarring Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 48.2

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 2 Scarring.

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 2 Scarring.

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 48.3

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 3 Microbiological cure of skin lesions.

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 3 Microbiological cure of skin lesions.

Open in table viewer
Comparison 49. Paromomycin (twice daily for 30 d) versus vehicle (twice daily for 30 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.71 [0.25, 2.06]

Analysis 49.1

Comparison 49 Paromomycin (twice daily for 30 d) versus vehicle (twice daily for 30 d), Outcome 1 Lesions cured.

Comparison 49 Paromomycin (twice daily for 30 d) versus vehicle (twice daily for 30 d), Outcome 1 Lesions cured.

Open in table viewer
Comparison 50. Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

2

383

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.86, 1.17]

Analysis 50.1

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 1 Participants complete cure.

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 50.2

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 2 Adverse effects.

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 2 Adverse effects.

2.1 Skin/local reaction

3

463

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.45, 1.93]

3 Microbiological cure of skin lesions Show forest plot

2

383

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.88, 1.20]

Analysis 50.3

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 3 Microbiological cure of skin lesions.

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 3 Microbiological cure of skin lesions.

Open in table viewer
Comparison 51. Paromomycin 15% (daily for 20 d) versus vehicle

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 51.1

Comparison 51 Paromomycin 15% (daily for 20 d) versus vehicle, Outcome 1 Participants complete cure.

Comparison 51 Paromomycin 15% (daily for 20 d) versus vehicle, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 52. Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus vehicle

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 52.1

Comparison 52 Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus vehicle, Outcome 1 Participants complete cure.

Comparison 52 Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus vehicle, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 53. Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus paromomycin 15% alone (daily for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 53.1

Comparison 53 Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus paromomycin 15% alone (daily for 20 d), Outcome 1 Participants complete cure.

Comparison 53 Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus paromomycin 15% alone (daily for 20 d), Outcome 1 Participants complete cure.

Open in table viewer
Comparison 54. Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 54.1

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 1 Participants complete cure.

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 1 Participants complete cure.

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 54.2

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 2 Recurrence.

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 2 Recurrence.

3 Scarring Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 54.3

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 3 Scarring.

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 3 Scarring.

Open in table viewer
Comparison 55. Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 55.1

Comparison 55 Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d), Outcome 1 Participants complete cure.

Comparison 55 Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d), Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 55.2

Comparison 55 Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d), Outcome 2 Adverse effects.

Comparison 55 Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d), Outcome 2 Adverse effects.

2.1 Cutaneous reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 56. Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 56.1

Comparison 56 Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d), Outcome 1 Participants complete cure.

Comparison 56 Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d), Outcome 1 Participants complete cure.

2 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 56.2

Comparison 56 Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d), Outcome 2 Microbiological cure of skin lesions.

Comparison 56 Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d), Outcome 2 Microbiological cure of skin lesions.

Open in table viewer
Comparison 57. Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 57.1

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 1 Lesions cured.

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 1 Lesions cured.

2 Scarring Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 57.2

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 2 Scarring.

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 2 Scarring.

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 57.3

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 3 Microbiological cure of skin lesions.

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 3 Microbiological cure of skin lesions.

Open in table viewer
Comparison 58. Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 58.1

Comparison 58 Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks), Outcome 1 Participants complete cure.

Comparison 58 Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks), Outcome 1 Participants complete cure.

2 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 58.2

Comparison 58 Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks), Outcome 2 Microbiological cure of skin lesions.

Comparison 58 Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks), Outcome 2 Microbiological cure of skin lesions.

Open in table viewer
Comparison 59. IL zinc 2% (twice a week for 2 weeks) versus ILSSG (100 mg/mL) for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 59.1

Comparison 59 IL zinc 2% (twice a week for 2 weeks) versus ILSSG (100 mg/mL) for 2 weeks), Outcome 1 Lesions cured.

Comparison 59 IL zinc 2% (twice a week for 2 weeks) versus ILSSG (100 mg/mL) for 2 weeks), Outcome 1 Lesions cured.

Open in table viewer
Comparison 60. IL zinc 2% (twice a week for 2 weeks) versus IL 7% HSCS for 2 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 60.1

Comparison 60 IL zinc 2% (twice a week for 2 weeks) versus IL 7% HSCS for 2 weeks, Outcome 1 Lesions cured.

Comparison 60 IL zinc 2% (twice a week for 2 weeks) versus IL 7% HSCS for 2 weeks, Outcome 1 Lesions cured.

Open in table viewer
Comparison 61. ILSSG (100 mg/mL) for 2 weeks versus IL 7% HSCS for 2 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 61.1

Comparison 61 ILSSG (100 mg/mL) for 2 weeks versus IL 7% HSCS for 2 weeks, Outcome 1 Lesions cured.

Comparison 61 ILSSG (100 mg/mL) for 2 weeks versus IL 7% HSCS for 2 weeks, Outcome 1 Lesions cured.

Open in table viewer
Comparison 62. IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 62.1

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 1 Lesions cured.

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 1 Lesions cured.

2 Participants complete cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 62.2

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 2 Participants complete cured.

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 2 Participants complete cured.

3 Adverse effects Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 62.3

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 3 Adverse effects.

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 3 Adverse effects.

3.1 Pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.2 Burning

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.3 Itching

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.4 Inflammation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.5 Pruritus and erythema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.6 Severe pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 63. IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 63.1

Comparison 63 IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks), Outcome 1 Lesions cured.

Comparison 63 IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks), Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 63.2

Comparison 63 IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks), Outcome 2 Adverse effects.

Comparison 63 IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks), Outcome 2 Adverse effects.

2.1 Inflammation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 64. Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 64.1

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 1 Participants complete cure.

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 1 Participants complete cure.

2 Participants with treated lesions that recur Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 64.2

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 2 Participants with treated lesions that recur.

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 2 Participants with treated lesions that recur.

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 64.3

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 3 Adverse effects.

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 3 Adverse effects.

3.1 Itch and burning

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 65. IL 7% HSCS (0.2 mL to 7 mL per lesion) versus ILSSG (max 2 mL) max 5 injections

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 65.1

Comparison 65 IL 7% HSCS (0.2 mL to 7 mL per lesion) versus ILSSG (max 2 mL) max 5 injections, Outcome 1 Lesions cured.

Comparison 65 IL 7% HSCS (0.2 mL to 7 mL per lesion) versus ILSSG (max 2 mL) max 5 injections, Outcome 1 Lesions cured.

Open in table viewer
Comparison 66. IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 66.1

Comparison 66 IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks, Outcome 1 Lesions cured.

Comparison 66 IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks, Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 66.2

Comparison 66 IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks, Outcome 2 Adverse effects.

Comparison 66 IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks, Outcome 2 Adverse effects.

2.1 Sporotrichotic dissemination

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 67. IL 7% HSCS (0.1 mL to 0.5 mL per lesion) versus IL 2% ciprofloxacin solution (0.1 mL to 0.5 mL per lesion)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 67.1

Comparison 67 IL 7% HSCS (0.1 mL to 0.5 mL per lesion) versus IL 2% ciprofloxacin solution (0.1 mL to 0.5 mL per lesion), Outcome 1 Lesions cured.

Comparison 67 IL 7% HSCS (0.1 mL to 0.5 mL per lesion) versus IL 2% ciprofloxacin solution (0.1 mL to 0.5 mL per lesion), Outcome 1 Lesions cured.

Open in table viewer
Comparison 68. IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 68.1

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 68.2

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.

3 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 68.3

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).

4 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 68.4

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.

4.1 Ulceration and necrosis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 69. ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 69.1

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 69.2

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.

3 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 69.3

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).

4 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 69.4

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.

4.1 Ulceration and necrosis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 70. ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 70.1

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 70.2

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.

3 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 70.3

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).

4 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 70.4

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.

4.1 Ulceration and necrosis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 71. IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 71.1

Comparison 71 IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks, Outcome 1 Lesions cured.

Comparison 71 IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks, Outcome 1 Lesions cured.

2 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 71.2

Comparison 71 IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks, Outcome 2 Microbiological cure of skin lesions.

Comparison 71 IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks, Outcome 2 Microbiological cure of skin lesions.

Open in table viewer
Comparison 72. WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 72.1

Comparison 72 WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d), Outcome 1 Participants complete cure.

Comparison 72 WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d), Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 72.2

Comparison 72 WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d), Outcome 2 Adverse effects.

Comparison 72 WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d), Outcome 2 Adverse effects.

2.1 Erythema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Mild pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Hearing acuity problems

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 73. IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 73.1

Comparison 73 IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks, Outcome 1 Participants complete cure.

Comparison 73 IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 73.2

Comparison 73 IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks, Outcome 2 Adverse effects.

Comparison 73 IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks, Outcome 2 Adverse effects.

2.1 Local inflammatory reactions

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 74. Topical miltefosine 6% (once daily) versus ILMA (twice a week) for up to 28 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 74.1

Comparison 74 Topical miltefosine 6% (once daily) versus ILMA (twice a week) for up to 28 d, Outcome 1 Participants complete cure.

Comparison 74 Topical miltefosine 6% (once daily) versus ILMA (twice a week) for up to 28 d, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 75. Dapsone gel 5% (twice a day) + ILMA (weekly) versus cryotherapy (every 2 weeks) + IMMA (weekly) for up to 16 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 75.1

Comparison 75 Dapsone gel 5% (twice a day) + ILMA (weekly) versus cryotherapy (every 2 weeks) + IMMA (weekly) for up to 16 weeks, Outcome 1 Lesions cured.

Comparison 75 Dapsone gel 5% (twice a day) + ILMA (weekly) versus cryotherapy (every 2 weeks) + IMMA (weekly) for up to 16 weeks, Outcome 1 Lesions cured.

Open in table viewer
Comparison 76. DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 76.1

Comparison 76 DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d, Outcome 1 Participants complete cure.

Comparison 76 DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 76.2

Comparison 76 DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d, Outcome 2 Adverse effects.

Comparison 76 DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d, Outcome 2 Adverse effects.

2.1 Reulceration

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Keloïd scars

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 77. DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 77.1

Comparison 77 DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 1 Participants complete cure.

Comparison 77 DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 77.2

Comparison 77 DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 2 Adverse effects.

Comparison 77 DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 2 Adverse effects.

2.1 Reulceration

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Keloïd scars

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 78. DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 78.1

Comparison 78 DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 1 Participants complete cure.

Comparison 78 DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 78.2

Comparison 78 DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 2 Adverse effects.

Comparison 78 DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 2 Adverse effects.

2.1 Reulceration

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Keloïd scars

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 79. Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 79.1

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 1 Lesions cured.

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 1 Lesions cured.

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 79.2

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 2 Recurrence.

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 2 Recurrence.

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 79.3

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 3 Adverse effects.

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 3 Adverse effects.

3.1 Dizziness and nausea

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.2 Hypersensitive reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 80. CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

433

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.53, 1.55]

Analysis 80.1

Comparison 80 CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d, Outcome 1 Lesions cured.

Comparison 80 CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d, Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 80.2

Comparison 80 CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d, Outcome 2 Adverse effects.

Comparison 80 CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d, Outcome 2 Adverse effects.

2.1 Hyperpigmentation and redness

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Hypertrophic scarring

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Systemic symptoms

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 81. CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 81.1

Comparison 81 CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks, Outcome 1 Lesions cured.

Comparison 81 CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks, Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 81.2

Comparison 81 CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks, Outcome 2 Adverse effects.

Comparison 81 CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks, Outcome 2 Adverse effects.

2.1 Hyperpigmentation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Atrophic scar

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Hypopigmentation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 82. Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Partcipants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 82.1

Comparison 82 Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser, Outcome 1 Partcipants complete cure.

Comparison 82 Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser, Outcome 1 Partcipants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 82.2

Comparison 82 Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser, Outcome 2 Adverse effects.

Comparison 82 Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser, Outcome 2 Adverse effects.

2.1 Erythema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 83. TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 83.1

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 1 Participants complete cure.

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 1 Participants complete cure.

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 83.2

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 2 Recurrence.

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 2 Recurrence.

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 83.3

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 3 Adverse effects.

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 3 Adverse effects.

3.1 Mild erythema and itch

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Microbiological cure of skin lesions Show forest plot

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.74, 1.26]

Analysis 83.4

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 4 Microbiological cure of skin lesions.

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 4 Microbiological cure of skin lesions.

Open in table viewer
Comparison 84. Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 84.1

Comparison 84 Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks, Outcome 1 Participants complete cure.

Comparison 84 Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks, Outcome 1 Participants complete cure.

2 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 84.2

Comparison 84 Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks, Outcome 2 Lesions cured.

Comparison 84 Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks, Outcome 2 Lesions cured.

2.1 Males

1

Risk Ratio (M‐H, Random, 99% CI)

0.0 [0.0, 0.0]

2.2 Females

1

Risk Ratio (M‐H, Random, 99% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 85. TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 85.1

Comparison 85 TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks), Outcome 1 Participants complete cure.

Comparison 85 TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks), Outcome 1 Participants complete cure.

2 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 85.2

Comparison 85 TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks), Outcome 2 Speed of healing (weeks).

Comparison 85 TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks), Outcome 2 Speed of healing (weeks).

Open in table viewer
Comparison 86. Fractional laser + ILMA (fortnightly 2 sessions) versus ILMA alone (twice a week for up to 8 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 86.1

Comparison 86 Fractional laser + ILMA (fortnightly 2 sessions) versus ILMA alone (twice a week for up to 8 weeks), Outcome 1 Participants complete cure.

Comparison 86 Fractional laser + ILMA (fortnightly 2 sessions) versus ILMA alone (twice a week for up to 8 weeks), Outcome 1 Participants complete cure.

Open in table viewer
Comparison 87. TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 87.1

Comparison 87 TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions), Outcome 1 Participants complete cure.

