Scolaris Content Display Scolaris Content Display

Антигельминтные средства в эндемичных по гельминтам районах: влияние на прогрессирование ВИЧ‐инфекции

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Abate 2014 ETH {published and unpublished data}

Abate E, Elias D, Getachew A, Alemu S, Diro E, Britton S, et al. Effects of albendazole on the clinical outcome and immunological responses in helminth co‐infected tuberculosis patients: a double blind randomised clinical trial. International Journal for Parasitology 2014;45(2‐3):133‐40.

Kallestrup 2005 ZWE {published and unpublished data}

Kallestrup P, Zinyama R, Gomo E, Butterworth AE, Mudenge B, van Dam GJ, et al. Schistosomiasis and HIV‐1 infection in rural Zimbabwe: effect of treatment of schistosomiasis on CD4 cell count and plasma HIV‐1 RNA load. The Journal of Infectious Diseases 2005;192(11):1956‐61.

Kelly 1996 ZMB {published data only}

Kelly P, Lungu F, Keane E, Baggaley R, Kazembe F, Pobee J, et al. Albendazole chemotherapy for treatment of diarrhoea in patients with AIDS in Zambia: a randomized double blind controlled trial. BMJ 1996;312(7040):1187‐91.

Nielsen 2007 TZA {published and unpublished data}

Nielsen NO, Simonsen PE, Dalgaard P, Krarup H, Magnussen P, Magesa S, et al. Effect of diethycarbamazine on HIV load, CD4%, and CD4/CD8 ratio in HIV‐infected adult Tanzanians with or without lymphatic filariasis: randomized double‐blind and placebo‐controlled cross‐over trial. American Journal of Tropical Medicine and Hygiene 2007;77(3):507‐13.
Nielsen NO, Simonsen PE, Kaestel P, Krarup H, Magnussen P, Magesa S, et al. Micronutrient status indicators in individuals single‐ or double‐infected with HIV and Wuchereria bancrofti before and after DEC treatment. Tropical Medicine & International Health 2009;14(1):44‐53.

Suputtamongkol 2011 THA {published and unpublished data}

Suputtamongkol Y, Premasathian N, Bhumimuang K, Waywa D, Nilganuwong S, Karuphong E, et al. Efficacy and safety of single and double doses of ivermectin versus 7‐day high dose albendazole for chronic strongyloidiasis. PLoS Neglected Tropical Diseases 2011;5(5):e1044.

Walson 2008 KEN {published and unpublished data}

Walson JL, Otieno PA, Mbuchi M, Richardson BA, Lohman‐Payne B, Macharia SW, et al. Albendazole treatment of HIV‐1 and helminth co‐infection: a randomized, double‐blind, placebo‐controlled trial. AIDS 2008;22(13):1601‐9.

Walson 2012 KEN {published and unpublished data}

Walson J, Singa B, Sangaré L, Naulikha J, Piper B, Richardson B, et al. Empiric deworming to delay HIV disease progression in adults with HIV who are ineligible for initiation of antiretroviral treatment (the HEAT study): a multi‐site, randomised trial. The Lancet Infectious diseases 2012;12(12):925‐32. [PUBMED: 22971323]

Webb 2012 UGA {published and unpublished data}

Webb EL, Kyosiimire‐Lugemwa J, Kizito D, Nkurunziza P, Lule S, Muhangi L, et al. The effect of anthelmintic treatment during pregnancy on HIV plasma viral load: results from a randomized, double‐blind, placebo‐controlled trial in Uganda. Journal of Acquired Immune Deficiency Syndromes 2012;60(3):307‐13.

References to studies excluded from this review

Blish 2010 {published data only}

Blish CA, Sangaré L, Herrin BR, Richardson BA, John‐Stewart G, Walson JL. Changes in plasma cytokines after treatment of Ascaris lumbricoides infection in individuals with HIV‐1 infection. The Journal of Infectious Diseases 2010;201(12):1816‐21.

Brown 2004 {published data only}

Brown M, Kizza M, Watera C, Quigley MA, Rowland A, Highes P, et al. Helminth infection is not associated with faster progression of HIV disease in coinfected adults in Uganda. The Journal of Infectious Diseases 2004;190(10):1869‐79.

Brown 2005 {published data only}

Brown M, Mawa PA, Joseph S, Bukusuba J, Watera C, Whitworth JAG, et al. Treatment of Schistosoma mansoni infection increases helminth‐specific type 2 cytokine responses and HIV‐1 loads in coinfected Ugandan adults. Journal of Infectious Diseases 2005;191(10):1648‐57.

Elliott 2003 {published data only}

Elliott AM, Mawa PA, Joseph S, Namujju PB, Kizza M, Nakiyingi JS, et al. Associations between helminth infection and CD4+ T cell count, viral load and cytokine responses in HIV‐1 infected Ugandan adults. Transactions of the Royal Society of Tropical Medicine and Hygiene 2003;97(1):103‐8.

Elliott 2007 {published data only}

Elliott AM, Kizza M, Quigley MA, Ndibazza J, Nampijja M, Muhangi L, et al. The impact of helminths on the response to immunization and on the incidence of infection and disease in childhood in Uganda: design of a randomized, double‐blind, placebo‐controlled, factorial trial of deworming interventions delivered in pregnancy and early childhood [ISRCTN32849447]. Clinical Trials 2007;4(1):42‐57.

Erikstrup 2008 {published data only}

Erikstrup C, Kallestrup P, Zinyama‐Gutsire RB, Gomo E, van Dam GJ, Deelder AM, et al. Schistosomiasis and infection with human immunodeficiency virus 1 in rural Zimbabwe: Systemic inflammation during co‐infection and after treatment for schistosomiasis. American Journal of Tropical Medicine and Hygiene 2008;79(3):331‐7.

Esan 2013 {published data only}

Esan MO, van Hensbroek MB, Nkhoma E, Musicha C, White SA, Ter Kuile FO, et al. Iron supplementation in HIV‐infected Malawian children with anemia: a double‐blind, randomized, controlled trial. Clinical Infectious Diseases 2013;57(11):1626‐34.

Finkelstein 2012 {published data only}

Finkelstein JL, Mehta S, Duggan CP, Spiegelman D, Aboud S, Kupka R, et al. Predictors of anaemia and iron deficiency in HIV‐infected pregnant women in Tanzania: a potential role for vitamin D and parasitic infections. Public Health Nutrition 2012;15(5):928‐37.

Fischer 1995 {published data only}

Fischer P, Kipp W, Kabwa P, Buttner DW. Onchocerciasis and human immunodeficiency virus in western Uganda: prevalences and treatment with ivermectin. American Journal of Tropical Medicine and Hygiene 1995;53(2):171‐8.

Gallagher 2005 {published data only}

Gallagher M, Malhotra I, Mungai PL, Wamachi AN, Kioko JM, Ouma JH, et al. The effects of maternal helminth and malaria infections on mother‐to‐child HIV transmission. AIDS 2005;19(16):1849‐55.

Ganley‐Leal 2006 {published data only}

Ganley‐Leal LM, Mwinzi PN, Cetre‐Sossah CB, Andove J, Hightower AW, Karanja DM, et al. Correlation between eosinophils and protection against reinfection with Schistosoma mansoni and the effect of human immunodeficiency virus type 1 coinfection in humans. Infection and Immunity 2006;74(4):2169‐76.

Geelhoed 2006 {published data only}

Geelhoed D, Agadzi F, Visser L, Ablordeppey E, Asare K, O'Rourke P, et al. Severe anemia in pregnancy in rural Ghana: a case‐control study of causes and management. Acta Obstetricia et Gynecologica Scandinavica 2006;85(10):1165‐71.

Heath 1996 {published data only}

Heath T, Riminton S, Garsia R, MacLeod C. Systemic strongyloidiasis complicating HIV: a promising response to ivermectin. International Journal of STD & AIDS 1996;7(4):294‐6.

Hosseinipour 2007 {published data only}

Hosseinipour MC, Napravnik S, Joaki G, Gama S, Mbeye N, Banda B, et al. HIV and parasitic infection and the effect of treatment among adult outpatients in Malawi. The Journal of Infectious Diseases 2007;195(9):1278‐82.