Comparison 87 TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions), Outcome 1 Participants complete cure.

2 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 87.2

Comparison 87 TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions), Outcome 2 Speed of healing (weeks).

Comparison 87 TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions), Outcome 2 Speed of healing (weeks).

Open in table viewer
Comparison 88. TCA fortnightly up to 8 weeks + ILMA (twice a week) versus ILMA alone (weekly for up to 8 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 88.1

Comparison 88 TCA fortnightly up to 8 weeks + ILMA (twice a week) versus ILMA alone (weekly for up to 8 weeks), Outcome 1 Participants complete cure.

Comparison 88 TCA fortnightly up to 8 weeks + ILMA (twice a week) versus ILMA alone (weekly for up to 8 weeks), Outcome 1 Participants complete cure.

Open in table viewer
Comparison 89. Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 89.1

Comparison 89 Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 89 Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 89.2

Comparison 89 Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 89 Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

2.1 Erythema and oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 90. Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 90.1

Comparison 90 Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 90 Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 90.2

Comparison 90 Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 90 Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

2.1 Erythema and oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 91. Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 91.1

Comparison 91 Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 91 Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 91.2

Comparison 91 Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 91 Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

2.1 Erythema and oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 92. Cryotherapy (weekly) versus ILMA (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 92.1

Comparison 92 Cryotherapy (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 92 Cryotherapy (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 93. Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 93.1

Comparison 93 Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks, Outcome 1 Lesions cured.

Comparison 93 Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks, Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 93.2

Comparison 93 Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 93 Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

2.1 Hypopigmentation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 94. Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 94.1

Comparison 94 Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 1 Lesions cured.

Comparison 94 Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 94.2

Comparison 94 Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 94 Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 2 Adverse effects.

2.1 Hypopigmentation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 95. Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 95.1

Comparison 95 Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 1 Lesions cured.

Comparison 95 Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

390

Risk Ratio (M‐H, Random, 95% CI)

14.64 [0.86, 247.99]

Analysis 95.2

Comparison 95 Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 95 Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 2 Adverse effects.

2.1 Hypopigmentation

1

390

Risk Ratio (M‐H, Random, 95% CI)

14.64 [0.86, 247.99]

Open in table viewer
Comparison 96. Cryotherapy (fortnightly) + 15% paromomycin + 10% urea cream (twice a day) + ILMA (twice a day for 4 weeks) versus ILMA (twice a week) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 96.1

Comparison 96 Cryotherapy (fortnightly) + 15% paromomycin + 10% urea cream (twice a day) + ILMA (twice a day for 4 weeks) versus ILMA (twice a week) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 96 Cryotherapy (fortnightly) + 15% paromomycin + 10% urea cream (twice a day) + ILMA (twice a day for 4 weeks) versus ILMA (twice a week) for up to 6 weeks, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 97. Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 97.1

Comparison 97 Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day), Outcome 1 Lesions cured.

Comparison 97 Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day), Outcome 1 Lesions cured.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 97.2

Comparison 97 Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day), Outcome 2 Adverse effects.

Comparison 97 Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day), Outcome 2 Adverse effects.

2.1 Mild skin symptoms

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 98. Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 98.1

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 1 Lesions cured.

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 1 Lesions cured.

2 Participants complete cure Show forest plot

2

499

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.97, 1.55]

Analysis 98.2

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 2 Participants complete cure.

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 2 Participants complete cure.

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 98.3

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 3 Adverse effects.

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 3 Adverse effects.

3.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 99. Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 99.1

Comparison 99 Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d), Outcome 1 Lesions cured.

Comparison 99 Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d), Outcome 1 Lesions cured.

2 Adverse effects (serious) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 99.2

Comparison 99 Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d), Outcome 2 Adverse effects (serious).

Comparison 99 Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d), Outcome 2 Adverse effects (serious).

Open in table viewer
Comparison 100. Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 100.1

Comparison 100 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 1 Participants complete cure.

Comparison 100 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 1 Participants complete cure.

2 Adverse event (secondary infection) Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 100.2

Comparison 100 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 2 Adverse event (secondary infection).

Comparison 100 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 2 Adverse event (secondary infection).

Open in table viewer
Comparison 101. Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 101.1

Comparison 101 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days), Outcome 1 Participants complete cure.

Comparison 101 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days), Outcome 1 Participants complete cure.

2 Adverse event (secondary infection) Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 101.2

Comparison 101 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days), Outcome 2 Adverse event (secondary infection).

Comparison 101 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days), Outcome 2 Adverse event (secondary infection).

Open in table viewer
Comparison 102. Radiofrequency waves versus ILSSG

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 102.1

Comparison 102 Radiofrequency waves versus ILSSG, Outcome 1 Participants complete cure.

Comparison 102 Radiofrequency waves versus ILSSG, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 103. Electrocauterisation + DAC n‐055 (daily) versus electrocauterisation

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 103.1

Comparison 103 Electrocauterisation + DAC n‐055 (daily) versus electrocauterisation, Outcome 1 Adverse effects.

Comparison 103 Electrocauterisation + DAC n‐055 (daily) versus electrocauterisation, Outcome 1 Adverse effects.

1.1 Superinfection

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Keloid formation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 104. PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 104.1

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 1 Lesions cured.

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 1 Lesions cured.

2 Scarring Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 104.2

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 2 Scarring.

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 2 Scarring.

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 104.3

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 3 Microbiological cure of skin lesions.

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 3 Microbiological cure of skin lesions.

Open in table viewer
Comparison 105. Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 105.1

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 105.2

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 2 Adverse effects.

2.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Development of cell‐mediated immunity Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 105.3

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 3 Development of cell‐mediated immunity.

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 3 Development of cell‐mediated immunity.

Open in table viewer
Comparison 106. Diminazene aceturate solution (weekly) versus cetrimide + chlorhexidine solution for 50 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 106.1

Comparison 106 Diminazene aceturate solution (weekly) versus cetrimide + chlorhexidine solution for 50 d, Outcome 1 Participants complete cure.

Comparison 106 Diminazene aceturate solution (weekly) versus cetrimide + chlorhexidine solution for 50 d, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 107. Topical garlic (twice a day) versus vehicle for 3 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 107.1

Comparison 107 Topical garlic (twice a day) versus vehicle for 3 weeks, Outcome 1 Participants complete cure.

Comparison 107 Topical garlic (twice a day) versus vehicle for 3 weeks, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 108. Topical herbal extract + placebo (5 d) versus IMMA (15‐20/mg/kg/d) + vehicle for 20 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 108.1

Comparison 108 Topical herbal extract + placebo (5 d) versus IMMA (15‐20/mg/kg/d) + vehicle for 20 d, Outcome 1 Participants complete cure.

Comparison 108 Topical herbal extract + placebo (5 d) versus IMMA (15‐20/mg/kg/d) + vehicle for 20 d, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 109. Topical honey (twice a day) + ILMA (weekly) versus ILMA (weekly) for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 109.1

Comparison 109 Topical honey (twice a day) + ILMA (weekly) versus ILMA (weekly) for 4 weeks, Outcome 1 Participants complete cure.

Comparison 109 Topical honey (twice a day) + ILMA (weekly) versus ILMA (weekly) for 4 weeks, Outcome 1 Participants complete cure.

Open in table viewer
Comparison 110. Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 110.1

Comparison 110 Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle, Outcome 1 Participants complete cure.

Comparison 110 Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 110.2

Comparison 110 Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle, Outcome 2 Adverse effects.

Comparison 110 Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle, Outcome 2 Adverse effects.

2.1 Itching

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 111. Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 111.1

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 1 Participants complete cure.

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 1 Participants complete cure.

2 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 111.2

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 2 Speed of healing (weeks).

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 2 Speed of healing (weeks).

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 111.3

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 3 Adverse effects.

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 3 Adverse effects.

3.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 112. Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 112.1

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 1 Participants complete cure.

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 1 Participants complete cure.

2 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 112.2

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 2 Speed of healing (weeks).

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 2 Speed of healing (weeks).

3 Adverse reaction Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 112.3

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 3 Adverse reaction.

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 3 Adverse reaction.

3.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 113. Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 113.1

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 1 Participants complete cure.

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 1 Participants complete cure.

2 Speed of healing (days) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 113.2

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 2 Speed of healing (days).

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 2 Speed of healing (days).

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 113.3

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 3 Adverse effects.

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 3 Adverse effects.

3.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 114. Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 114.1

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 1 Participants complete cure.

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 1 Participants complete cure.

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 114.2

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 2 Adverse effects.

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 2 Adverse effects.

2.1 Itching

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 114.3

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 3 Microbiological cure of skin lesions.

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 3 Microbiological cure of skin lesions.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgments about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

Risk of bias summary: review authors' judgments about each risk of bias item for each included study.

Risk of bias graph: review authors' judgments about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

Risk of bias graph: review authors' judgments about each risk of bias item presented as percentages across all included studies.

Forest Plot of Primary Outcome: Lesions Cured (Various comparisons)
Figuras y tablas -
Figure 4

Forest Plot of Primary Outcome: Lesions Cured (Various comparisons)

Forest plot of primary outcome: 1.2 Participants cured (Various comparisons)
Figuras y tablas -
Figure 5

Forest plot of primary outcome: 1.2 Participants cured (Various comparisons)

Comparison 1 ILMA weekly versus ILMA fortnightly for up to 8 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 1.1

Comparison 1 ILMA weekly versus ILMA fortnightly for up to 8 weeks, Outcome 1 Lesions cured.

Comparison 2 ILMA (every other day) versus IMMA (6 d/week) for up to 4 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 2.1

Comparison 2 ILMA (every other day) versus IMMA (6 d/week) for up to 4 weeks, Outcome 1 Lesions cured.

Comparison 3 IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (30 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 3.1

Comparison 3 IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (30 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.

Comparison 4 IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 4.1

Comparison 4 IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.

Comparison 5 IMMA (60 mg/kg/d for 3 weeks) + placebo versus IMMA (30 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 5.1

Comparison 5 IMMA (60 mg/kg/d for 3 weeks) + placebo versus IMMA (30 mg/kg/d for 3 weeks) + placebo, Outcome 1 Lesions cured.

Comparison 6 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 6.1

Comparison 6 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Comparison 6 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 6.2

Comparison 6 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 2 Adverse effects.

Comparison 7 IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 7.1

Comparison 7 IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Comparison 8 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 8.1

Comparison 8 IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Comparison 9 IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 9.1

Comparison 9 IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo, Outcome 1 Participants complete cure.

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 10.1

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 1 Lesions cured.

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 2 Adverse effects (itching and burning).
Figuras y tablas -
Analysis 10.2

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 2 Adverse effects (itching and burning).

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 3 Adverse effects (oedema).
Figuras y tablas -
Analysis 10.3

Comparison 10 ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 3 Adverse effects (oedema).

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 11.1

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 1 Lesions cured.

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 2 Adverse effects (itching and burning).
Figuras y tablas -
Analysis 11.2

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 2 Adverse effects (itching and burning).

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 3 Adverse effects (oedema).
Figuras y tablas -
Analysis 11.3

Comparison 11 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks), Outcome 3 Adverse effects (oedema).

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 12.1

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 1 Lesions cured.

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 2 Adverse effects (itching and burning).
Figuras y tablas -
Analysis 12.2

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 2 Adverse effects (itching and burning).

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 3 Adverse effects (oedema).
Figuras y tablas -
Analysis 12.3

Comparison 12 ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing, Outcome 3 Adverse effects (oedema).

Comparison 13 ILMA (weekly injections for 6 weeks) + gel mask twice a day versus ILMA (weekly injections for 6 weeks) + vehicle, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 13.1

Comparison 13 ILMA (weekly injections for 6 weeks) + gel mask twice a day versus ILMA (weekly injections for 6 weeks) + vehicle, Outcome 1 Participants complete cure.

Comparison 14 ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5) versus ILSSG (1000 mg/mL days 1, 3, 5), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 14.1

Comparison 14 ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5) versus ILSSG (1000 mg/mL days 1, 3, 5), Outcome 1 Lesions cured.

Comparison 15 ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 15.1

Comparison 15 ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 1 Participants complete cure.

Comparison 15 ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 2 Adverse effects (mild heart symptoms).
Figuras y tablas -
Analysis 15.2

Comparison 15 ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 2 Adverse effects (mild heart symptoms).

Comparison 16 Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 16.1

Comparison 16 Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks, Outcome 1 Participants complete cure.

Comparison 16 Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks, Outcome 2 Adverse effects (nausea and vomiting).
Figuras y tablas -
Analysis 16.2

Comparison 16 Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks, Outcome 2 Adverse effects (nausea and vomiting).

Comparison 17 Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections), Outcome 1 Participants cured.
Figuras y tablas -
Analysis 17.1

Comparison 17 Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections), Outcome 1 Participants cured.

Comparison 17 Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections), Outcome 2 Adverse effect (liver enzymes increase).
Figuras y tablas -
Analysis 17.2

Comparison 17 Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections), Outcome 2 Adverse effect (liver enzymes increase).

Comparison 18 ILSSG (100 mg/mL days 1, 3, 5) + oral ketoconazole (600 mg/d for 4 weeks) versus ILSSG (100 mg/mL days 1, 3, 5), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 18.1

Comparison 18 ILSSG (100 mg/mL days 1, 3, 5) + oral ketoconazole (600 mg/d for 4 weeks) versus ILSSG (100 mg/mL days 1, 3, 5), Outcome 1 Lesions cured.

Comparison 19 ILSSG (100 mg/mL days 1, 3, 5) + ketoconazole (600 mg/d for 4 weeks) versus ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 19.1

Comparison 19 ILSSG (100 mg/mL days 1, 3, 5) + ketoconazole (600 mg/d for 4 weeks) versus ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5), Outcome 1 Lesions cured.