Ivan 2015 {published data only}

Ivan E, Crowther NJ, Mutimura E, Rucogoza A, Janssen S, Njunwa KK, et al. Effect of deworming on disease progression markers in HIV‐1‐infected pregnant women on antiretroviral therapy: a longitudinal observational study from Rwanda. Clinical Infectious Diseases 2015;60(1):135‐42.

Joseph 2004 {published data only}

Joseph S, Jones FM, Laidlaw ME, Mohamed G, Mawa PA, Namujju PB, et al. Impairment of the Schistosoma mansoni‐specific immune responses elicited by treatment with praziquantel in Ugandans with HIV‐1 coinfection. The Journal of Infectious Diseases 2004;190(3):613‐8.

Kallestrup 2006 {published data only}

Kallestrup P, Zinyama R, Gomo E, Butterworth AE, van Dam GJ, Gerstoft J, et al. Schistosomiasis and HIV in rural Zimbabwe: efficacy of treatment of schistosomiasis in individuals with HIV coinfection. Clinical Infectious Diseases 2006;42(12):1781‐9.

Karanja 1998 {published data only}

Karanja DM, Boyer AE, Strand M, Colley DG, Nahlen BL, Ouma JH, et al. Studies on schistosomiasis in western Kenya: II. Efficacy of praziquantel for treatment of schistosomiasis in persons coinfected with human immunodeficiency virus‐1. American Journal of Tropical Medicine and Hygiene 1998;59(2):307‐11.

Kassu 2003 {published data only}

Kassu A, Tsegaye A, Wolday D, Petros B, Aklilu M, Sanders EJ, et al. Role of incidental and/or cured intestinal parasitic infections on profile of CD4+ and CD8+ T cell subsets and activation status in HIV‐1 infected and uninfected adult Ethiopians. Clinical Experimental Immunology 2003;132(1):113‐9.

Kipp 2005 {published data only}

Kipp W, Bamhuhiiga J, Rubaale T, Kabagambe G. Adverse reactions to the ivermectin treatment of onchocerciasis patients: does infection with the human immunodeficiency virus play a role?. Annals of Tropical Medicine and Parasitology 2005;99(4):395‐402.

Kleppa 2014 {published data only}

Kleppa E, Ramsuran V, Zulu S, Karlsen GH, Bere A, Passmore JA, et al. Effect of female genital schistosomiasis and anti‐schistosomal treatment on monocytes, CD4+ T‐cells and CCR5 expression in the female genital tract. PLoS One 2014;9(6):e98593.

Lankowski 2014 {published data only}

Lankowski AJ, Tsai AC, Kanyesigye M, Bwana M, Haberer JE, Wenger M, et al. Empiric deworming and CD4 count recovery in HIV‐infected Ugandans initiating antiretroviral therapy. PLoS Neglected Tropical Diseases 2014;8(8):e3036.

Lawn 2000 {published data only}

Lawn SD, Karanja DM, Mwinzi P, Andove J, Colley DG, Folks TM, et al. The effect of treatment of schistosomiasis on blood plasma HIV‐1 RNA concentration in coinfected individuals. AIDS 2000;14(16):2437‐43.

McElroy 2005 {published data only}

McElroy MD, Elrefaei M, Jones N, Ssali F, Mugyenyi P, Barugahare B, et al. Coinfection with Schistosoma mansoni is associated with decreased HIV‐specific cytolysis and increased IL‐10 production. The Journal of Immunology 2005;174(8):5119‐23.

Modjarrad 2005 {published data only}

Modjarrad K, Zulu I, Redden DT, Njobvu L, Lane HC, Bentwich Z, et al. Treatment of intestinal helminths does not reduce plasma concentrations of HIV‐1 RNA in coinfected Zambian adults. The Journal of Infectious Diseases 2005;192(7):1277‐83.

Mulu 2013 {published data only}

Mulu A, Maier M, Liebert UG. Deworming of intestinal helminths reduces HIV‐1 subtype C viraemia in chronically co‐infected individuals. International Journal of Infectious Diseases 2013;17(10):e897–901.

Muok 2013 {published data only}

Muok EM, Simiyu EW, Ochola EA, Ng'ang'a ZW, Secor WE, Karanja DM, et al. Association between CD4+ T‐lymphocyte counts and fecal excretion of Schistosoma mansoni eggs in patients coinfected with S. mansoni and human immunodeficiency virus before and after initiation of antiretroviral therapy. American Journal of Tropical Medicine and Hygiene 2013;89(1):42‐5.

Mwanakasale 2003 {published data only}

Mwanakasale V, Vounatsou P, Sukwa TY, Ziba M, Ernest A, Tanner M. Interactions between Schistosoma haematobium and human immunodeficiency virus type 1: the effects of coinfection on treatment outcomes in rural Zambia. American Journal of Tropical Medicine and Hygiene 2003;69(4):420‐8.

Ndibazza 2012 {published data only}

Ndibazza J, Mpairwe H, Webb EL, Mawa PA, Nampijja M, Muhangi L, et al. Impact of anthelminthic treatment in pregnancy and childhood on immunisations, infections and eczema in childhood: a randomised controlled trial. PLoS One 2012;7(12):e50325.

Watanabe 2007 {published data only}

Watanabe K, Mwinzi PN, Black CL, Muok EM, Karanja DM, Secor WE, et al. T regulatory cell levels decrease in people infected with Schistosoma mansoni on effective treatment. American Journal of Tropical Medicine and Hygiene 2007;77(4):676‐82.

Wolday 2002 {published data only}

Wolday D, Mayaan S, Mariam ZG, Berhe N, Seboxa T, Britton S, et al. Treatment of intestinal worms is associated with decreased HIV plasma viral load. Journal of Acquired Immune Deficiency Syndromes 2002;31(1):56‐62.

Zinyama 2009 {published data only}

Zinyama‐Gutsire R, Gomo E, Kallestrup P, Erikstrup C, Ullum H, Butterworth AE, et al. Downregulation of MIP‐1alpha/CCL3 with praziquantel treatment in Schistosoma haematobium and HIV‐1 co‐infected individuals in a rural community in Zimbabwe. BMC Infectious Diseases 2009;9:174.

Zulu 2002 {published data only}

Zulu I, Veitch A, Sianongo S, McPhail G, Feakins R, Farthing MJ, et al. Albendazole chemotherapy for AIDS‐related diarrhoea in Zambia‐‐clinical, parasitological and mucosal responses. Alimentary Pharmacology & Therapeutics 2002;16(3):595‐601.

Baggaley 2015

Baggaley RF, Hollingsworth TD. Brief report: HIV‐1 transmissions during asymptomatic infection: exploring the impact of changes in HIV‐1 viral load due to coinfections. Journal of Acquired Immune Deficiency Syndromes 2015;68(5):594‐8.

Bentwich 1995

Bentwich Z, Kalinkovich A, Weisman Z. Immune activation is a dominant factor in the pathogenesis of African AIDS. Immunology Today 1995;16(4):187‐91.

Bentwich 1996

Bentwich Z, Weisman Z, Moroz C, Bar‐Yehuda S, Kalinkovich A. Immune dysregulation in Ethiopian immigrants in Israel: relevance to helminth infections?. Clinical and Experimental Immunology 1996;103(2):239‐43.

Borkow 2006

Borkow G, Bentwich Z. HIV and helminth co‐infection: is deworming necessary?. Parasite Immunology 2006;28(11):605‐12.

Brown 2006

Brown M, Mawa PA, Kaleebu P, Elliott AM. Helminths and HIV infection: epidemiological observations on immunological hypotheses. Parasite Immunology 2006;28(11):613‐23.

Bundy 2009

Bundy DAP, Kremer M, Bleakley H, Jukes MCH, Miguel E. Deworming and development: asking the right questions, asking the questions right. PLoS Neglected Tropical Diseases 2009;3(1):e362. [10.1371/ journal.pntd.0000362]

Chachage 2014

Chachage M, Podola L, Clowes P, Nsojo A, Bauer A, Mgaya O, et al. Helminth‐associated systemic immune activation and HIV co‐receptor expression: response to albendazole/praziquantel treatment. PLoS Neglected Tropical Diseases 2014;8(3):e2755.

DerSimonian 1986

DerSimonian R, Laird N. Meta‐analysis in clinical trials. Controlled Clinical Trials 1986;7(3):177‐89.