Comparison 20 Itraconazole (200 mg for 6 weeks) versus placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 20.1

Comparison 20 Itraconazole (200 mg for 6 weeks) versus placebo, Outcome 1 Participants complete cure.

Comparison 21 Itraconazole (200 mg for 3 weeks) versus placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 21.1

Comparison 21 Itraconazole (200 mg for 3 weeks) versus placebo, Outcome 1 Participants complete cure.

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 22.1

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 1 Participants complete cure.

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 22.2

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 2 Adverse effects.

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 3 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 22.3

Comparison 22 Itraconazole (200 mg for 6 to 8 weeks) versus placebo, Outcome 3 Microbiological cure of skin lesions.

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 23.1

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 1 Participants complete cure.

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 2 Adverse effects (headache and dizziness).
Figuras y tablas -
Analysis 23.2

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 2 Adverse effects (headache and dizziness).

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 3 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 23.3

Comparison 23 Itraconazole (200 mg for 6 to 8 weeks) versus no treatment, Outcome 3 Microbiological cure of skin lesions.

Comparison 24 Fluconazole (200 mg for 6 weeks) versus placebo, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 24.1

Comparison 24 Fluconazole (200 mg for 6 weeks) versus placebo, Outcome 1 Lesions cured.

Comparison 24 Fluconazole (200 mg for 6 weeks) versus placebo, Outcome 2 Participants complete cure.
Figuras y tablas -
Analysis 24.2

Comparison 24 Fluconazole (200 mg for 6 weeks) versus placebo, Outcome 2 Participants complete cure.

Comparison 25 Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 25.1

Comparison 25 Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks), Outcome 1 Participants complete cure.

Comparison 25 Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 25.2

Comparison 25 Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks), Outcome 2 Adverse effects.

Comparison 26 Oral dapsone (200 mg/d for 6 weeks) versus placebo, Outcome 1 Participants complete Cure.
Figuras y tablas -
Analysis 26.1

Comparison 26 Oral dapsone (200 mg/d for 6 weeks) versus placebo, Outcome 1 Participants complete Cure.

Comparison 26 Oral dapsone (200 mg/d for 6 weeks) versus placebo, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 26.2

Comparison 26 Oral dapsone (200 mg/d for 6 weeks) versus placebo, Outcome 2 Adverse effects.

Comparison 27 Allopurinol (15 mg/kg/d for 3 weeks) + IMMA (20 mg/kg/d for 2 weeks) versus allopurinol (15 mg/kg/d for 3 weeks), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 27.1

Comparison 27 Allopurinol (15 mg/kg/d for 3 weeks) + IMMA (20 mg/kg/d for 2 weeks) versus allopurinol (15 mg/kg/d for 3 weeks), Outcome 1 Lesions cured.

Comparison 28 Allopurinol (15mg/kg/d for 3 weeks)+ IMMA (20 mg/kg/d for 2 weeks) versus IMMA (20 mg/kg/d for 2 weeks), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 28.1

Comparison 28 Allopurinol (15mg/kg/d for 3 weeks)+ IMMA (20 mg/kg/d for 2 weeks) versus IMMA (20 mg/kg/d for 2 weeks), Outcome 1 Lesions cured.

Comparison 29 Allopurinol (15 mg/kg/d for 3 weeks) versus IMMA (20 mg/kg/d for 2 weeks), Outcome 1 Lesions Cured.
Figuras y tablas -
Analysis 29.1

Comparison 29 Allopurinol (15 mg/kg/d for 3 weeks) versus IMMA (20 mg/kg/d for 2 weeks), Outcome 1 Lesions Cured.

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 30.1

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 1 Lesions cured.

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 30.2

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 2 Adverse effects.

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 3 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 30.3

Comparison 30 Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d), Outcome 3 Microbiological cure of skin lesions.

Comparison 31 Allopurinol (20 mg/kg/d for 3 weeks)+ IMMA (10 mg/kg/d for 20 d) versus IMMA (20 mg/kg/d for 28 d), Outcome 1 Adverse effects.
Figuras y tablas -
Analysis 31.1

Comparison 31 Allopurinol (20 mg/kg/d for 3 weeks)+ IMMA (10 mg/kg/d for 20 d) versus IMMA (20 mg/kg/d for 28 d), Outcome 1 Adverse effects.

Comparison 32 Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d), Outcome 1 Participants complete cured.
Figuras y tablas -
Analysis 32.1

Comparison 32 Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d), Outcome 1 Participants complete cured.

Comparison 32 Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 32.2

Comparison 32 Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d), Outcome 2 Adverse effects.

Comparison 33 Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 33.1

Comparison 33 Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo, Outcome 1 Participants complete cure.

Comparison 33 Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo, Outcome 2 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 33.2

Comparison 33 Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo, Outcome 2 Microbiological cure of skin lesions.

Comparison 34 Oral rifampicin (10 mg/kg/d) + omeprazole (20 mg/d) for 6 weeks versus placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 34.1

Comparison 34 Oral rifampicin (10 mg/kg/d) + omeprazole (20 mg/d) for 6 weeks versus placebo, Outcome 1 Participants complete cure.

Comparison 35 Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 35.1

Comparison 35 Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d), Outcome 1 Lesions cured.

Comparison 35 Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 35.2

Comparison 35 Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d), Outcome 2 Adverse effects.

Comparison 36 Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 36.1

Comparison 36 Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d), Outcome 1 Participants complete cure.

Comparison 36 Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 36.2

Comparison 36 Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d), Outcome 2 Adverse effects.

Comparison 37 Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 37.1

Comparison 37 Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d, Outcome 1 Participants complete cure.

Comparison 37 Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 37.2

Comparison 37 Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d, Outcome 2 Adverse effects.

Comparison 38 Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 38.1

Comparison 38 Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks), Outcome 1 Participants complete cure.

Comparison 39 Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 39.1

Comparison 39 Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks), Outcome 1 Participants complete cure.

Comparison 40 Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 40.1

Comparison 40 Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d, Outcome 1 Participants complete cure.

Comparison 40 Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 40.2

Comparison 40 Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d, Outcome 2 Adverse effects.

Comparison 41 Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 41.1

Comparison 41 Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 1 Participants complete cure.

Comparison 41 Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 41.2

Comparison 41 Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 2 Adverse effects.

Comparison 42 Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 42.1

Comparison 42 Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 1 Participants complete cure.

Comparison 42 Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 42.2

Comparison 42 Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d, Outcome 2 Adverse effects.

Comparison 43 Oral zinc sulphate (10 mg/kg/d for 45 d) versus IMMA (20 mg/kg/d for 20 d), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 43.1

Comparison 43 Oral zinc sulphate (10 mg/kg/d for 45 d) versus IMMA (20 mg/kg/d for 20 d), Outcome 1 Participants complete cure.

Comparison 44 Artesunate 400 mg + sulphamethoxypyrazine/pyrimethamine 1000 mg/50 mg 4 times daily for 4 d versus placebo, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 44.1

Comparison 44 Artesunate 400 mg + sulphamethoxypyrazine/pyrimethamine 1000 mg/50 mg 4 times daily for 4 d versus placebo, Outcome 1 Participants complete cure.

Comparison 45 Topical 2% miconazole (twice a day) versus topical 1% clotrimazole (twice a day) for 30 d, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 45.1

Comparison 45 Topical 2% miconazole (twice a day) versus topical 1% clotrimazole (twice a day) for 30 d, Outcome 1 Lesions cured.

Comparison 46 Topical ketoconazole (twice a day) versus vehicle (twice a day) for 30 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 46.1

Comparison 46 Topical ketoconazole (twice a day) versus vehicle (twice a day) for 30 d, Outcome 1 Participants complete cure.

Comparison 47 Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks, Outcome 1 Participants complete cure (ITT).
Figuras y tablas -
Analysis 47.1

Comparison 47 Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks, Outcome 1 Participants complete cure (ITT).

Comparison 47 Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 47.2

Comparison 47 Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks, Outcome 2 Adverse effects.

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 48.1

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 1 Lesions cured.

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 2 Scarring.
Figuras y tablas -
Analysis 48.2

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 2 Scarring.

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 3 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 48.3

Comparison 48 Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d), Outcome 3 Microbiological cure of skin lesions.

Comparison 49 Paromomycin (twice daily for 30 d) versus vehicle (twice daily for 30 d), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 49.1

Comparison 49 Paromomycin (twice daily for 30 d) versus vehicle (twice daily for 30 d), Outcome 1 Lesions cured.

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 50.1

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 1 Participants complete cure.

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 50.2

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 2 Adverse effects.

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 3 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 50.3

Comparison 50 Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d), Outcome 3 Microbiological cure of skin lesions.

Comparison 51 Paromomycin 15% (daily for 20 d) versus vehicle, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 51.1

Comparison 51 Paromomycin 15% (daily for 20 d) versus vehicle, Outcome 1 Participants complete cure.

Comparison 52 Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus vehicle, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 52.1

Comparison 52 Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus vehicle, Outcome 1 Participants complete cure.

Comparison 53 Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus paromomycin 15% alone (daily for 20 d), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 53.1

Comparison 53 Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus paromomycin 15% alone (daily for 20 d), Outcome 1 Participants complete cure.

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 54.1

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 1 Participants complete cure.

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 2 Recurrence.
Figuras y tablas -
Analysis 54.2

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 2 Recurrence.

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 3 Scarring.
Figuras y tablas -
Analysis 54.3

Comparison 54 Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months), Outcome 3 Scarring.

Comparison 55 Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 55.1

Comparison 55 Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d), Outcome 1 Participants complete cure.

Comparison 55 Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 55.2

Comparison 55 Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d), Outcome 2 Adverse effects.

Comparison 56 Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 56.1

Comparison 56 Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d), Outcome 1 Participants complete cure.

Comparison 56 Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d), Outcome 2 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 56.2

Comparison 56 Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d), Outcome 2 Microbiological cure of skin lesions.

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 57.1

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 1 Lesions cured.

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 2 Scarring.
Figuras y tablas -
Analysis 57.2

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 2 Scarring.

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 3 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 57.3

Comparison 57 Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks), Outcome 3 Microbiological cure of skin lesions.

Comparison 58 Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 58.1

Comparison 58 Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks), Outcome 1 Participants complete cure.

Comparison 58 Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks), Outcome 2 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 58.2

Comparison 58 Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks), Outcome 2 Microbiological cure of skin lesions.

Comparison 59 IL zinc 2% (twice a week for 2 weeks) versus ILSSG (100 mg/mL) for 2 weeks), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 59.1

Comparison 59 IL zinc 2% (twice a week for 2 weeks) versus ILSSG (100 mg/mL) for 2 weeks), Outcome 1 Lesions cured.

Comparison 60 IL zinc 2% (twice a week for 2 weeks) versus IL 7% HSCS for 2 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 60.1

Comparison 60 IL zinc 2% (twice a week for 2 weeks) versus IL 7% HSCS for 2 weeks, Outcome 1 Lesions cured.

Comparison 61 ILSSG (100 mg/mL) for 2 weeks versus IL 7% HSCS for 2 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 61.1

Comparison 61 ILSSG (100 mg/mL) for 2 weeks versus IL 7% HSCS for 2 weeks, Outcome 1 Lesions cured.

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 62.1

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 1 Lesions cured.

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 2 Participants complete cured.
Figuras y tablas -
Analysis 62.2

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 2 Participants complete cured.

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 3 Adverse effects.
Figuras y tablas -
Analysis 62.3

Comparison 62 IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks), Outcome 3 Adverse effects.

Comparison 63 IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 63.1

Comparison 63 IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks), Outcome 1 Lesions cured.

Comparison 63 IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 63.2

Comparison 63 IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks), Outcome 2 Adverse effects.

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 64.1

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 1 Participants complete cure.

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 2 Participants with treated lesions that recur.
Figuras y tablas -
Analysis 64.2

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 2 Participants with treated lesions that recur.

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 3 Adverse effects.
Figuras y tablas -
Analysis 64.3

Comparison 64 Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA, Outcome 3 Adverse effects.

Comparison 65 IL 7% HSCS (0.2 mL to 7 mL per lesion) versus ILSSG (max 2 mL) max 5 injections, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 65.1

Comparison 65 IL 7% HSCS (0.2 mL to 7 mL per lesion) versus ILSSG (max 2 mL) max 5 injections, Outcome 1 Lesions cured.

Comparison 66 IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 66.1

Comparison 66 IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks, Outcome 1 Lesions cured.

Comparison 66 IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 66.2

Comparison 66 IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks, Outcome 2 Adverse effects.

Comparison 67 IL 7% HSCS (0.1 mL to 0.5 mL per lesion) versus IL 2% ciprofloxacin solution (0.1 mL to 0.5 mL per lesion), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 67.1

Comparison 67 IL 7% HSCS (0.1 mL to 0.5 mL per lesion) versus IL 2% ciprofloxacin solution (0.1 mL to 0.5 mL per lesion), Outcome 1 Lesions cured.

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 68.1

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.
Figuras y tablas -
Analysis 68.2

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).
Figuras y tablas -
Analysis 68.3

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.
Figuras y tablas -
Analysis 68.4

Comparison 68 IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 69.1

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.
Figuras y tablas -
Analysis 69.2

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).
Figuras y tablas -
Analysis 69.3

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.
Figuras y tablas -
Analysis 69.4

Comparison 69 ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 70.1

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 1 Lesions cured.

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.
Figuras y tablas -
Analysis 70.2

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 2 Recurrence.

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).
Figuras y tablas -
Analysis 70.3

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 3 Speed of healing (weeks).

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.
Figuras y tablas -
Analysis 70.4

Comparison 70 ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion), Outcome 4 Adverse effects.

Comparison 71 IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 71.1

Comparison 71 IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks, Outcome 1 Lesions cured.

Comparison 71 IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks, Outcome 2 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 71.2

Comparison 71 IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks, Outcome 2 Microbiological cure of skin lesions.