Eggena 2005

Eggena MP, Barugahare B, Okello M, Mutyala S, Jones N, Ma Y, et al. T cell activation in HIV‐seropositive Ugandans: differential associations with viral load, CD4+ T cell depletion, and coinfection. The Journal of Infectious Diseases 2005;191(5):694‐701.

Fincham 2003

Fincham JE, Markus MB, Adams VJ. Could control of soil‐transmitted helminthic infection influence the HIV/AIDS pandemic. Acta Tropica 2003;86(2‐3):315‐33.

Gordin 1984

Gordin FM, Simon GL, Wofsy CB, Mills J. Adverse reactions to trimethoprim‐sulphamethoxazole in patients with the acquired immunodeficiency syndrome. Annals of Internal Medicine 1984;100(4):495–9.

GRADEpro GDT 2015

GRADEpro GDT: GRADEpro Guideline Development Tool [Software] McMaster University, 2015 (developed by Evidence Prime, Inc.). Available from www.gradepro.org.

Gupta 2007

Gupta SB, Jacobson LP, Margolick JB, Rinaldo CR, Phair JP, Jamieson BD, et al. Estimating the benefit of an HIV‐1 vaccine that reduces viral load set point. The Journal of Infectious Diseases 2007;195(4):546‐50.

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Kalinkovich 1999

Kalinkovich A, Weisman Z, Bentwich Z. Chemokines and chemokine receptors: role in HIV infection. Immunology Letters 1999;68(2‐3):281‐7.

Kalinkovich 2001

Kalinkovich A, Borkow G, Weisman Z, Tsimanis A, Stein M, Bentwich Z. Increased CCR5 and CXCR4 expression in Ethiopians living in Israel: environmental and constitutive factors. Clinical Immunology 2001;100(1):107‐17.

Kallestrup 2005

Kallestrup P, Zinyama R, Gomo E, Butterworth A, van Dam GJ, Erikstrup C, et al. Schistosomiasis and HIV‐1 infection in rural Zimbabwe: implications of coinfection for excretion of eggs. Journal of Infectious Diseases 2005;191(8):1311–20.

Kinter 2007

Kinter AL, Horak R, Sion M, Riggin L, McNally J, Lin Y, et al. CD25+ regulatory T cells isolated from HIV‐infected individuals suppress the cytolytic and nonlytic antiviral activity of HIV‐specific CD8+ T cells in vitro. AIDS Research and Human Retroviruses 2007;23(3):438‐50.

Lawn 2001

Lawn SD, Butera ST, Folks TM. Contribution of immune activation to the pathogenesis and transmission of human immunodeficiency virus type 1 infection. Clinical Microbiology Reviews 2001;14(4):753‐77.

Modjarrad 2008

Modjarrad K, Chamot E, Vermund SH. Impact of small reductions in plasma HIV RNA levels on the risk of heterosexual transmission and disease progression. AIDS 2008;22(16):2179‐85.

Modjarrad 2010

Modjarrad K, Vermund SH. Effect of treating co‐infections on HIV‐1 viral load: a systematic review. The Lancet. Infectious Diseases 2010;10(7):455‐63.

Nesheim 2007

Nesheim SR, Kapogiannis BG, Soe MM, Sullivan KM, Abrams E, Farley J, et al. Trends in opportunistic infections in the pre‐ and post‐highly active antiretroviral therapy eras among HIV‐infected children in the Perinatal AIDS Collaborative Transmission Study, 1986‐2004. Pediatrics 2007;120(1):100‐9.

Nunn 1991

Nunn P, Kibuga D, Gathua S, Brindle R, Imalingat A, Wasunna K, et al. Cutaneous hypersensitivity reactions due to thiacetazone in HIV‐1 seropositive patients treated for tuberculosis. The Lancet 1991;377(8742):627–30.

Partnership for Child Development 1998

Partnership for Child Development. Cost of school‐based treatment in Tanzania. Health Policy and Planning 1998;13(4):384‐96.

RevMan 2014 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Secor 2003

Secor WE, Shah A, Mwinzi PM, Ndenga BA, Watta CO, Karanja DM. Increased density of human immunodeficiency virus type 1 coreceptors CCR5 and CXCR4 on the surfaces of CD4(+) T cells and monocytes of patients with Schistosoma mansoni infection. Infection and Immunity 2003;71(11):6668–71.

Seden 2013

Seden K, Khoo S, Back D, Prevatt N, Lamorde M, Byakika‐Kibwika P, et al. Drug–drug interactions between antiretrovirals and drugs used in the management of neglected tropical diseases: important considerations in the WHO 2020 Roadmap and London Declaration on Neglected Tropical Diseases. AIDS 2013;27(5):675‐86.

Shapira‐Nahor 1998

Shapira‐Nahor O, Kalinkovich A, Weisman Z, Greenberg Z, Nahmias J, Shapiro M, et al. Increased susceptibility to HIV‐1 infection of peripheral blood mononuclear cells from chronically immune‐activated individuals. AIDS 1998;12(13):1731‐3.

UNAIDS 2007

UNAIDS. 2007 AIDS Epidemic Update. http://data.unaids.org/pub/EPISlides/2007/2007_epiupdate_en.pdf (accessed 1 August 2015).

Uniting to Combat NTDs 2012

Uniting to Combat NTDs. The London Declaration. http://unitingtocombatntds.org/sites/default/files/resource_file/london_declaration_on_ntds.pdf (accessed 1 August 2015).

Webb 2011

Webb EL, Mawa PA, Ndibazza J, Kizito D, Namatovu A, Kyosiimire‐Lugemwa J, et al. Effect of single‐dose anthelmintic treatment during pregnancy on an infant’s response to immunisation and on susceptibility to infectious diseases in infancy: a randomised, double‐blind, placebo‐controlled trial. The Lancet 2011;377(9759):52–62.

WHO 2006

World Health Organization. Preventive chemotherapy in human helminthiasis. Coordinated use of anthelminthic drugs in control interventions: a manual for health professionals and programme managers. Geneva: World Health Organization, 2006.

WHO 2014

WHO. Soil‐transmitted helminth infections: fact sheet. April 2014. http://www.who.int/mediacentre/factsheets/fs366/en/(accessed February 2015).

References to other published versions of this review

Walson 2007a

Walson JL, John‐Stewart G. Treatment of helminth co‐infection in individuals with HIV‐1: a systematic review of the literature. PLoS Neglected Tropical Diseases 2007;1(3):e102.

Walson 2007b

Walson JL, John‐Stewart G. Treatment of helminth co‐infection in HIV‐1 infected individuals in resource‐limited settings. Cochrane Database of Systematic Reviews 2007, Issue 1. [DOI: 10.1002/14651858.CD006419]

Walson 2008

Walson JL, John‐Stewart G. Treatment of helminth co‐infection in HIV‐1 infected individuals in resource‐limited settings. Cochrane Database of Systematic Reviews 2008, Issue 1. [DOI: 10.1002/14651858.CD006419.pub2]

Walson 2009

Walson JL, Herrin BR, John‐Stewart G. Deworming helminth co‐infected individuals for delaying HIV disease progression. Cochrane Database of Systematic Reviews 2009, Issue 3. [DOI: 10.1002/14651858.CD006419.pub3]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Abate 2014 ETH

Methods

Trial design: randomized, double‐blind, placebo‐controlled trial.

Participants: adults with and without HIV infection who were found to be co‐infected with soil‐transmitted helminths (STHs) and tuberculosis (TB). Data of the HIV seropositive cohort only are included in this analysis
Length of follow‐up: relevant outcomes (CD4+ T cell count) were recorded at baseline and after three months.
Monitoring and diagnostics: all helminth co‐infections were diagnosed using triplicate stool samples, direct microscopy, and Kato‐Katz techniques. Participants were routinely tested for human immunodeficiency virus (HIV) at TB treatment centres using rapid test kits, Stat‐Pak, and Unigold.

Participants

Number of participants: a total of 140 helminth‐positive TB participants were enrolled and randomized. 72 participants were randomized to the treatment arm and 68 to the placebo arm. We included only data from the 18 HIV‐positive participants in the treatment arm and 14 HIV‐positive participants in the placebo arm in this Cochrane Review.
Inclusion criteria: newly diagnosed TB participants (15 to 60 years of age) presenting at a university referral hospital and co‐infected with an STH infection.