Comparison 72 WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 72.1

Comparison 72 WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d), Outcome 1 Participants complete cure.

Comparison 72 WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 72.2

Comparison 72 WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d), Outcome 2 Adverse effects.

Comparison 73 IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 73.1

Comparison 73 IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks, Outcome 1 Participants complete cure.

Comparison 73 IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 73.2

Comparison 73 IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks, Outcome 2 Adverse effects.

Comparison 74 Topical miltefosine 6% (once daily) versus ILMA (twice a week) for up to 28 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 74.1

Comparison 74 Topical miltefosine 6% (once daily) versus ILMA (twice a week) for up to 28 d, Outcome 1 Participants complete cure.

Comparison 75 Dapsone gel 5% (twice a day) + ILMA (weekly) versus cryotherapy (every 2 weeks) + IMMA (weekly) for up to 16 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 75.1

Comparison 75 Dapsone gel 5% (twice a day) + ILMA (weekly) versus cryotherapy (every 2 weeks) + IMMA (weekly) for up to 16 weeks, Outcome 1 Lesions cured.

Comparison 76 DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 76.1

Comparison 76 DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d, Outcome 1 Participants complete cure.

Comparison 76 DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 76.2

Comparison 76 DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d, Outcome 2 Adverse effects.

Comparison 77 DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 77.1

Comparison 77 DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 1 Participants complete cure.

Comparison 77 DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 77.2

Comparison 77 DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 2 Adverse effects.

Comparison 78 DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 78.1

Comparison 78 DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 1 Participants complete cure.

Comparison 78 DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 78.2

Comparison 78 DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d, Outcome 2 Adverse effects.

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 79.1

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 1 Lesions cured.

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 2 Recurrence.
Figuras y tablas -
Analysis 79.2

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 2 Recurrence.

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 3 Adverse effects.
Figuras y tablas -
Analysis 79.3

Comparison 79 Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks, Outcome 3 Adverse effects.

Comparison 80 CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 80.1

Comparison 80 CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d, Outcome 1 Lesions cured.

Comparison 80 CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 80.2

Comparison 80 CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d, Outcome 2 Adverse effects.

Comparison 81 CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 81.1

Comparison 81 CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks, Outcome 1 Lesions cured.

Comparison 81 CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 81.2

Comparison 81 CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks, Outcome 2 Adverse effects.

Comparison 82 Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser, Outcome 1 Partcipants complete cure.
Figuras y tablas -
Analysis 82.1

Comparison 82 Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser, Outcome 1 Partcipants complete cure.

Comparison 82 Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 82.2

Comparison 82 Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser, Outcome 2 Adverse effects.

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 83.1

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 1 Participants complete cure.

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 2 Recurrence.
Figuras y tablas -
Analysis 83.2

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 2 Recurrence.

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 3 Adverse effects.
Figuras y tablas -
Analysis 83.3

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 3 Adverse effects.

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 4 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 83.4

Comparison 83 TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks), Outcome 4 Microbiological cure of skin lesions.

Comparison 84 Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 84.1

Comparison 84 Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks, Outcome 1 Participants complete cure.

Comparison 84 Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks, Outcome 2 Lesions cured.
Figuras y tablas -
Analysis 84.2

Comparison 84 Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks, Outcome 2 Lesions cured.

Comparison 85 TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 85.1

Comparison 85 TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks), Outcome 1 Participants complete cure.

Comparison 85 TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks), Outcome 2 Speed of healing (weeks).
Figuras y tablas -
Analysis 85.2

Comparison 85 TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks), Outcome 2 Speed of healing (weeks).

Comparison 86 Fractional laser + ILMA (fortnightly 2 sessions) versus ILMA alone (twice a week for up to 8 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 86.1

Comparison 86 Fractional laser + ILMA (fortnightly 2 sessions) versus ILMA alone (twice a week for up to 8 weeks), Outcome 1 Participants complete cure.

Comparison 87 TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 87.1

Comparison 87 TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions), Outcome 1 Participants complete cure.

Comparison 87 TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions), Outcome 2 Speed of healing (weeks).
Figuras y tablas -
Analysis 87.2

Comparison 87 TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions), Outcome 2 Speed of healing (weeks).

Comparison 88 TCA fortnightly up to 8 weeks + ILMA (twice a week) versus ILMA alone (weekly for up to 8 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 88.1

Comparison 88 TCA fortnightly up to 8 weeks + ILMA (twice a week) versus ILMA alone (weekly for up to 8 weeks), Outcome 1 Participants complete cure.

Comparison 89 Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 89.1

Comparison 89 Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 89 Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 89.2

Comparison 89 Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 90 Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 90.1

Comparison 90 Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 90 Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 90.2

Comparison 90 Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 91 Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 91.1

Comparison 91 Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 91 Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 91.2

Comparison 91 Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 92 Cryotherapy (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 92.1

Comparison 92 Cryotherapy (weekly) versus ILMA (weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 93 Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 93.1

Comparison 93 Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks, Outcome 1 Lesions cured.

Comparison 93 Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 93.2

Comparison 93 Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 94 Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 94.1

Comparison 94 Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 1 Lesions cured.

Comparison 94 Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 94.2

Comparison 94 Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 95 Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 95.1

Comparison 95 Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 1 Lesions cured.

Comparison 95 Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 95.2

Comparison 95 Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 96 Cryotherapy (fortnightly) + 15% paromomycin + 10% urea cream (twice a day) + ILMA (twice a day for 4 weeks) versus ILMA (twice a week) for up to 6 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 96.1

Comparison 96 Cryotherapy (fortnightly) + 15% paromomycin + 10% urea cream (twice a day) + ILMA (twice a day for 4 weeks) versus ILMA (twice a week) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 97 Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 97.1

Comparison 97 Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day), Outcome 1 Lesions cured.

Comparison 97 Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day), Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 97.2

Comparison 97 Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day), Outcome 2 Adverse effects.

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 98.1

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 1 Lesions cured.

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 2 Participants complete cure.
Figuras y tablas -
Analysis 98.2

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 2 Participants complete cure.

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 3 Adverse effects.
Figuras y tablas -
Analysis 98.3

Comparison 98 Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks, Outcome 3 Adverse effects.

Comparison 99 Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 99.1

Comparison 99 Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d), Outcome 1 Lesions cured.

Comparison 99 Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d), Outcome 2 Adverse effects (serious).
Figuras y tablas -
Analysis 99.2

Comparison 99 Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d), Outcome 2 Adverse effects (serious).

Comparison 100 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 100.1

Comparison 100 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 1 Participants complete cure.

Comparison 100 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 2 Adverse event (secondary infection).
Figuras y tablas -
Analysis 100.2

Comparison 100 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks), Outcome 2 Adverse event (secondary infection).

Comparison 101 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days), Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 101.1

Comparison 101 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days), Outcome 1 Participants complete cure.

Comparison 101 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days), Outcome 2 Adverse event (secondary infection).
Figuras y tablas -
Analysis 101.2

Comparison 101 Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days), Outcome 2 Adverse event (secondary infection).

Comparison 102 Radiofrequency waves versus ILSSG, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 102.1

Comparison 102 Radiofrequency waves versus ILSSG, Outcome 1 Participants complete cure.

Comparison 103 Electrocauterisation + DAC n‐055 (daily) versus electrocauterisation, Outcome 1 Adverse effects.
Figuras y tablas -
Analysis 103.1

Comparison 103 Electrocauterisation + DAC n‐055 (daily) versus electrocauterisation, Outcome 1 Adverse effects.

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 1 Lesions cured.
Figuras y tablas -
Analysis 104.1

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 1 Lesions cured.

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 2 Scarring.
Figuras y tablas -
Analysis 104.2

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 2 Scarring.

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 3 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 104.3

Comparison 104 PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks), Outcome 3 Microbiological cure of skin lesions.

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 105.1

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 1 Participants complete cure.

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 105.2

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 2 Adverse effects.

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 3 Development of cell‐mediated immunity.
Figuras y tablas -
Analysis 105.3

Comparison 105 Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks, Outcome 3 Development of cell‐mediated immunity.

Comparison 106 Diminazene aceturate solution (weekly) versus cetrimide + chlorhexidine solution for 50 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 106.1

Comparison 106 Diminazene aceturate solution (weekly) versus cetrimide + chlorhexidine solution for 50 d, Outcome 1 Participants complete cure.

Comparison 107 Topical garlic (twice a day) versus vehicle for 3 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 107.1

Comparison 107 Topical garlic (twice a day) versus vehicle for 3 weeks, Outcome 1 Participants complete cure.

Comparison 108 Topical herbal extract + placebo (5 d) versus IMMA (15‐20/mg/kg/d) + vehicle for 20 d, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 108.1

Comparison 108 Topical herbal extract + placebo (5 d) versus IMMA (15‐20/mg/kg/d) + vehicle for 20 d, Outcome 1 Participants complete cure.

Comparison 109 Topical honey (twice a day) + ILMA (weekly) versus ILMA (weekly) for 4 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 109.1

Comparison 109 Topical honey (twice a day) + ILMA (weekly) versus ILMA (weekly) for 4 weeks, Outcome 1 Participants complete cure.

Comparison 110 Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 110.1

Comparison 110 Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle, Outcome 1 Participants complete cure.

Comparison 110 Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 110.2

Comparison 110 Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle, Outcome 2 Adverse effects.

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 111.1

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 1 Participants complete cure.

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 2 Speed of healing (weeks).
Figuras y tablas -
Analysis 111.2

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 2 Speed of healing (weeks).

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 3 Adverse effects.
Figuras y tablas -
Analysis 111.3

Comparison 111 Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 3 Adverse effects.

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 112.1

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 1 Participants complete cure.

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 2 Speed of healing (weeks).
Figuras y tablas -
Analysis 112.2

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 2 Speed of healing (weeks).

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 3 Adverse reaction.
Figuras y tablas -
Analysis 112.3

Comparison 112 Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks, Outcome 3 Adverse reaction.

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 113.1

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 1 Participants complete cure.

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 2 Speed of healing (days).
Figuras y tablas -
Analysis 113.2

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 2 Speed of healing (days).

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 3 Adverse effects.
Figuras y tablas -
Analysis 113.3

Comparison 113 Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks, Outcome 3 Adverse effects.

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 1 Participants complete cure.
Figuras y tablas -
Analysis 114.1

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 1 Participants complete cure.

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 2 Adverse effects.
Figuras y tablas -
Analysis 114.2

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 2 Adverse effects.

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 3 Microbiological cure of skin lesions.
Figuras y tablas -
Analysis 114.3

Comparison 114 Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks, Outcome 3 Microbiological cure of skin lesions.

Summary of findings for the main comparison. Itraconazole (200 mg for 6 to 8 weeks) versus placebo for Old World cutaneous leishmaniasis

Itraconazole (200 mg for 6‐8 weeks) versus placebo for Old World cutaneous leishmaniasis

Patient or population: patients with Old World cutaneous leishmaniasis
Settings: Kuwait, India, and Iran
Intervention: itraconazole (200 mg for 6‐8 weeks)
Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo

Itraconazole (200 mg for 6‐8 weeks)

Percentage of lesions cured after the end of treatment

Not measured in this comparison

Percentage of participants with complete cure
Follow‐up: mean 2.5 months

Study population

RR 3.70
(0.35 to 38.99)

244
(3 studies)

⊕⊝⊝⊝
Very lowa

454 per 1000

1000 per 1000
(159 to 1000)

Moderate

100 per 1000

370 per 1000
(35 to 1000)

Adverse effects

Mild abdominal pain and nausea

Adverse effects

Mild abnormal liver function

40 per 1000

0 per 1000

95 per 1000
(30 to 302)

0 per 1000
(0 to 0)

RR 2.36
(0.74 to 7.47)

RR 3.08
(0.53 to 17.98)

204
(3 studies)

84
(3 studies)

⊕⊝⊝⊝
Very lowb

⊕⊝⊝⊝
Very lowc

Speed of healing (time taken to be 'cured')

Neither of the studies reported speed of healing (time taken to be 'cured') in this comparison.

Microbiological or histopathological cure of skin lesions
Follow‐up: mean 2.5 months

Not estimable

Not estimable

RR 17.00
(0.47 to 612.21)

20
(1 study)

⊕⊝⊝⊝
Very lowd

There were zero events in the placebo group

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio.

GRADE Working Group grades of evidence
High quality/certainty: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality/certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality/certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality/certainty: we are very uncertain about the estimate.

aDowngraded by 4 levels due to: risk of bias (2 RCTs have many uncertain items), inconsistency (there is considerable heterogeneity ‐ I² = 73%), and imprecision (2 levels due to wide 95% confidence intervals, crossing the line of no effect).
bDowngraded by 3 levels due to: risk of bias (many uncertain items in the risk of bias judgment), and imprecision (2 levels due to wide 95% confidence intervals, crossing the line of no effect).
cDowngraded by 3 levels due to: risk of bias (many uncertain items in the risk of bias judgment), and imprecision (2 levels due to wide 95% confidence intervals, crossing the line of no effect).
dDowngraded by 3 levels due to: risk of bias (many uncertain items in the risk of bias judgment), and imprecision (2 levels due to wide 95% confidence intervals; this outcome is only reported for one study involving 20 participants).