Exclusion criteria: participants that required hospital admission, were pregnant, infected with Schistosoma spp., displayed symptoms of active helminth infection, or displayed signs of any concomitant chronic or infectious disease other than TB/HIV.

Interventions

Intervention: albendazole treatment (400 mg/day) for 3 consecutive days. All helminth‐positive TB participants, including the placebo group, received deworming treatment at week 12. Randomization occurred 2 weeks following TB treatment.

Control: identical placebo tablets.

Outcomes

Outcomes included in this review: change in CD4+ T cells after three months, adverse events, and mortality events.

Other trial outcomes: the primary outcome of the trial was change in TB score after 2 months. Other secondary outcomes were sputum smear conversion after 2 months, changes in the chest x‐ray pattern at week 12, IgE and eosinophil responses, as well as changes in the frequency of Tregs and IFN‐c, IL‐5, and IL‐10 producing peripheral blood mononuclear cells (PBMCs) after 3 months.

Notes

Location: Gondar, Ethiopia.

Participant helminth status: ascertained.

Participant antiretroviral (ART) status: 94% and 100% of participants were on ART in the intervention and placebo groups, respectively.

Note: this is the only trial in which all participants had a major co‐infection beyond the helminth and HIV relationship of interest, which might limit comparability to other findings in this review.

Author contact: we requested additional data for the HIV cohort alone from the trial authors, who provided the data.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Random numbers were generated in a block size of eight by the Addis Continental Institute of Public Health, Ethiopia."

Allocation concealment (selection bias)

Low risk

"All tablets looked identical and were assigned a treatment code by the manufacturer.The treatment allocated for each patient was concealed in an individual envelope."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Both the investigators and clinic staff were blinded to the treatment given."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The treatment code was kept in a sealed envelope at the manufacturer and opened after the last patient had been to a follow‐up visit and the data had been analysed."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No risk of attrition; 15% lost to follow‐up with 72 participants in the intervention group and 68 participants in the control group.

Kallestrup 2005 ZWE

Methods

Trial design: randomized, unblinded, controlled trial.

Participants: adults with and without HIV‐1 infection who were found to be infected with schistosomes. We only included data from the HIV seropositive cohort in this analysis.

Length of follow‐up: HIV‐1 RNA and CD4+ T cell counts were measured at baseline and after 3 months.

Monitoring and diagnostics: from Kallestrup 2005; Status for HIV was determined by a rapid HIV‐1/2 test in the field on a dry blood spot and two enzyme‐linked immunosorbent assays on serum. Infection with Schistosoma haematobium was diagnosed by microscopic identification and quantification of fixed‐volume urine samples filtered on Nytrel filters. Diagnosis of infection with Schistosoma mansoni and other helminth eggs or parasites was assessed by the modified formol‐ether concentration technique on 1 g of stool from each participant.

Participants

Number of participants: 287 individuals were enrolled of whom we included 130 with HIV‐1 and schistosome co‐infection in this analysis. 64 participants received early praziquantel treatment and 66 received delayed treatment.

Inclusion criteria: HIV‐1 and schistosomiasis co‐infection, HIV‐negative schistosomiasis infected, HIV‐1 negative but schistosomiasis infected, or neither infection.

Exclusion criteria: pregnant women and participants presenting with clinical signs/symptoms of TB, terminal stages of schistosomiasis, or severe anaemia.

Interventions

Intervention: participants received a single oral dose of praziquantel (40 mg/kg) at enrolment or after a delay of 3 months.

Control: participants received a single oral dose of praziquantel (40 mg/kg) after a delay of 3 months following enrolment.

Outcomes

Outcomes included in this review: changes in plasma HIV‐1 RNA levels, CD4+ T cell count, and haemoglobin levels between individuals randomized to early versus delayed treatment.

Other trial outcomes: none

Notes

Location: Shamva District, Zimbabwe.

Participant helminth status: ascertained.

Participant ART status: not stated.

Author contact: we requested additional haemoglobin data from the trial authors for this update, who provided the data.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Exact random sequence generation used unclear. From Kallestrup 2005: "Allocation of participants to these 3 control groups was done randomly: 1 participant with schistosomiasis was selected for every 2 coinfected participants, and 1 HIV‐1–positive participant or 1 healthy participant was selected for every 4 coinfected participants."

Allocation concealment (selection bias)

High risk

Allocation concealment was not done. "On inclusion, all participants infected with schistosomes within each HIV‐1 group were openly randomised".

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Non‐blinded; "On inclusion, all participants infected with schistosomes within each HIV‐1 group were openly randomised into 2 equally sized groups".

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

From Kallestrup 2005: "At all stages, when performing parasitological examinations, the technician was blinded to clinical and serological information".

Incomplete outcome data (attrition bias)
All outcomes

High risk

There is a risk of attrition as there was < 80% follow‐up.

Kelly 1996 ZMB

Methods

Trial design: randomized double blind placebo controlled trial.

Participants: HIV‐1 or HIV‐2 seropositive adults with persistent diarrhoea.

Length of follow‐up: adverse and mortality events were recorded over 6 months of follow‐up.

Monitoring and diagnostics: ELISA tests were used to determine HIV seropositivity. Home care staff noted any potential adverse effects.

Participants

Number of participants: 174 participants were initially randomized but only 138 participants were followed‐up after 1 month and considered correctly randomized, with 69 in the intervention group and 69 in the placebo‐controlled group.

Inclusion criteria: HIV‐positive adults with persistent diarrhoea (defined as loose but not bloody stools 3 or more times a day for 3 weeks or longer).

Exclusion criteria: participants were excluded if they had received antibiotics in the preceding week, or were deteriorating clinically (Karnofsky score ≤ 20).

Interventions

Intervention: 800 mg albendazole twice daily for 14 days for treatment of persistent diarrhoea in HIV‐positive participants.

Control: identical placebos twice daily for 14 days.

Outcomes

Outcomes included in this review: incidence of adverse events and mortality. Incidence of adverse events defined as exacerbated diarrhoea, cutaneous reaction, dizziness, headache, cough, and difficulty swallowing.

Other trial outcomes: proportion of time periods during which diarrhoea was experienced after completion of treatment and proportion of participants with full remission after completion of treatment.

Notes

Location: 3 urban hospitals/health centres in Zambia.

Participant helminth status: not ascertained.

Participant ART status: not specified, but the study took place before treatment was widely available in Zambia.

Author contact: we did not contact the trial authors to provide additional information.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"the patients were randomised by allocation of a study pack containing either albendazole or placebo, which had been prepared in London according to a randomisation code."

Allocation concealment (selection bias)

Low risk

"The code (constructed so that the numbers of patients randomised to albendazole and placebo balanced every 20 patients) was kept in London during the study".

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Each pack contained 112 tablets of albendazole or placebo, which were indistinguishable."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear whether or not the assessor was blinded.

Incomplete outcome data (attrition bias)
All outcomes

High risk

There is a risk of attrition as there was < 80% follow‐up.

Nielsen 2007 TZA

Methods

Trial design: randomized double‐blind placebo‐controlled cross‐over trial.

Participants: HIV‐positive individuals with or without Wuchereria bancrofti co‐infection.

Length of follow‐up: all subjects were followed‐up at 1, 12, 13, and 24 weeks after the 1st round of treatment. The data presented in the published manuscript considered the 12‐week visit as the initial visit and the 24‐week visit as the final visit. Individuals treated at the initial visit with diethylcarbamazine (DEC) were then considered as the 'placebo' arm and those who were not treated until the 12‐week visit were the 'treatment' arm. For the purposes of this analysis, we considered only individuals with confirmed HIV‐1 infection who were also filariasis‐infected at the baseline visit. We considered individuals treated with DEC at the initial visit to be in the 'treatment' arm and those who did not receive treatment to be in the 'placebo' arm. We reported outcomes as indicated for the 12‐week visit. As all participants were treated with DEC by the 12 week visit, we did not include data from the 24‐week follow‐up period.

Monitoring and diagnostics: diagnosis of infection with lymphatic filariasis (LF) infection with W. bancrofti was performed using immunochromatographic tests (ICT) followed by ELISA testing for circulating filarial antigens (CFA) in serum samples. Participants were also screened for malaria and intestinal helminth eggs.

Participants

Number of participants: the trial authors screened 858 adults and 34 HIV‐1 infected individuals were enrolled and randomized in the trial, of which 27 were followed‐up. Eighteen were co‐infected with W. bancrofti, and 16 were not co‐infected. In the co‐infected group, 10 participants were randomized to early treatment and 8 participants were randomized to delayed treatment. Twelve of these participants were followed‐up, 6 in each treatment group. The HIV RNA from one of the participants in the delayed treatment arm could not be amplified.

Inclusion criteria: HIV‐positive individuals without clinical manifestations of HIV, with and without LF infection.

Exclusion criteria: none specified.

Interventions

Intervention: DEC (6 mg/kg) at randomization.

Control: equivalent placebo and treatment after a delay of 3 months.

Outcomes

Outcomes included in this review: plasma HIV‐1 RNA levels, CD4+ cell count, CD4 percent, serum concentrations of ferritin, adverse events.

Other trial outcomes: CD4/CD8 ratio between individuals randomized to early versus delayed treatment (3 months later) (Nielsen 2007 TZA). Baseline and change in serum retinol, β‐carotene, α‐tocopherol, and the acute phase reactant a‐1 antichymotrypsin after 3 months (Nielsen 2009).

Notes

Location: Northeastern Tanzania

Participant helminth status: ascertained

Participation ART status: the trial authors did not specify if any participants were receiving ART treatment at the start or during the trial.

Author contact: we requested additional CD4 and HIV‐1 RNA data from the trial authors, who provided the data.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Individuals were randomised (1:1). "The identical DEC and placebo tablets were packed in containers with different color codes (“red” or “blue”). Individuals were randomised (1:1) to receive treatment in the order of “red” followed by “blue” or “blue” followed by “red” by using a list of random numbers.The selected study participants were listed and numbered from 1 to 34, and the first 17 numbers (between 1 and 34) encountered in the table (when starting on a randomly chosen figure) were assigned to receive treatment in the order red–blue, and the remaining 17 received treatment in the order blue–red."

Allocation concealment (selection bias)

Unclear risk

This was not specified.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"All study personnel and participants were blinded to treatment assignment throughout the study."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear whether or not the assessor was blinded.

Incomplete outcome data (attrition bias)
All outcomes

High risk

There is a risk of attrition as there was < 80% follow‐up. However, this reflects 7 participants who were lost to follow‐up and therefore excluded from the analyses. The excluded participants did not significantly differ at baseline from the included participants.

Suputtamongkol 2011 THA

Methods

Trial design: randomized, non‐blinded study.

Participants: adult participants with chronic strongyloidiasis.

Length of follow‐up: participants were followed up 2 weeks after treatment initiation, then 1 month, 3 months, 6 months, 9 months, and 1 year post‐treatment.

Monitoring and diagnostics: infection with Strongyloides stercoralis was ascertained using the direct smear, formol‐ether concentration method, and modified Koga agar plate culture method.

Participants

Number of participants: 90 adult participants with chronic Strongyloides infection were recruited. There were 10 participants with HIV co‐infections, with 3 HIV‐positive participants randomized to albendazole, 2 HIV‐positive participants randomized to single dose ivermectin, and 5 HIV‐positive participants randomized to double dose ivermectin.

Inclusion criteria: adult participants with characteristic rhabditiform larvae of S. stercoralis present on faecal microscopy.

Exclusion criteria: history of allergic reaction to either study medication, treatment within the month prior to the trial with any drug known to have anti‐Strongyloides activity, pregnancy, or lactation, and any suggestion of disseminated strongyloidiasis.

Interventions

Intervention: Group 1: ivermectin delivered as a single dose of 200 µg/kg; Intervention. Group 2: 2 doses of ivermectin (200 µg/kg) delivered 2 weeks apart. For the purpose of this analysis we considered both groups that received ivermectin together.

Control: participants received 7 days of albendazole (800 mg per day).

Outcomes

Outcomes included in this review: incidence of adverse events defined as "symptoms or signs that developed after the study drug administration and had not been reported prior to the administration of the first dose of the antihelmintic."

Other trial outcomes: treatment cure (defined as clinical improvement (if symptomatic before treatment) and the absence of rhabditiform larvae in the stool at day 14 of treatment and throughout the follow‐up period) and treatment failure (defined as the presence of larvae two weeks after initiation of treatment or the reappearance of larvae during follow‐up).

Notes

Location: Siriraj Hospital, Thailand

Participant helminth status: ascertained

Participant ART status: it was unspecified if any individuals were receiving ART treatment at the start or during the trial.

Author contact: we requested additional data regarding the incidence of adverse events in the HIV‐positive participants specifically from the trial authors, who provided this information.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Computer generated, simple, random allocation sequences were prepared for 3 study groups by the investigator team."

Allocation concealment (selection bias)

Low risk

"These were sealed in an opaque envelope and numbered. The investigator assigned study participants to their respective treatment group after opening the sealed envelope."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"prospective open‐label, randomised, controlled study".

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear whether or not the assessor was blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There was 10% loss to follow‐up; "Ten patients were excluded from analysis because they did not receive or complete the study treatment (3 in albendazole group, 2 in ivermectin‐II group), or they were lost to follow‐up immediately after treatment (3 in albendazole group, 1 each in ivermectin‐I and ivermectin‐II respectively)."

Walson 2008 KEN

Methods

Trial design: randomized double‐blind placebo‐controlled trial.

Partcipants: HIV‐1 positive adults with evidence of co‐infection with albendazole‐treatable STHs.

Length of follow‐up: all subjects were followed‐up at 12 weeks post‐randomization.

Monitoring and diagnostics: helminth diagnosis was performed using stool samples processed and evaluated with wet preparation, Kato‐Katz, and formol‐ether concentration techniques. HIV‐1 was diagnosed using Determine™ rapid test qualitative immunoassay. The CD4 lymphocyte count was determined using Multiset™ software on a FACSCalibur machine. Plasma HIV‐1 RNA was quantified using the Gen‐Probe HIV‐1 viral load assay.

Participants

Number of participants: the trial screened 1551 adults attending HIV care clinics and 299 were infected with at least 1 helminth species. Regarding enrolment, 234 ART‐naive individuals were enrolled, of whom 208 HIV and STH co‐infected (hookworm, Ascaris, Trichuris, or Strongyloides) individuals were included in the final analysis, 108 were randomized to receive early treatment and 100 to receive placebo.

Inclusion criteria: HIV‐1 seropositive adults, not pregnant, and ineligible for initiation of ART based on WHO guidelines (CD4 < 200 cells/mm³, any stage 4 and some stage 3 disease).

Exclusion criteria: ever used ART drugs, took medicine for helminth infection in the preceding 6 months, evidence of active TB or TB treatment in the past 3 months, and clinical signs of severe anaemia.

Interventions

Intervention: albendazole (400 mg per day) for 3 days versus placebo at enrolment.

Control: placebo at enrolment. After a delay of 3 months, all participants showing evidence of helminth infection were treated with albendazole, regardless of randomization arm.

Outcomes

Outcomes included in this review: changes in plasma HIV‐1 RNA levels and CD4+ count between individuals randomized to early versus delayed treatment (3 months later) and adverse events.

Other trial outcomes: none.

Notes

Location: 10 sites throughout Kenya.

Participant helminth status: ascertained.

Participant ART status: treatment naive.

Author contact: the trial author provided additional CD4 and HIV‐1 RNA data for inclusion in this Cochrane Review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Participants were randomly assigned to two groups using a 1:1 allocation scheme with block randomisation of 30 patients and following a random‐allocation list generated independently."

Allocation concealment (selection bias)

Low risk

"Pre‐labeled, sequentially numbered treatment packs were used. Both the active drug (albendazole) and an identical appearing placebo were provided by the drug manufacturer."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Investigators, clinic staff and patients were blinded to study‐group assignment."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear whether or not the assessor was blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No risk of attrition; 0.5% lost to follow‐up, with 4 in the intervention arm and 5 in the control arm.

Walson 2012 KEN

Methods

Trial design: non‐blinded randomized study.

Partcipants: HIV‐positive ART naive adults in Kenya.

Length of follow‐up: CD4+ cell counts measured every 6 months and plasma RNA measured every 12 months. Participants were followed for 24 months.

Monitoring and diagnostics: HIV serological testing was conducted with Determine™ rapid tests, CD4 cell counts using a FACSCalibur™, plasma HIV RNA assays with the COBAS® Amplicor assay.