Figuras y tablas -
Summary of findings for the main comparison. Itraconazole (200 mg for 6 to 8 weeks) versus placebo for Old World cutaneous leishmaniasis
Summary of findings 2. Paromomycin ointment versus vehicle for Old World cutaneous leishmaniasis

Paromomycin ointment versus matched vehicle for Old World cutaneous leishmaniasis

Patient or population: patients with Old World cutaneous leishmaniasis
Settings: primary health centres, Iran and Tunisia
Intervention: paromomycin ointment (15% + 10% urea) twice daily for 14 days
Comparison: vehicle

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Certainty of the evidence
(GRADE)

Assumed risk

Corresponding risk

Vehicle

Paromomycin ointment (15% + 10% urea) twice daily for 14 days

Percentage of lesions cured after the end of treatment

Not measured in this comparison

Percentage of participants with complete cure
Follow‐up: mean 2.5 months

Study population

RR 1.00
(0.86 to 1.17)

383
(2 studies)

⊕⊝⊝⊝
Very lowa

623 per 1000

623 per 1000
(536 to 729)

Moderate

619 per 1000

619 per 1000
(532 to 724)

Adverse effects Skin/local reactions

Study population

RR 1.42
(0.67 to 3.01)

713
(4 studies)

⊕⊝⊝⊝
Very lowb

96 per 1000

136 per 1000
(64 to 287)

Moderate

90 per 1000

128 per 1000
(60 to 271)

Speed of healing (time taken to be 'cured')

Not measured in this comparison

Microbiological or histopathological cure of skin lesions
Follow‐up: mean 2.5 months

Study population

RR 1.03
(0.88 to 1.2)

383
(2 studies)

⊕⊝⊝⊝
Very lowc

859 per 1000

884 per 1000
(756 to 1000)

Moderate

792 per 1000

816 per 1000
(697 to 950)

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; N/A: not applicable.

GRADE Working Group grades of evidence
High quality/certainty: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality/certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality/certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality/certainty: we are very uncertain about the estimate.

aDowngraded by 4 levels due to risk of bias (1 RCT has many uncertain risks), indirectness (2 levels because one of the studies focused on young people), and imprecision (the confidence interval around the estimate risk ratio ranges from a 14% reduction to a 17% increase in the risk ratio for healing with paromomycin).
bDowngraded by 4 levels due to risk of bias (2 RCTs have many uncertain risks), indirectness (2 levels because one of the studies focused on young people), and imprecision (the confidence interval crosses the line of no effect).
cDowngraded by 5 levels due to risk of bias (1 RCT has many uncertain risks), inconsistency (there is considerable heterogeneity ‐ I² = 84%), indirectness (2 levels because one of the studies focused on young people), and imprecision (the confidence interval around the estimate risk ratio ranges from a 12% reduction to a 20% increase in the risk ratio for healing with paromomycin).

Figuras y tablas -
Summary of findings 2. Paromomycin ointment versus vehicle for Old World cutaneous leishmaniasis
Table 1. Glossary

Term

Definition

Antimonials

Pharmaceutical agents containing antimony. Antimony‐containing compounds (meglumine antimoniate and sodium stibogluconate) are the principal medications used to treat leishmaniases, an infection caused by a protozoan parasite.

Arthralgia

Pain in the joints. The causes of arthralgia are varied and range, from a joints perspective, from degenerative and destructive processes such as osteoarthritis and sports injuries to inflammation of tissues surrounding the joints, such as bursitis.

Cardiac arrhythmia

An arrhythmia is an abnormal heart rhythm. Many types of arrhythmia have no symptoms. When symptoms are present these may include palpitations or feeling a pause between heartbeats. More seriously there may be lightheadedness, passing out, shortness of breath, or chest pain.

Cutaneous necrosis

The death of living tissues in response to disease or injury.

Cytolysis

The degeneration or dissolution of cell caused by the disruption of cell membrane.

Exudate

A fluid with a high content of protein and cellular debris that has escaped from blood vessels and has been deposited in tissues or on tissue surfaces, usually as a result of inflammation.

Human monocytes

Monocytes are the biggest type of white blood cell in the immune system. Originally formed in the bone marrow, they are released into our blood and migrate into the connective tissue where they differentiate into macrophages. When certain germs enter the body, they quickly rush to the site of attack.

Hypotension

A systolic blood pressure reading (the top number) of 90 millimetres of mercury (mmHg) or less a diastolic blood pressure reading (the bottom number) of 60 mmHg or less is generally considered low blood pressure. The causes of low blood pressure can range from dehydration to serious medical or surgical disorders.

Immune response modifier

Any of a broad family of biomolecules that up‐ or down‐regulate, or restore immune responsiveness, which are generated after T cells recognise an antigen present on the surface of a self‐antigen‐presenting cell, which, once activated, produce multiple cytokines.

Immunolabeling

A biochemical process that enables the detection and localisation of an antigen to a particular site within a cell, tissue, or organ. Antigens are organic molecules, usually proteins, capable of binding to an antibody. These antigens can be visualised using a combination of antigen‐specific antibodies as well as a means of detection, called a tag, that is covalently linked to the antibody. If the immunolabeling process is meant to reveal information about a cell or its substructures, the process is called immunocytochemistry. Immunolabeling of larger structures is called immunohistochemistry.

In vitro

Biological processes or reactions made to occur outside the living organism in an artificial environment, such as a culture medium.

Intralesional meglumine antimoniate

Meglumine antimoniate (or Glucantime) is a medicine used for treating leishmaniasis. It belongs to a group of compounds known as the pentavalent antimonials.

Lymphadenopathies

Lymph nodes that have an abnormal in size, number or consistency; often used as a synonym for swollen or enlarged lymph nodes. Common causes of lymphadenopathy are infection, autoimmune disease, or malignancy.

Lymphatic channels

The vessels that transport lymph throughout the body. Lymph is a clear fluid that contains cells important for forming antibodies that fight infection.

Lymphokine

Any of various soluble protein mediators released by sensitised lymphocytes on contact with antigen, and believed to play a role in macrophage activation, lymphocyte transformation, and cell‐mediated immunity. They regulate immune responses through differentiation, amplification, and inhibition of cell functions. Lymphokines may also have a cytotoxic effector function. Used as biologic response modifiers in the treatment of cancer.

Macrophages

White blood cells (activated monocytes) that protect the body against infection and foreign substances by breaking them down into antigenic peptides recognised by circulating T cells.

Miltefosine

An oral alkyl phosphocholine analogue used to treat cutaneous and visceral leishmaniasis. Interacts with lipids and sterols in the Leishmania membrane resulting in inhibition of mitochondria and apoptotic cell death.

Mucous membranes

The mucous membranes (or mucosae or mucosas; singular mucosa) are linings of mostly endodermal origin, covered in epithelium, which are involved in absorption and secretion. They line cavities that are exposed to the external environment and internal organs.

Myalgia

Myalgia, or muscle pain, is a symptom of many diseases and disorders. The most common causes are the overuse or over‐stretching of a muscle or group of muscles. Myalgia without a traumatic history is often due to viral infections. Long‐term myalgias may be indicative of a metabolic myopathy, some nutritional deficiencies or chronic fatigue syndrome.

Nodular lymphangitis

Nodular lymphangitis is a distinct clinical entity, separate from lymphangitis. This disorder is characterised by inflammatory nodules along the lymphatics draining a primary skin infection

Papule

A solid, rounded growth that is elevated from the skin, usually inflammatory but nonsuppurative. A papule is usually less than 1 cm across.

Parenteral

Administration of a medicinal or therapeutic substance, other than through the gastrointestinal or respiratory tracts, e.g. by intravenous, intramuscular or subcuticular injection.

Pentamidine

Pentamidine (e.g. isethionate) is an antiprotozoal and antifungal agent of the class of aromatic diamidines, administered intravenously or intramuscularly in treatment of early African trypanosomiasis and leishmaniasis, and intravenously, intramuscularly, or by oral inhalation in treatment and prophylaxis of Pneumocystis carinii pneumonia.

Pentavalent antimony

Pentavalent antimonials are a group of compounds used for the treatment of leishmaniasis. The first pentavalent antimonial used was urea stibamine: first introduced in the 1930s, it fell out of favour in the 1950s due to higher toxicity compared to sodium stibogluconate. The compounds currently available for clinical use are: sodium stibogluconate (Pentostam; manufactured by GlaxoSmithKline; available in the USA and UK), which is administered by slow intravenous injection, intralesional or intramuscular injection, and meglumine antimoniate (Glucantime; manufactured by Aventis; available in Brazil, France and Italy), which is administered by intramuscular, intralesional, or intravenous injection.

Promastigotes

Term now generally used instead of 'leptomonad' or 'leptomonad stage' to avoid confusion with the flagellate genus Leptomonas. It denotes the flagellate stage of a trypanosomatid protozoan in which the flagellum arises from a kinetoplast in front of the nucleus and emerges from the anterior end of the organism; usually an extracellular phase, as in the insect intermediate host (or in culture) of Leishmania parasites.

Protozoan

Any of a group of single‐celled, usually microscopic, eukaryotic organisms, such as amoebas, ciliates, flagellates, and sporozoans.

ThermoMed device

The ThermoMed is a battery‐operated device that delivers precisely controlled localised current field radiofrequency heat to selectively destroy certain diseased tissue and is recommended by the World Health Organization as an alternative therapy for cutaneus leishmaniasis.

Thermotherapy

The treatment of disease by the application of heat. Thermotherapy may be administered as dry heat with heat lamps, diathermy machines, electric pads, or hot water bottles or as moist heat with warm compresses or immersion in warm water. Warm soaks or compresses may be used to treat local infections, relax muscles and relieve pain in patients with motor problems, and promote circulation in peripheral vascular disorders such as thrombophlebitis.

Figuras y tablas -
Table 1. Glossary
Table 2. Interventions for Old World cutaneous leishmaniasis

Drug

Doses

Systemic antimonials

Sodium stibogluconate (Pentostam, Stibanate)

Meglumine antimonate (Glucantime)

Combined with pentoxifylline

20 mgSb v+/kg/d intramuscularly or intravenously for 20‐30 days

400 mg orally 3 times a day for 10–20 days

Intralesional antimonials

Sodium stibogluconate (Pentostam, Stibanate)

Meglumine antimonate (Glucantime)

1–5 mL per session every 3–7 days. Up to 10 sessions depending on the clinical response, but most patients require ≤ 5 sessions

Non‐antimonial systemic treatments

Fluconazole

200 mg orally daily for 6 weeks

Miltefosine

50 mg orally three times daily for 28 days

Liposomal amphotericin B

3 mg/kg/d IV on days 1‐5 and 10 (18 mg/kg total dose)

Non‐antimonial topical or intralesional therapies

15% paromomycin/12% methylbenzethonium chloride

Ointment twice daily for 10‐20 days

15% paromomycin/0.5% gentamicin sulphate

Twice a day for 20 days

Physical therapies

Cryotherapy with liquid nitrogen

Frozen for 10‐30 s and thaw applied locally 2‐3 times in each session, repeated every 1‐4 weeks to complete healing (usually 2‐4 sessions)

Local heat therapy

50°‐55ºC for 30 s by:

Infrared light

Direct current electrical stimulation

Ultrasound

Laser

Radiofrequency waves

ThermoMed device

Figuras y tablas -
Table 2. Interventions for Old World cutaneous leishmaniasis
Table 3. Adverse effects of oral antibiotics

Study

Method of assessment

Timing

Interventions

Adverse effects

Kochar 2000

Quote: "Biochemical tests were done to detect any toxic effects of the drug."

Biochemical tests were done at the end of 1 week, 2 weeks and 4 weeks post‐ treatment.

I1: oral rifampicin 1200 mg/d

I2: placebo

Intervention

23 participants evaluated for AEs. Quote: "The drug was well‐tolerated and no side‐effects were seen in any participant."

Placebo

23 participants evaluated for AEs. Not reported

Jaffar 2006

Quote: "clinical examination, liver function tests, renal function tests"

Quote: "Before, during, and after completion of treatment"

I1: oral rifampicin 10 mg/kg/d

I2: placebo

Intervention

46 participants evaluated for AEs. Elevation liver enzymes: 1 (2%)

Placebo

16 participants evaluated for AEs. Not reported

Kochar 2006

Haemoglobin, leukocyte count, and liver test

Biochemical tests were done at the end of 2 week, 4 weeks and 6 weeks post‐treatment

I1: oral rifampicin + omeprazole

I2: placebo

Intervention 1

23 participants evaluated for AEs.

Intervention 2

21 participants evaluated for AEs.

Quote: "All participants tolerated the drug and placebo very well and no side effect was reported."

Layegh 2007

Not described

Azythromycin group: monthly up to 4 months

I1: azithromycin 500 mg/d

I2: IMMA 60 mg/kg/d

Intervention 1

22 participants evaluated for AEs (35 lesions). Nausea and vomiting: 2 (9%)

Intervention 2

27 participants evaluated for AEs (58 lesions). Myalgia: 3 (11%);

Erythema: 1 (3.7%).

Adam 2009

Quote: "Complete haemogram, including haemoglobin and liver and renal function tests Participants were questioned about expected adverse effects for 3 days (Days 5–7) following administration of the doses."

Haemogram: 1 and 2 months

Clinical AEs: days 5‐7; 19‐21; 33‐35; 47‐19.

I1: artesunate

I2: placebo

Intervention 1

20 participants evaluated for AEs.

Placebo

21 participants evaluated for AEs.

Skin rash with itching: 1 (4.7%)

Quote: "There was no significant difference in biological tests (liver and renal function tests) in all the participants before and after treatment."

Ben Salah 2009

Interview, physical examination, laboratory test, evaluation for pain, standardised questionnaire for the occurrence of systemic side effects (e.g. vertigo, tinnitus). Diminished hearing was verified with audiometer.

Laboratory test.

Quote: "Investigators observed each participant each day that the topical creams were administered and at follow‐up study visits (days 50‐100‐180). Clinical and laboratory evidence of side effects was determined on D10 and D20."

I1: WR 279,396

I2: placebo

Intervention

50 participants evaluated for AEs. Erythema at the site of application: 15 (30%); mild pain within 30 minutes of application: 7 (14 %); mild increases and decreases in hearing acuity from baseline: 14 (28%); change hearing acuity: 14 (28%); vertigo: 0 (0%); Increase serum creatinine: 0 (0%); Death: 0 (0%)

Placebo

42 participants evaluated for AEs. Erythema at the site of application: 10 (24%); mild pain within 30 minutes of application: 6 (14 %); mild increases and decreases in hearing acuity from baseline: 9 (21%); change hearing acuity: 9 (21%); vertigo: 0 (0%); increase serum creatinine: 0 (0%); death: 0 (0%)

Dastgheib 2012

Quote: "Participants were interviewed and underwent laboratory tests three times"

Quote: "In Allopurinol group, the participants which were visited and received medication underwent laboratory tests three times (before, one month a2fter, and end of the treatment)

I1: azithromycin + allopurinol

I2: IMMA

Intervention 1

36 participants evaluated for AEs.

Gastrointestinal complaints and headache severe: 1 (2.7%); slight gastrointestinal complications (nausea, heartburn, and epigastric pain): 3 (8.3%)

Intervention 2

35 participants evaluated for AEs.

Myalgia: 2 (5.7%)

AE: adverse effect; IMMA: intramuscular meglumine antimoniate.

Figuras y tablas -
Table 3. Adverse effects of oral antibiotics
Table 4. Adverse effects of topical paromomycin

Study

Method of assessment

Timing

Interventions

Adverse effects

Asilian 1995

Clinical evaluation and laboratory tests

Days 15, 45 and 105

I1: paromomycin

I2: placebo

126 participants evaluated for AEs.

During treatment: oedema, local pain, vesiculation: 1 (0.7%)

After treatment: redness, pain, vesiculation, and inflammation: 8 (6.3%)

Quote: "There were no significant differences in four laboratory test results of safety (SGOT, BUN, Hb, and WBC) between the groups either before or after treatment."

Ben Salah 1995

Clinical evaluation, physical examination, advice to participants, laboratory test: liver function, haemoglobin and white blood cell count

Days 15, 45 and 105

I1: paromomycin

I2: placebo

57 participants evaluated for AEs.

Quote: "A local reaction (inflammation, vesication, pain and/or red ness) was recorded for 12 participants, with no significant difference between the 2 groups."

Laboratory test changes: 0 (0%)

Özgöztasi 1997

Not described

At the end of treatment (day 30) and 1 month post‐treatment

I1: paromomycin + MBCL

I2: oral ketoconazole

40 participants (62 lesions) evaluated for AEs

Quote: "Treatment‐related adverse effects were only observed in the paromomycin group. The most common side‐effect was the development of irritant contact dermatitis. No subjects withdrew because of this adverse effect."

Asilian 2003

Clinical evaluation

Days 15, 29, 45 and 105

I1: paromomycin

I2: placebo

108 participants evaluated for AEs.

Quote: "Treatment was well tolerated, and no adverse reactions to the ointment were observed or reported in either group."

Faghihi 2003

Not described

Quote "Clinical evaluation and follow‐up were performed fortnightly until 1 month post treatment and then monthly until 3 months post treatment, and finally every 3 months until 1 year post treatment"

I1: paromomycin + urea

I2: ILMA weekly

Not described

Shazad 2005

Not described

Week 1 and week 6 post‐treatment and at 6 months after treatment was completed

I1: paromomycin + urea

I2: ILMA weekly

30 participants evaluated for AEs.

Cutaneous reactions (erythematosus, urticaria or lymphadenitis with pain): 1 (3%)

Quote: "No systemic toxic reaction attributable to the drug was observed."

Iraji 2005

Clinical evaluation

Days 7, 14, 21 and 30

I1: paromomycin

I2: placebo

30 participants evaluated for AEs.

Mild contact dermatitis: 3 (10%)

Asilian 2006

Not described

Weekly during treatment and monthly for up 2 months

I1: photodynamic therapy

I2: paromomycin

I3: placebo

19 participants (34 lesions) evaluated for AEs.

Quote: "Adverse side‐effects seen in some participants in all groups were pruritus, burning, redness, discharge, oedema, and pain, but all were generally mild and tolerable."

Ben Salah 2009

Quote: "Renal toxic effects and ototoxic effects from aminoglycoside exposure were ascertained by means of serum creatinine measurements at the end of therapy (at 20 days) and participants' daily reports of tinnitus and vertigo."

Quote: "Safety

end points were assessed daily during therapy (20 days)."

I1:Paromomycin ‐Gentamicin

I2: paromomycin Alone

I3: vehicle Control

Intervention 1:

125 participants evaluated for AEs.

Erythema: 6 (5%); local infection: 0 (0%); inflammation: 0 (0%); vesicles mild‐moderate: 31 (25%); mild oedema: 2 (2%); pain: 2 (2%);

mild bronchitis: 5 (4%); paronychia: 2 (2%); superinfection: 3 (2%); upper respiratory tract infection: 0 (0%); oropharyngeal pain: 4 (3%); skin irritation: 3 (2%); tinnitus: 0 (0%); vertigo: 0 (0%); creatinine serum changes: 0 (0%)

Intervention 2:

125 participants evaluated for AEs.

Erythema: 7 (6%); local infection: 0 (0%); inflammation: 0 (0%); vesicles mild‐moderate: 32 (26%); mild‐moderate oedema: 3 (3%); pain: 2 (2%); bronchitis mild‐moderate: 3 (3%); paronychia: 0 (0%); superinfection: 0 (0%); upper respiratory tract infection: 2 (2%); oropharyngeal pain: 3 (2%); skin irritation: 9 (7%); tinnitus: 0 (0%); vertigo: 0 (0%); creatinine serum changes: 0 (0%)

AE: adverse effect; BUN: blood urea nitrogen; Hb: haemoglobin; ILMA: intralesional meglumine antimoniate; SGOT: serum glutamic‐oxaloacetic transaminase; WBC: white blood cells.

Figuras y tablas -
Table 4. Adverse effects of topical paromomycin
Table 5. Adverse effects of intralesional zinc sulphate

Study

Method of assessment

Timing

Interventions

Adverse effects

Sharquie 1997

Not described

Quote: "Participants were seen at 10‐15 day intervals after injection, and at 6 weeks post treatment"

I1: IL 2% zinc sulphate

I2: IL 7% sodium chloride

I3: ILSSG 2‐5 mL per lesion

19 participants evaluated for AEs.

Quote: "Apart from pain at the time of injection, no appreciable side‐effect was noted."

Iraji 2004

Not described

Not described

I1: IL zinc sulphate

I2: ILMA weekly

31 participants evaluated for AEs.

Severe pain caused vasovagal shock: 2 (6.4%)

Firooz 2005

Not described

Not described

I1: IL zinc sulphate

I2: ILMA weekly

36 participants evaluated for AEs.

Pain: 13 (36.1%); burning at site injection: 3 (8.4%); itching: 3 (8.4%); inflammation: 7 (19.4%)

Maleki 2012

Not described

14, 28, 42, and 56 days after starting the treatment

I1: IL 2% zinc sulphate

I2: ILMA weekly

24 participants evaluated for AEs.

Quote: "The side effects seen in both groups were pain after injection and hyperpigmentation."

Burning after injection and necrosis of the lesions: 24 (100%); inflammation and swelling: 3 (12.5%)

AE: adverse effect; IL: intralesional; ILMA: intralesional meglumine antimoniate; ILSSG: intralesional sodium stibogluconate.

Figuras y tablas -
Table 5. Adverse effects of intralesional zinc sulphate
Table 6. Adverse effects of intralesional hypertonic sodium chloride solution

Study

Method of assessment

Timing

Interventions

Adverse effects

Sharquie 1997

Not described

Quote: "Participants were seen at 10‐15 day intervals after injection, and at 6 week post treatment"

I1: IL 2% zinc sulphate

I2: IL 7% HSCS

I3: ILSSG 2‐5 mL per lesion

17 participants evaluated for AEs.

Quote: "No side‐effect other than pain at the time of injection was noted."

Sadeghian 2006b

Not described

Not described

I1: IL 5% HSCS

I2: ILMA 0.5‐1 mL/week

36 participants evaluated for AEs. Allergic reaction (erythema, oedema, and pruritus): 0 (0%); sporotrichoid dissemination: 3 (8.3%)

Ranawaka 2010

Not described

Quote: "Participants were seen weekly for the first three injections; fortnightly for the fourth and fifth injections; then monthly until cure. Participants were followed‐up every 3 months after cure for 18 months to assess recurrences and evidence of visceralization."

I1: ILSSG

I2: IL 7% HSCS

67 participants evaluated for AEs.

Leishmaniasis recidivans: 0 (0%)

Quote: "There were no systemic side effects with SSG or HS. Pain during injection was the only local side effect noted with both therapies. After healing, scarring was minimal, but postinflammatory hyperpigmentation

was observed in all participants for both treatments, which faded out over 6–8 months."

AE: adverse effect; IL: intralesional; ILSSG: intralesional sodium stibogluconate; HSCS: hypertonic sodium chloride solution.

Figuras y tablas -
Table 6. Adverse effects of intralesional hypertonic sodium chloride solution
Table 7. Adverse effects of laser

Study

Method of assessment

Timing

Interventions

Adverse effects

Asilian 2004b

Not described

1, 3, 4, 8, 12 and 24 weeks after treatment

I1: CO₂

I2: IMMA 50 mg/kg/d

123 participants evaluated for AEs. Quote: "Complications were seen in (4) 4.5% of participants and included hyperpigmentation, persistent redness." Hypertonic scars: 5 (4%)

Shamsi Meymandi 2011

Quote: "Follow‐up was performed and any side‐effects were recorded."

Quote: "Follow‐up evaluation was performed by clinical assessment of treated lesions at weeks 2, 6, 12 and 16."

I1: CO₂

I2: cryotherapy + MA

80 participants (95 lesions) evaluated for AEs.

Hyperpigmentation + trivial scar: 20 (25%); atrophic scar: 7 (8.75%); hypertrophic scar: 1 (1.25%); sporotrichoid: 1 (1.25%); raised papular lesions: 1 (1.25%); persistent erythema: 3 (3.75%); hypopigmentation + trivial scar: 4 (5%)

Nilforoushzadeh 2014a

Not described

Quote: "Participants were followed in the first, third, and sixth months after treatment with the final evaluation in the sixth month."

I1: ablative CO₂ laser

I2: fractional CO₂ laser

Intervention 1:

30 participants evaluated for AEs.

Erythema: 2 (6.7%)

Intervention 2:

30 participants evaluated for AEs.

Erythema: 4 (13.3%)

AE: adverse effect; IMMA: intramuscular meglumine antimoniate; MA: meglumine antimoniate.

Figuras y tablas -
Table 7. Adverse effects of laser
Table 8. Adverse effects of cryotherapy

Study

Method of assessment

Timing

Interventions

Adverse effects

Asilian 2004a

Not described

Fortnightly until 6 months post‐treatment and 2 weeks and 4 weeks post‐treatment

I1: cryotherapy + ILMA

I2: cryotherapy alone

I3: ILMA alone

Intervention 1:

100 participants evaluated for AEs. Postinflammatory hypopigmentation: 5 (5%)

Intervention 2:

200 participants evaluated for AEs. Postinflammatory hypopigmentation: 10 (5%)

Salmanpour 2006

Not described

Not described

I1: ILMA alone

I2: cryotherapy alone

I3: cryotherapy + ILMA

Intervention 2:

20 participants evaluated for AEs.

Erythema and oedema of the lesions and perilesional area: (28%)

Intervention 3:

20 participants evaluated for AEs.

Erythema and oedema of the lesions and perilesional area: (33%)

Quote: "There were no serious side‐effects in any of the treatment groups"

Layegh 2009

Not described

Quote: "Weekly for up to six weeks of treatment and six months after."

I1: cryotherapy

I2: ILMA

36 participants evaluated for AEs.

Hypopigmentation: 2 (5.5%); hyperpigmentation: 7 (19.4%).

Quote: "the most common adverse reactions were erythema and oedema of the treated site, which appeared during the initial hours of treatment, and blistering of the treatment site, which became evident 1–2 days after treatment and responded well to local treatment."

Shamsi Meymandi 2011

Quote: "Follow‐up was performed and any side‐effects were recorded."

Quote: "Follow‐up evaluation was performed by clinical assessment of treated lesions at weeks 2, 6, 12 and 16."

I1: CO₂

I2: cryotherapy + MA

80 participants (95 lesions) evaluated for AEs.

Hyperpigmentation + trivial scar: 15 (18.7%);

atrophic scar: 6 (7.5%); hypertrophic scar: 0 (0%); sporotrichoid: 0 (0%); raised papular lesions: 0 (0%); persistent erythema: 0 (0%); hypopigmentation + trivial scar: 15 (18.8%)

Jowkar 2012

Quote: "During these visits the healing process of the ulcer, change of diameter and induration of lesions and complications were assessed."

Quote: "The participants were evaluated every 2 weeks up to 12 weeks."

I1: cryotherapy + 3% salicylic + 3% sodium nitrite

I2: cryotherapy + 3% salicylic + placebo

Intervention 1:

36 participants evaluated for AEs.

Erythema, a burning sensation and skin irritation: 7 (19.4%)

Intervention 2:

27 participants evaluated for AEs.

Erythema, a burning sensation and skin irritation: 1 (3.7%)

AE: adverse effect; ILMA: intralesional meglumine antimoniate; MA: meglumine antimoniate.

Figuras y tablas -
Table 8. Adverse effects of cryotherapy
Table 9. Adverse effects of thermotherapy

Study

Method of assessment

Timing

Interventions

Adverse effects

Reithinger 2005

Quote: "The occurrence of adverse effects was evaluated blindly by means of participant interviews and physical examinations."

Quote: "The occurrence of adverse effects was evaluated … during follow‐up visits."