Participants

Number of participants: 979 individuals were screened for enrolment in the trial and 917 individuals were enrolled and eligible, with 449 participants randomized to the treatment group and 468 randomized to the control group. All participants were administered cotrimoxazole prophylaxis.

Inclusion criteria: adults ≥ 18 years, HIV seropositive, and do not meet criteria for ART initiation (on the basis of documented WHO disease stage and CD4+ cell count within the previous 3 months and a clinical assessment at enrolment).

Exclusion criteria: pregnancy, reports of taking antihelminthics in the previous 6 months, reports of having previously received ART ((except for the prevention of mother‐to‐child transmission).

Interventions

Intervention: empiric deworming with repeat single‐dose albendazole (400 mg) given every 3 months plus single dose praziquantel (25 mg/kg) given annually. Participants were excluded if they missed two or more consecutive doses of study drug.

Control: standard of care.

Outcomes

Outcomes included in this review: changes in plasma HIV‐1 RNA levels, CD4+ cell count, non‐traumatic death, and adverse events between individuals randomized to treatment versus no intervention.

Other trial outcomes: time to CD4+ count of < 350 cells/μL and first occurrence of any of the following: CD4+ count < 350 cells/μL, first reported use of ART (excluding that used for the prevention of mother‐to‐child transmission), or non‐traumatic death. Secondary analyses included time to death and ART initiation separately.

Notes

Location: 3 sites in Kenya

Participant helminth status: helminth status of participants was not evaluated at baseline.

Participant ART status: treatment naive

Author contact: the trial author provided additional CD4+ and HIV‐1 RNA data for inclusion in this Cochrane Review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Using a computer‐generated randomisation sequence, we assigned participants (1:1) to either the treatment group or control group."

Allocation concealment (selection bias)

Low risk

"We used a computerised database to ensure that treatment allocation was not disclosed to study staff and participants until randomisation was complete."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial participants and personnel were not blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear whether or not the assessor was blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There is no risk of attrition; 4.2% of participants were lost to follow‐up with 16 in the intervention arm and 24 in the control arm.

Webb 2012 UGA

Methods

Trial design: 2 x 2 randomized double‐blind placebo controlled trial.

Partcipants: HIV‐positive ART‐naive pregnant women.

Length of follow‐up: viral load was measured 6 weeks post‐treatment and at delivery. However, this analysis included data at 6 weeks post‐treatment only in order to minimize the effects of prenatal HIV care provided to study participants.

Monitoring and diagnostics: stool samples were processed and examined for helminth ova using a duplicate Kato‐Katz method and by charcoal culture for Strongyloides.

Participants

Number of participants: 2507 women were enrolled in the parent study (EMaBS), of whom 299 tested positive for HIV. Of these, 222 participants were randomized and followed‐up to 6 weeks post‐enrolment. Sixty‐four participants were randomized to single‐dose albendazole, 54 participants to praziquantel, 67 participants to both albendazole and praziquantel, and 70 participants to placebo only.

Inclusion criteria: pregnant women presenting at the government‐funded antenatal clinic at Entebbe General Hospital, who were resident in the study area, planning to deliver in the hospital, willing to know their HIV status, and in the 2nd or 3rd trimester of pregnancy.

Exclusion criteria: evidence of possible helminth‐induced pathology (haemoglobin < 8 g/dL, clinically apparent severe liver disease, diarrhoea with blood in stool), history of adverse reaction to antihelminthics, prior enrolment in an earlier pregnancy, or abnormal pregnancy as assessed by the midwife.

Interventions

Intervention 1: albendazole (400 mg).

Intervention 2: praziquantel (40 mg/kg).

Intervention 3: both albendazole (400 mg) and praziquantel (40 mg/kg).

Control: equivalent placebos.

Outcomes

Outcomes included in this review: changes in plasma HIV‐1 RNA levels 6 weeks post‐treatment, adverse events (defined as post‐treatment hospitalizations), and mortality.

Other trial outcomes: primary outcomes for EMaBS were response to immunisation and incidence of infectious diseases in the offspring, and vertical transmission of HIV (Webb 2011). Secondary outcomes in this analysis included viral load at delivery.

Notes

Location: Entebbe, Uganda.

Participant helminth status: ascertained.

Participant ART status: ART naive. However, in accordance with guidelines at the time, HIV‐positive women were counselled and given intrapartum and neonatal single dose nevirapine for prevention of mother‐to‐child HIV transmission. After enrolment, women received standard antenatal care, including haematinics, tetanus immunization, and intermittent presumptive treatment for malaria.

Author contact: we requested additional HIV‐1 RNA and adverse events data from the trial authors, who provided this information.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Women were then randomised in a 1:1:1:1 ratio to single‐dose albendazole 400 mg or matching placebo and praziquantel 40 mg/kg or matching placebo in a 2×2 factorial design. The randomisation code was generated by the trial statistician in blocks of 100."

Allocation concealment (selection bias)

Low risk

"Sealed envelopes containing the study intervention were prepared by colleagues at the Medical Research Council Unit in Entebbe with no other involvement in the trial. Treatments were allocated in numerical order by trained interviewer‐counsellors and taken under observation."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"All participants and staff were blinded to treatment allocation."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear whether or not the assessor was blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There is no risk of attrition; 10% of participants were lost to follow‐up.

Abbreviations: antiretroviral treatment (ART), circulating filarial antigens (CFA), diethylcarbamazine (DEC), human immunodeficiency virus (HIV), immunochromatographic tests (ICT), soil‐transmitted helminths (STHs), tuberculosis (TB)

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Blish 2010

This manuscript presents further analyses using data already presented in Walson 2008 KEN

Brown 2004

Observational, prospective cohort study.

Brown 2005

Observational, prospective cohort study.

Elliott 2003

Observational, prospective cohort study.

Elliott 2007

Study protocol for included study Webb 2012 UGA.

Erikstrup 2008

This manuscript presents further analyses using data already presented in Kallestrup 2005 ZWE.

Esan 2013

A RCT with non‐relevant intervention (that is, iron supplementation and multivitamins) to assess effects of anaemia in children 6 to 59 months of age.

Finkelstein 2012

Observational, prospective cohort study.

Fischer 1995

Observational, prospective cohort study.

Gallagher 2005

Observational, retrospective cohort study.

Ganley‐Leal 2006

Observational, prospective cohort study.

Geelhoed 2006

Observational, prospective case‐control study, with irrelevant outcomes to this Cochrane Review.

Heath 1996

Case study of a single participant.

Hosseinipour 2007

Observational, cross‐sectional study.

Ivan 2015

Observational, cross‐sectional study.

Joseph 2004

Observational, prospective cohort study.

Kallestrup 2006

This manuscript presents further analyses using data already presented in Kallestrup 2005 ZWE.

Karanja 1998

Observational, prospective cohort study.

Kassu 2003

Observational, prospective cohort study.

Kipp 2005

Observational, prospective cohort study.

Kleppa 2014

Observational, case‐control study.

Lankowski 2014

Observational retrospective cohort study.

Lawn 2000

Observational, prospective cohort study with no comparison group.

McElroy 2005

Observational, cross‐sectional study.

Modjarrad 2005

Observational, prospective cohort study.

Mulu 2013

Observational, prospective cohort study.

Muok 2013

Observational, prospective cohort study

Mwanakasale 2003

Observational, prospective cohort study.

Ndibazza 2012

A RCT with irrelevant outcomes to this Cochrane Review. The study authors hypothesize that antihelminthic treatment in pregnancy and early childhood would improve responses to immunization and modulate disease incidence in early childhood.

Watanabe 2007

Observational, prospective cohort study.

Wolday 2002

Observational, prospective cohort study.

Zinyama 2009

Observational, prospective cohort study.

Zulu 2002

Observational, prospective cohort study.

Abbreviations: RCT: randomized controlled trial.

Data and analyses

Open in table viewer
Comparison 1. Deworming drugs versus placebo in people with unknown helminth infection status

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Viral load: change in log10 HIV‐1 RNA Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Deworming drugs versus placebo in people with unknown helminth infection status, Outcome 1 Viral load: change in log10 HIV‐1 RNA.

Comparison 1 Deworming drugs versus placebo in people with unknown helminth infection status, Outcome 1 Viral load: change in log10 HIV‐1 RNA.