I1: ILSSG 2‐5 mL per lesion

I2: IMSSG 20 mg/kg

I3: thermotherapy

138‐108 participants evaluated for AEs.

Secondary infections: 8 (5.7%).

Quote: "The original CL ulcer often increased in size immediately after and up to 2 weeks after treatment."

Sadeghian 2007

Quote: "Appearance of lesions at subsequent follow‐up visits and occurrence of unwanted side‐effects were also recorded on the form."

Weekly 4 weeks and monthly up to 6 months

I1: thermotherapy

I2: ILMA weekly

57 participants (83 lesions) evaluated for AEs.

Satellite lesions: 1 (1.7%)

Aronson 2010

Quote: "Interview, physical examination, laboratory testing (complete blood count, creatine phosphokinase, amylase, lipase, complete metabolic profile), and electrocardiograms."

Quote: "Daily for the first 10 days and follow‐up at 2, 6, and 12–24 months post treatment"

I1: thermotherapy

I2: ILSSG

27 participants evaluated for AEs.

Serious AE: 4 (15%); ECG changes: 10 (37%); abdominal discomfort: 1 (4%); wound infection: 5 (19%); musculoskeletal: 5 (19%); headache: 3 (11%); fatigue: 5 (19%); rash: 1 (4%); blister reaction: 25 (93%); erythema: 7 (26%); oozing: 21 (78%)

Safi 2012

Quote: "The occurrences of adverse effects were evaluated by means of participant interviews and physical examinations during follow‐up visits."

Quote: "During treatment, all participants were then followed for four visits at weekly intervals … After initial treatment, all participants were scheduled for four subsequent follow‐up visits: 10 days after baseline and 1‐month, 2 months and 6 months after treatment."

I1: thermotherapy

I2: ILMA weekly

189 participants evaluated for AEs

Not reported

Bumb 2013

Not described

Not described

I1: radiofrequency heat treatment

I2: ILSSG

Quote: "RFHT was cosmetically acceptable because it was associated with less scarring and hyperpigmentation compared with intralesional SSG injections."

Jebran 2014

Quote: "In case of clinical signs for a superinfection, a smear was taken, Gram stained and microscopically evaluated for the presence of bacteria and/or fungi."

Quote: "Adverse events such as bacterial or fungal superinfections of the wounds, the formation or scars and the rate of re‐ulcerations were monitored during the treatment and follow‐up period."

I1: electrocauterisation + DAC N‐055.

I2: electrocauterisation + placebo.

Intervention 1:

38 participants evaluated for AEs.

Bacterial and fungal superinfections: 3 (8.0%); Keloïd formation: 2 (5%)

Intervention 2:

32 participants evaluated for AEs.

Bacterial and fungal superinfections: 3 (9.0%); Keloïd formation: 2 (6%)

AE: adverse effect; CL: cutaneous leishmaniasis; ILMA: intralesional meglumine antimoniate; ILSSG: intralesional sodium stibogluconate; IMSSG: intramuscular sodium stibogluconate.

Figuras y tablas -
Table 9. Adverse effects of thermotherapy
Comparison 1. ILMA weekly versus ILMA fortnightly for up to 8 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 1. ILMA weekly versus ILMA fortnightly for up to 8 weeks
Comparison 2. ILMA (every other day) versus IMMA (6 d/week) for up to 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 2. ILMA (every other day) versus IMMA (6 d/week) for up to 4 weeks
Comparison 3. IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (30 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 3. IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (30 mg/kg/d for 3 weeks) + placebo
Comparison 4. IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 4. IMMA (30 mg/kg/d for 3 weeks) + cimetidine versus IMMA (60 mg/kg/d for 3 weeks) + placebo
Comparison 5. IMMA (60 mg/kg/d for 3 weeks) + placebo versus IMMA (30 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 5. IMMA (60 mg/kg/d for 3 weeks) + placebo versus IMMA (30 mg/kg/d for 3 weeks) + placebo
Comparison 6. IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

2

148

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.71, 0.96]

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Serious adverse effects

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Skin reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Cardiac toxicity 'QT prolongation'

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 6. IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo
Comparison 7. IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 7. IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo
Comparison 8. IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 8. IMMA (30 mg/kg/d for 3 weeks) + placebo versus IMMA (60 mg/kg/d for 3 weeks) + placebo
Comparison 9. IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 9. IMMA (30 mg/kg/d for 3 weeks) + 40 mg omeprazole versus IMMA (60 mg/kg/d for 3 weeks) + placebo
Comparison 10. ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects (itching and burning) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Adverse effects (oedema) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 10. ILMA + non‐silver polyester dressing versus ILMA (weekly injections for 6 weeks)
Comparison 11. ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects (itching and burning) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Adverse effects (oedema) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 11. ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks)
Comparison 12. ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects (itching and burning) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Adverse effects (oedema) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 12. ILMA (weekly injections for 6 weeks) + silver polyester dressing versus ILMA (weekly injections for 6 weeks) + non‐silver polyester dressing
Comparison 13. ILMA (weekly injections for 6 weeks) + gel mask twice a day versus ILMA (weekly injections for 6 weeks) + vehicle

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 13. ILMA (weekly injections for 6 weeks) + gel mask twice a day versus ILMA (weekly injections for 6 weeks) + vehicle
Comparison 14. ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5) versus ILSSG (1000 mg/mL days 1, 3, 5)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 14. ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5) versus ILSSG (1000 mg/mL days 1, 3, 5)
Comparison 15. ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects (mild heart symptoms) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 15. ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 d for 29 days) versus IMSSG (20 mg/kg/d for 3 weeks)
Comparison 16. Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects (nausea and vomiting) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 16. Ketoconazole 600 mg/d for 6 weeks versus ketoconazole 800 mg/d for 6 weeks
Comparison 17. Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effect (liver enzymes increase) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 17. Ketoconazole 600 mg/d for 30 d versus ILMA (6 to 8 biweekly injections)
Comparison 18. ILSSG (100 mg/mL days 1, 3, 5) + oral ketoconazole (600 mg/d for 4 weeks) versus ILSSG (100 mg/mL days 1, 3, 5)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 18. ILSSG (100 mg/mL days 1, 3, 5) + oral ketoconazole (600 mg/d for 4 weeks) versus ILSSG (100 mg/mL days 1, 3, 5)
Comparison 19. ILSSG (100 mg/mL days 1, 3, 5) + ketoconazole (600 mg/d for 4 weeks) versus ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 19. ILSSG (100 mg/mL days 1, 3, 5) + ketoconazole (600 mg/d for 4 weeks) versus ILSSG (20 mg/kg/d) + IMSSG (remaining total dose days 1, 3, 5)
Comparison 20. Itraconazole (200 mg for 6 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Figuras y tablas -
Comparison 20. Itraconazole (200 mg for 6 weeks) versus placebo
Comparison 21. Itraconazole (200 mg for 3 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Figuras y tablas -
Comparison 21. Itraconazole (200 mg for 3 weeks) versus placebo
Comparison 22. Itraconazole (200 mg for 6 to 8 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

3

244

Risk Ratio (M‐H, Random, 99% CI)

3.70 [0.35, 38.99]

2 Adverse effects Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Mild abdominal pain and nausea

3

204

Risk Ratio (M‐H, Random, 95% CI)

2.36 [0.74, 7.47]

2.2 Mild abnormal liver function

3

84

Risk Ratio (M‐H, Random, 95% CI)

3.08 [0.53, 17.98]

2.3 Headache and dizziness

1

20

Risk Ratio (M‐H, Random, 95% CI)

2.63 [0.16, 43.63]

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Figuras y tablas -
Comparison 22. Itraconazole (200 mg for 6 to 8 weeks) versus placebo
Comparison 23. Itraconazole (200 mg for 6 to 8 weeks) versus no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

2 Adverse effects (headache and dizziness) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 23. Itraconazole (200 mg for 6 to 8 weeks) versus no treatment
Comparison 24. Fluconazole (200 mg for 6 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 24. Fluconazole (200 mg for 6 weeks) versus placebo
Comparison 25. Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Rise creatinine and liver enzymes

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Cheilitis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Nausea

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 25. Fluconazole (400 mg/d for 6 weeks) versus fluconazole (200 mg/d for 6 weeks)
Comparison 26. Oral dapsone (200 mg/d for 6 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete Cure Show forest plot

2

160

Risk Ratio (M‐H, Random, 95% CI)

24.08 [1.44, 403.43]

2 Adverse effects Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Nausea

2

160

Risk Ratio (M‐H, Random, 95% CI)

21.86 [3.04, 157.29]

Figuras y tablas -
Comparison 26. Oral dapsone (200 mg/d for 6 weeks) versus placebo
Comparison 27. Allopurinol (15 mg/kg/d for 3 weeks) + IMMA (20 mg/kg/d for 2 weeks) versus allopurinol (15 mg/kg/d for 3 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

3.83 [1.71, 8.60]

Figuras y tablas -
Comparison 27. Allopurinol (15 mg/kg/d for 3 weeks) + IMMA (20 mg/kg/d for 2 weeks) versus allopurinol (15 mg/kg/d for 3 weeks)
Comparison 28. Allopurinol (15mg/kg/d for 3 weeks)+ IMMA (20 mg/kg/d for 2 weeks) versus IMMA (20 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

1.92 [1.08, 3.41]

Figuras y tablas -
Comparison 28. Allopurinol (15mg/kg/d for 3 weeks)+ IMMA (20 mg/kg/d for 2 weeks) versus IMMA (20 mg/kg/d for 2 weeks)
Comparison 29. Allopurinol (15 mg/kg/d for 3 weeks) versus IMMA (20 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions Cured Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.35, 1.62]

Figuras y tablas -
Comparison 29. Allopurinol (15 mg/kg/d for 3 weeks) versus IMMA (20 mg/kg/d for 2 weeks)
Comparison 30. Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Mild abdominal pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Skin eruption

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Muscle pain and weakness

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Microbiological cure of skin lesions Show forest plot

1

72

Risk Ratio (M‐H, Random, 95% CI)

1.11 [0.88, 1.41]

Figuras y tablas -
Comparison 30. Allopurinol (20 mg/kg/d for 3 weeks) + IMMA (30 mg/kg/d for 20 days) versus IMMA (60 mg/kg/d for 20 d)
Comparison 31. Allopurinol (20 mg/kg/d for 3 weeks)+ IMMA (10 mg/kg/d for 20 d) versus IMMA (20 mg/kg/d for 28 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

1.1 Secondary infection

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Myalgia

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.3 ECG changes

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.4 Chest pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.5 Pain injection site

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.6 Abscess

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 31. Allopurinol (20 mg/kg/d for 3 weeks)+ IMMA (10 mg/kg/d for 20 d) versus IMMA (20 mg/kg/d for 28 d)
Comparison 32. Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Abdominal symptoms

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Liver abnormalities

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Myalgia

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.4 Rash

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 32. Allopurinol (20 mg/kg/d for 3 weeks) versus IVSSG (20 mg/kg/d for 15 d)
Comparison 33. Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

1.1 Evaluated 3 months after treatment

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Evaluated after treatment

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 33. Oral rifampicin (10 mg/kg/d for 4 to 6 weeks) versus placebo
Comparison 34. Oral rifampicin (10 mg/kg/d) + omeprazole (20 mg/d) for 6 weeks versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 34. Oral rifampicin (10 mg/kg/d) + omeprazole (20 mg/d) for 6 weeks versus placebo
Comparison 35. Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Nausea and vomiting

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 35. Azythromicin (500 mg/d for 5 d/month up to 4 months) versus IMMA (60 mg/kg/d for 20 d)
Comparison 36. Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Gastrointestinal complaints and headache

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Gastrointestinal complications

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Myalgia

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 36. Azythromicin (10 mg/kg/d) + allopurinol (10 mg/kg/d) for 1 month versus IMMA (20 mg/kg/d for 20 d)
Comparison 37. Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Allergic macule‐papular

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 37. Oral pentoxifylline (400 mg 3 times daily) + IMMA (20 mg/kg/d) for 20 d versus placebo + IMMA (20 mg/kg/d) for 20 d
Comparison 38. Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 38. Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks)
Comparison 39. Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 39. Oral miltefosine (2.5 mg/kg/d for 4 weeks) versus IMMA (60 mg/kg/d for 2 weeks)
Comparison 40. Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Nausea and vomiting

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 40. Oral zinc sulphate 2.5 mg/kg/d for 45 days versus oral zinc sulphate 5 mg/kg/d for 45 d
Comparison 41. Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Nausea and vomiting

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 41. Oral zinc sulphate 2.5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d
Comparison 42. Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Nausea and vomiting

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 42. Oral zinc sulphate 5 mg/kg/d for 45 d versus oral zinc sulphate 10 mg/kg/d for 45 d
Comparison 43. Oral zinc sulphate (10 mg/kg/d for 45 d) versus IMMA (20 mg/kg/d for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 43. Oral zinc sulphate (10 mg/kg/d for 45 d) versus IMMA (20 mg/kg/d for 20 d)
Comparison 44. Artesunate 400 mg + sulphamethoxypyrazine/pyrimethamine 1000 mg/50 mg 4 times daily for 4 d versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 44. Artesunate 400 mg + sulphamethoxypyrazine/pyrimethamine 1000 mg/50 mg 4 times daily for 4 d versus placebo
Comparison 45. Topical 2% miconazole (twice a day) versus topical 1% clotrimazole (twice a day) for 30 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 45. Topical 2% miconazole (twice a day) versus topical 1% clotrimazole (twice a day) for 30 d
Comparison 46. Topical ketoconazole (twice a day) versus vehicle (twice a day) for 30 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 46. Topical ketoconazole (twice a day) versus vehicle (twice a day) for 30 d
Comparison 47. Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure (ITT) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Hypersensitivity

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Mild pruritus

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Erithema and oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 47. Topical amphotericin B (3 to 7 drops twice daily for 8 weeks) versus ILMA (max 2 mL) once a week for 8 weeks
Comparison 48. Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Scarring Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 48. Paromomycin 15% + 12% MBCL (twice daily for 28 d) versus vehicle (twice daily for 28 d)
Comparison 49. Paromomycin (twice daily for 30 d) versus vehicle (twice daily for 30 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.71 [0.25, 2.06]

Figuras y tablas -
Comparison 49. Paromomycin (twice daily for 30 d) versus vehicle (twice daily for 30 d)
Comparison 50. Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

2

383

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.86, 1.17]

2 Adverse effects Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Skin/local reaction

3

463

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.45, 1.93]

3 Microbiological cure of skin lesions Show forest plot

2

383

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.88, 1.20]

Figuras y tablas -
Comparison 50. Paromomycin 15% + 10% urea (twice daily for 14 d) versus vehicle (twice daily for 14 d)
Comparison 51. Paromomycin 15% (daily for 20 d) versus vehicle

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 51. Paromomycin 15% (daily for 20 d) versus vehicle
Comparison 52. Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus vehicle

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 52. Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus vehicle
Comparison 53. Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus paromomycin 15% alone (daily for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 53. Paromomycin 15% + gentamicin 0.5% (daily for 20 d) versus paromomycin 15% alone (daily for 20 d)
Comparison 54. Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Scarring Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 54. Paromomycin 15% + 10% urea (twice daily for 45 d) versus ILMA (weekly for up to 3 months)
Comparison 55. Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Cutaneous reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 55. Paromomycin 15% + 10% urea (twice daily for 20 d) versus ILMA (weekly for up to 20 d)
Comparison 56. Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 56. Paromomycin + MBCL (twice daily for 15 d) versus ketoconazole (weekly for up to 30 d)
Comparison 57. Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Scarring Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 57. Paromomycin (15% + 12% MBCL twice daily for 28 days) versus PDT (weekly for 4 weeks)
Comparison 58. Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 58. Paromomycin (4 weeks) versus paromomycin (2 weeks) + vehicle (2 weeks)
Comparison 59. IL zinc 2% (twice a week for 2 weeks) versus ILSSG (100 mg/mL) for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 59. IL zinc 2% (twice a week for 2 weeks) versus ILSSG (100 mg/mL) for 2 weeks)
Comparison 60. IL zinc 2% (twice a week for 2 weeks) versus IL 7% HSCS for 2 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 60. IL zinc 2% (twice a week for 2 weeks) versus IL 7% HSCS for 2 weeks
Comparison 61. ILSSG (100 mg/mL) for 2 weeks versus IL 7% HSCS for 2 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 61. ILSSG (100 mg/mL) for 2 weeks versus IL 7% HSCS for 2 weeks
Comparison 62. IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Participants complete cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Adverse effects Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 Pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.2 Burning

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.3 Itching

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.4 Inflammation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.5 Pruritus and erythema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.6 Severe pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 62. IL zinc 2% (weekly for up to 6 weeks) versus ILMA (max 2 mL weekly for up to 6 weeks)
Comparison 63. IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Inflammation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 63. IL zinc 2% (twice a week for 2 weeks) versus ILMA (60 mg/kg/d for 2 weeks)
Comparison 64. Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Participants with treated lesions that recur Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 Itch and burning

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 64. Imiquimod (5% 3 times/week for 28 d) + IMMA (20 mg/kg/d for 14 d) versus vehicle + IMMA
Comparison 65. IL 7% HSCS (0.2 mL to 7 mL per lesion) versus ILSSG (max 2 mL) max 5 injections

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 65. IL 7% HSCS (0.2 mL to 7 mL per lesion) versus ILSSG (max 2 mL) max 5 injections
Comparison 66. IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Sporotrichotic dissemination

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 66. IL 5% HSCS (0.5 mL to 1 mL per lesion) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 6 to 10 weeks
Comparison 67. IL 7% HSCS (0.1 mL to 0.5 mL per lesion) versus IL 2% ciprofloxacin solution (0.1 mL to 0.5 mL per lesion)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 67. IL 7% HSCS (0.1 mL to 0.5 mL per lesion) versus IL 2% ciprofloxacin solution (0.1 mL to 0.5 mL per lesion)
Comparison 68. IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

4 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

4.1 Ulceration and necrosis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 68. IL 15% HSCS (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion)
Comparison 69. ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

4 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

4.1 Ulceration and necrosis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 69. ILSSG (0.2 mL to 4 mL per lesion) max 5 injections versus IL 10% HSCS (0.2 mL to 4 mL per lesion)
Comparison 70. ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

3 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

4 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

4.1 Ulceration and necrosis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 70. ILSSG (0.2 mL to 4 mL per lesion) maximum 5 injections versus IL 15% HSCS (0.2 mL to 4 mL per lesion)
Comparison 71. IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 71. IL IFN‐γ (weekly for 5 weeks) versus ILMA (0.5 mL to 1 mL per lesion) weekly for 5 weeks
Comparison 72. WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Erythema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Mild pain

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Hearing acuity problems

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 72. WR279,396 (twice a day for 20 d) versus vehicle (twice a day for 20 d)
Comparison 73. IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Local inflammatory reactions

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 73. IL metronidazole (2.5 mg to 10 mg each lesion) versus ILMA (150 mg to 600 mg each lesion) for up to 8 weeks
Comparison 74. Topical miltefosine 6% (once daily) versus ILMA (twice a week) for up to 28 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 74. Topical miltefosine 6% (once daily) versus ILMA (twice a week) for up to 28 d
Comparison 75. Dapsone gel 5% (twice a day) + ILMA (weekly) versus cryotherapy (every 2 weeks) + IMMA (weekly) for up to 16 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 75. Dapsone gel 5% (twice a day) + ILMA (weekly) versus cryotherapy (every 2 weeks) + IMMA (weekly) for up to 16 weeks
Comparison 76. DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Reulceration

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Keloïd scars

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 76. DAC‐055 + MWT (for 15 min) versus DAC‐055 alone for up to 75 d
Comparison 77. DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Reulceration

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Keloïd scars

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 77. DAC‐055 + heat (for 15 min) versus ILSSG (0.6 mL) for up to 75 d
Comparison 78. DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Reulceration

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Keloïd scars

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 78. DAC‐055 alone (for 15 min) versus ILSSG (0.6 mL) for up to 75 d
Comparison 79. Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 Dizziness and nausea

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.2 Hypersensitive reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 79. Thio‐Ben (1 mL to 2 mL daily) + cryotherapy (fortnightly) versus ILMA (0.5 mL to 2 mL per lesions) weekly + cryotherapy (fortnightly) for up to 12 weeks
Comparison 80. CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

433

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.53, 1.55]

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Hyperpigmentation and redness

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Hypertrophic scarring

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Systemic symptoms

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 80. CO₂ laser (30 W continuous) versus IMMA (50 mg/kg/d) for up to 15 d
Comparison 81. CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Hyperpigmentation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Atrophic scar

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 Hypopigmentation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 81. CO₂ laser (30 W continuous) versus cryotherapy (fortnightly) + ILMA (weekly) for up to 12 weeks
Comparison 82. Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Partcipants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Erythema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 82. Ablative CO₂ laser (25 kW for 1 session) versus 3 weeks fractional CO₂ laser
Comparison 83. TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Recurrence Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 Mild erythema and itch

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Microbiological cure of skin lesions Show forest plot

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.74, 1.26]

Figuras y tablas -
Comparison 83. TCA (50% wt/vol) fortnightly up to 3 times versus ILMA alone (weekly for up to 6 weeks)
Comparison 84. Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

2.1 Males

1

Risk Ratio (M‐H, Random, 99% CI)

0.0 [0.0, 0.0]

2.2 Females

1

Risk Ratio (M‐H, Random, 99% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 84. Topical TCA 50% + local heat versus ILMA twice a week for up to 8 weeks
Comparison 85. TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 85. TCA + ILMA (weekly for up to 8 weeks) versus ILMA alone (twice a week for up to 8 weeks)
Comparison 86. Fractional laser + ILMA (fortnightly 2 sessions) versus ILMA alone (twice a week for up to 8 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 86. Fractional laser + ILMA (fortnightly 2 sessions) versus ILMA alone (twice a week for up to 8 weeks)
Comparison 87. TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 87. TCA + ILMA (weekly for up to 8 weeks) versus fractional laser + ILMA (fortnightly 2 sessions)
Comparison 88. TCA fortnightly up to 8 weeks + ILMA (twice a week) versus ILMA alone (weekly for up to 8 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 88. TCA fortnightly up to 8 weeks + ILMA (twice a week) versus ILMA alone (weekly for up to 8 weeks)
Comparison 89. Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Erythema and oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 89. Cryotherapy + ILMA (weekly) versus cryotherapy (weekly) for up to 6 weeks
Comparison 90. Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Erythema and oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 90. Cryotherapy + ILMA (weekly) versus ILMA (weekly) for up to 6 weeks
Comparison 91. Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Erythema and oedema

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 91. Cryotherapy + ILMA (weekly) versus ILMA alone (weekly) for up to 6 weeks
Comparison 92. Cryotherapy (weekly) versus ILMA (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 92. Cryotherapy (weekly) versus ILMA (weekly) for up to 6 weeks
Comparison 93. Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Hypopigmentation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 93. Cryotherapy + ILMA (weekly) versus cryotherapy alone (weekly) for up to 6 weeks
Comparison 94. Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Hypopigmentation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 94. Cryotherapy + ILMA (weekly) versus ILMA (fortnightly) for up to 6 weeks
Comparison 95. Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

390

Risk Ratio (M‐H, Random, 95% CI)

14.64 [0.86, 247.99]

2.1 Hypopigmentation

1

390

Risk Ratio (M‐H, Random, 95% CI)

14.64 [0.86, 247.99]

Figuras y tablas -
Comparison 95. Cryotherapy alone (weekly) versus ILMA (fortnightly) for up to 6 weeks
Comparison 96. Cryotherapy (fortnightly) + 15% paromomycin + 10% urea cream (twice a day) + ILMA (twice a day for 4 weeks) versus ILMA (twice a week) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 96. Cryotherapy (fortnightly) + 15% paromomycin + 10% urea cream (twice a day) + ILMA (twice a day for 4 weeks) versus ILMA (twice a week) for up to 6 weeks
Comparison 97. Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Mild skin symptoms

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 97. Cryotherapy (weekly) + 3% salicylic + 3% sodium nitrite cream (twice a day) for up to 12 weeks versus cryotherapy (weekly) + 3% salicylic cream (twice a day)
Comparison 98. Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Participants complete cure Show forest plot

2

499

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.97, 1.55]

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 98. Radiofrequency waves versus ILMA (1 mL to 7 mL per lesion) weekly for 4 weeks
Comparison 99. Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects (serious) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 99. Radiofrequency waves (50 uCTM applied for 30 s) versus ILSSG (10 days of 20 mg/kg/d)
Comparison 100. Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse event (secondary infection) Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Figuras y tablas -
Comparison 100. Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus IMSSG (20 mg/kg/d for 3 weeks)
Comparison 101. Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse event (secondary infection) Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Figuras y tablas -
Comparison 101. Radiofrequency waves (1 treatment of > 1 consecutive application at 50ºC for 30 s) versus ILSSG (5 injections of 2 mL to 5 mL every 5 to 7 days)
Comparison 102. Radiofrequency waves versus ILSSG

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 102. Radiofrequency waves versus ILSSG
Comparison 103. Electrocauterisation + DAC n‐055 (daily) versus electrocauterisation

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

1.1 Superinfection

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 Keloid formation

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 103. Electrocauterisation + DAC n‐055 (daily) versus electrocauterisation
Comparison 104. PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lesions cured Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Scarring Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 104. PDT (weekly for 4 weeks) versus placebo (twice a day for 4 weeks)
Comparison 105. Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Development of cell‐mediated immunity Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 105. Mesotherapy gun (0.5 mL of MA weekly) versus ILMA (0.1 mL weekly) for up to 6 weeks
Comparison 106. Diminazene aceturate solution (weekly) versus cetrimide + chlorhexidine solution for 50 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 106. Diminazene aceturate solution (weekly) versus cetrimide + chlorhexidine solution for 50 d
Comparison 107. Topical garlic (twice a day) versus vehicle for 3 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 107. Topical garlic (twice a day) versus vehicle for 3 weeks
Comparison 108. Topical herbal extract + placebo (5 d) versus IMMA (15‐20/mg/kg/d) + vehicle for 20 d

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 108. Topical herbal extract + placebo (5 d) versus IMMA (15‐20/mg/kg/d) + vehicle for 20 d
Comparison 109. Topical honey (twice a day) + ILMA (weekly) versus ILMA (weekly) for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 109. Topical honey (twice a day) + ILMA (weekly) versus ILMA (weekly) for 4 weeks
Comparison 110. Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Itching

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 110. Cassia fistula (topical gel) + ILMA (0.5 mL to 2 mL), twice a week versus ILMA (0.5 mL to 2 mL), twice a week + vehicle
Comparison 111. Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 111. Cassia fistula boiled (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks
Comparison 112. Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Speed of healing (weeks) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Adverse reaction Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 112. Cassia fistula hydroalcoholic (topical) versus ILMA (0.5 mL to 2 mL), twice a week for 4 weeks
Comparison 113. Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Speed of healing (days) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 Allergic reaction

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 113. Cassia fistula boiled (topical) versusC fistula hydroalcoholic (topical) for 4 weeks
Comparison 114. Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants complete cure Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Itching

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Microbiological cure of skin lesions Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 114. Topical gel Achilles millefollium (twice daily) + ILMA (weekly 20 mg/kg/d) versus ILMA (weekly 20 mg/kg/d) + vehicle (twice daily) for 4 weeks