1.1 At 6 weeks after one dose

1

166

Mean Difference (IV, Random, 95% CI)

‐0.14 [‐0.35, 0.07]

1.2 At 2 years after multiple doses

1

917

Mean Difference (IV, Random, 95% CI)

0.01 [‐0.03, 0.05]

2 Change in CD4 count Show forest plot

1

917

Mean Difference (IV, Random, 95% CI)

2.60 [‐10.15, 15.35]

Analysis 1.2

Comparison 1 Deworming drugs versus placebo in people with unknown helminth infection status, Outcome 2 Change in CD4 count.

Comparison 1 Deworming drugs versus placebo in people with unknown helminth infection status, Outcome 2 Change in CD4 count.

2.1 At 2 years after multiple doses

1

917

Mean Difference (IV, Random, 95% CI)

2.60 [‐10.15, 15.35]

Open in table viewer
Comparison 2. Deworming drugs versus placebo in people with confirmed helminth infection

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Viral load: change in log10 HIV‐1 RNA Show forest plot

4

445

Mean Difference (IV, Random, 95% CI)

‐0.13 [‐0.26, ‐0.00]

Analysis 2.1

Comparison 2 Deworming drugs versus placebo in people with confirmed helminth infection, Outcome 1 Viral load: change in log10 HIV‐1 RNA.

Comparison 2 Deworming drugs versus placebo in people with confirmed helminth infection, Outcome 1 Viral load: change in log10 HIV‐1 RNA.

1.1 Soil‐transmitted helminths

2

273

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.31, 0.12]

1.2 Schistosomiasis

2

161

Mean Difference (IV, Random, 95% CI)

‐0.13 [‐0.36, 0.10]

1.3 Lymphatic filariasis

1

11

Mean Difference (IV, Random, 95% CI)

‐0.08 [‐0.93, 0.77]

2 Change in CD4 count Show forest plot

3

358

Mean Difference (IV, Random, 95% CI)

37.86 [7.36, 68.35]

Analysis 2.2

Comparison 2 Deworming drugs versus placebo in people with confirmed helminth infection, Outcome 2 Change in CD4 count.

Comparison 2 Deworming drugs versus placebo in people with confirmed helminth infection, Outcome 2 Change in CD4 count.

2.1 Soil‐transmitted helminths

2

228

Mean Difference (IV, Random, 95% CI)

40.69 [1.51, 79.87]

2.2 Schistosomiasis

1

130

Mean Difference (IV, Random, 95% CI)

33.5 [‐15.06, 82.06]

Open in table viewer
Comparison 3. Deworming drugs versus placebo; all trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in log10 HIV RNA, by antihelminthic Show forest plot

5

1534

Mean Difference (IV, Random, 95% CI)

‐0.09 [‐0.19, 0.01]

Analysis 3.1

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 1 Change in log10 HIV RNA, by antihelminthic.

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 1 Change in log10 HIV RNA, by antihelminthic.

1.1 Albendazole

2

302

Mean Difference (IV, Random, 95% CI)

‐0.18 [‐0.37, 0.02]

1.2 Praziquantel

2

213

Mean Difference (IV, Random, 95% CI)

‐0.17 [‐0.32, ‐0.03]

1.3 Diethylcarbamazine

1

11

Mean Difference (IV, Random, 95% CI)

‐0.08 [‐0.93, 0.77]

1.4 Albendazole and praziquantel

2

1008

Mean Difference (IV, Random, 95% CI)

0.01 [‐0.04, 0.05]

2 Difference in adverse events between treatment and no treatment groups Show forest plot

7

1649

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.53, 2.83]

Analysis 3.2

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 2 Difference in adverse events between treatment and no treatment groups.

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 2 Difference in adverse events between treatment and no treatment groups.

3 Difference in mortality events between treatment and no treatment groups Show forest plot

5

1627

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.52, 1.14]

Analysis 3.3

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 3 Difference in mortality events between treatment and no treatment groups.

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 3 Difference in mortality events between treatment and no treatment groups.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

'Risk of bias' summary: review authors' judgements about each 'Risk of bias' item for each included trial.
Figuras y tablas -
Figure 2

'Risk of bias' summary: review authors' judgements about each 'Risk of bias' item for each included trial.

'Risk of bias' graph: review authors' judgements about each methodological quality item presented as percentages across all included trials.
Figuras y tablas -
Figure 3

'Risk of bias' graph: review authors' judgements about each methodological quality item presented as percentages across all included trials.

Comparison 1 Deworming drugs versus placebo in people with unknown helminth infection status, Outcome 1 Viral load: change in log10 HIV‐1 RNA.
Figuras y tablas -
Analysis 1.1

Comparison 1 Deworming drugs versus placebo in people with unknown helminth infection status, Outcome 1 Viral load: change in log10 HIV‐1 RNA.

Comparison 1 Deworming drugs versus placebo in people with unknown helminth infection status, Outcome 2 Change in CD4 count.
Figuras y tablas -
Analysis 1.2

Comparison 1 Deworming drugs versus placebo in people with unknown helminth infection status, Outcome 2 Change in CD4 count.

Comparison 2 Deworming drugs versus placebo in people with confirmed helminth infection, Outcome 1 Viral load: change in log10 HIV‐1 RNA.
Figuras y tablas -
Analysis 2.1

Comparison 2 Deworming drugs versus placebo in people with confirmed helminth infection, Outcome 1 Viral load: change in log10 HIV‐1 RNA.

Comparison 2 Deworming drugs versus placebo in people with confirmed helminth infection, Outcome 2 Change in CD4 count.
Figuras y tablas -
Analysis 2.2

Comparison 2 Deworming drugs versus placebo in people with confirmed helminth infection, Outcome 2 Change in CD4 count.

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 1 Change in log10 HIV RNA, by antihelminthic.
Figuras y tablas -
Analysis 3.1

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 1 Change in log10 HIV RNA, by antihelminthic.

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 2 Difference in adverse events between treatment and no treatment groups.
Figuras y tablas -
Analysis 3.2

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 2 Difference in adverse events between treatment and no treatment groups.

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 3 Difference in mortality events between treatment and no treatment groups.
Figuras y tablas -
Analysis 3.3

Comparison 3 Deworming drugs versus placebo; all trials, Outcome 3 Difference in mortality events between treatment and no treatment groups.

Summary of findings for the main comparison. Summary of findings for participants with unknown helminth infection status

Deworming drugs compared with placebo for people with HIV and an unknown helminth infection status

Participant or population: HIV‐positive people

Settings: urban and rural areas co‐endemic for helminths and HIV

Intervention: deworming drugs (albendazole or praziquantel or a combination)

Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Number of participants
(trials)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Placebo

Deworming drugs

Viral load

At 6 weeks after a single dose

166

(1 trial)

⊕⊕⊝⊝1,2,3,4
low

in the control group, the mean change in viral load was an increase of

0.09 log10 viral RNA

On average, with deworming, there was a suppressive effect on mean viral load of

0.14 log10 viral RNA

(0.35 benefit to 0.07 harm)

At 2 years after multiple doses

917

(1 trial)

⊕⊕⊕⊝1,2,5,6
moderate

In the control group, the mean viral load increased by

0.03 log10 viral RNA

On average, with deworming, there was a suppressive effect on mean viral load of

0.01 log10 viral RNA

(0.03 benefit to 0.05 harm)

CD4+ cell count

At 2 years after multiple doses

917

(1 trial)

⊕⊕⊝⊝1,2,4,5
low

In the control group, the mean CD4+ cell count reduced by

37.3 CD4+ cells/µL

On average, with deworming, there was a favourable effect on mean CD4+ cell count of

2.60 CD4+ cells/µL

(15.35 benefit 10.15 harm)

*The basis for the assumed risk (for example, the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
Abbreviations: CI: confidence interval; RR: risk ratio; RNA: ribonucleic acid; HIV: human immunodeficiency virus.

GRADE Working Group grades of evidence
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: 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: we are very uncertain about the estimate.

1No serious risk of bias: this single trial was at low risk of selection bias.
2No serious inconsistency: this was not applicable as there was only a single trial.
3We downgraded by 1 for serious indirectness: this single trial is based upon a specific sample of pregnant women treated with albendazole or praziquantel, or both. The overall finding of an effect cannot be easily generalized to all populations or settings.
4We downgraded by 1 for imprecision: the 95% CI includes potentially clinically important differences as well as no effect. Further larger studies are needed.
5We downgraded by 1 for serious indirectness: this trial was conducted in three sites in rural and urban Kenya. Helminth infection was expected to be high but was not assessed at baseline. The findings are not easily generalized to all helminth endemic settings.
6No serious imprecision: the 95% CI includes no effect but is narrow around the estimate and excludes clinically important differences.

Figuras y tablas -
Summary of findings for the main comparison. Summary of findings for participants with unknown helminth infection status
Summary of findings 2. Summary of findings for participants with confirmed helminth infections

Deworming drugs compared with placebo for people with HIV and confirmed helminth infections

Participant or population: HIV‐positive people with proven helminth infection

Settings: urban and rural areas co‐endemic for helminths and HIV

Intervention: deworming drugs (albendazole, praziquantel, albendazole and praziquantel, or diethylcarbamazine)

Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Number of participants
(trials)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Placebo

Deworming drugs

Viral load

At 6 to 12 weeks

445

(4 trials)

⊕⊕⊝⊝
low1,2,3,4

In the control groups, the mean change in viral load ranged from

a 0.13 decrease to an increase of 0.21 log10 viral RNA

On average, with deworming, there was a small suppressive effect on mean viral load of

0.13 log10 viral RNA

(0.26 benefit to 0.00 benefit)

CD4+ cell count

At 6 to 12 weeks

358

(3 trials)

⊕⊕⊝⊝
low2,5,6,7

In the control groups, the mean change in CD4+ cell count ranged from

a decrease of 68 to an increase of 45 CD4+ cells/µL

On average, with deworming, there was favourable effect on mean CD4+ cell count of

37.86 CD4+ cells/µL

(7.36 benefit to 68.35 benefit)

*The basis for the assumed risk (for example, the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
Abbreviations: CI: confidence interval; RR: risk ratio; RNA: ribonucleic acid; HIV: human immunodeficiency virus.

GRADE Working Group grades of evidence
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: 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: we are very uncertain about the estimate.

1We downgraded by 1 for serious risk of bias: of the five studies, only one had CIs that excluded the possibility of no effect and this study was at high risk of selection bias.
2No serious inconsistency: statistical heterogeneity was low.
3We downgraded by 1 for serious indirectness: the only trial for which the CIs excluded the possibility of no effect administered praziquantel to people with schistosomiasis. The overall finding of an effect cannot be easily generalized to all deworming drugs, helminth infections, or settings.
4No serious imprecision: the overall 95% CI is wide and includes both clinically important effects and no effect. However, as potential harm is excluded, we did not further downgrade the evidence.
5No serious risk of bias: one trial was at high risk of selection bias, but excluding this trial did not substantially change the result.
6We downgraded by 1 for serious indirectness: the included trials are from a very limited number of settings and participants. The overall finding is not easily generalized to all deworming drugs, helminth infections, or settings, and further trials are needed.
7We downgraded by 1 for imprecision: the 95% CI is wide and includes potentially clinically important differences as well as no effect. Further larger studies are needed.

Figuras y tablas -
Summary of findings 2. Summary of findings for participants with confirmed helminth infections
Summary of findings 3. Summary of findings for secondary outcomes

Deworming drugs compared with placebo for people with HIV and both known and unknown helminth infection status

Participant or population: HIV‐positive people

Settings: urban and rural areas co‐endemic for helminths and HIV

Intervention: deworming drugs (albendazole, praziquantel, albendazole and praziquantel, or diethylcarbamazine)

Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

Number of participants
(trials)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Placebo

Albendazole

Iron deficiency (serum ferritin)

log10 mean increase of 0.04 µg/L

log10 mean increase of 0.07 µg/L

Deworming drugs associated with a 0.03 higher µg/L log10 mean ferritin measure (0.30 lower to 0.35 higher)

16

(1 trial)

⊕⊝⊝⊝1,2,3,4
very low

Anaemia (serum haemoglobin)

Increase in 0.15 g/dL

Decrease in 0.10 g/dL

Deworming drugs associated with a 0.25 lower g/dL haemoglobin

(0.58 lower to 0.08 higher)

130

(1 trial)

⊕⊝⊝⊝2,4,5,6
very low

Mortality

41 per 1000

52 per 1000

RR 0.77

(0.52 to 1.14)

1627

(5 trials)

⊕⊕⊝⊝7,8,9,10
low

Adverse events

32 per 1000

35 per 1000

RR 1.23

(0.53 to 2.83)

1649

(7 trials)

⊕⊝⊝⊝4,9,11,12
very low

*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% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
Abbreviations: CI: confidence interval; RR: risk ratio; HIV: human immunodeficiency virus.

GRADE Working Group grades of evidence
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: 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: we are very uncertain about the estimate.

1No serious risk of bias: this single trial was at risk of selection bias. However, the outcome is an objective laboratory measure. We did not downgrade the quality of the evidence.
2No serious inconsistency: not applicable as this is a single trial.
3We downgraded by 2 for serious indirectness: this single trial is based upon a very small sample size of people treated with diethylcarbamazine for lymphatic filariasis. The overall finding of an effect can not be easily generalized to all deworming drugs, helminth infections, or settings.
4We downgraded by 1 for imprecision: the 95% CI is wide and includes potentially clinically important differences as well as no effect. Larger studies are needed.
5We downgraded by 1 for serious risk of bias: this single trial was at high risk of selection bias.
6Downgraded by 1 for serious indirectness: this single trial is based upon a very small sample size of people treated with praziquantel for schistosomiasis. The overall finding of an effect can not be easily generalized to all deworming drugs or helminth infections or settings.
7No serious risk of bias: although some of the trials are at risk of selection bias the outcome is an objective measure. We did not downgrade the evidence.
8No serious inconsistency: statistical heterogeneity was low.
9No serious indirectness: these trials took place in different settings, with different drugs, and with different helminth infections present. However, effects of treatment were consistent across the trials.
10We downgraded by 2 for imprecision: the 95% CI is wide and includes potentially clinically important differences as well as no effect. Additionally the trials were not powered to detect changes in the rare outcome of mortality. A trial in which 3% of controls died would require a sample size of 7648 participants to attain 80% power to detect a 33% reduction in mortality.
11We downgraded by 2 for serious risk of bias: it is unclear how some of the trial authors defined adverse events and thus there is risk of bias in outcome assessment.
12No serious inconsistency: however, the analysis has an I² statistic value of 34%, which may represent low to moderate heterogeneity.

Figuras y tablas -
Summary of findings 3. Summary of findings for secondary outcomes
Table 1. Cochrane librarian search strategy for MEDLINE

Number

Search terms

#1

Search (HIV Infections[MeSH] OR HIV[MeSH] OR hiv[tiab] OR hiv‐1*[tiab] OR hiv‐2*[tiab] OR hiv1[tiab] OR hiv2[tiab] OR hiv infect*[tiab] OR human immunodeficiency virus[tiab] OR human immunedeficiency virus[tiab] OR human immuno‐deficiency virus[tiab] OR human immune‐deficiency virus[tiab] OR ((human immun*[tiab]) AND (deficiency virus[tiab])) OR acquired immunodeficiency syndrome[tiab] OR acquired immunedeficiency syndrome[tiab] OR acquired immuno‐deficiency syndrome[tiab] OR acquired immune‐deficiency syndrome[tiab] OR ((acquired immun*[tiab]) AND (deficiency syndrome[tiab])) OR "sexually transmitted diseases, Viral"[MeSH:NoExp]))

#2

Search (randomized controlled trial [pt] OR controlled clinical trial [pt] OR randomized [tiab] OR placebo [tiab] OR drug therapy [sh] OR randomly [tiab] OR trial [tiab] OR groups [tiab]) NOT (animals [mh] NOT humans [mh])

#3

Search (helminths[mh] OR helminth*[tiab] OR nematode*[tiab] OR worm*[tiab] OR parasites[mh] OR parasit*[tiab] OR round worm*[tiab] OR roundworm*[tiab] OR hookworm*[tiab] OR hook worm*[tiab] OR ancylostoma*[tiab] OR cestode*[tiab] OR tapeworm*[tiab] OR tape worm*[tiab] OR trematode*[tiab] OR fluke*[tiab] OR whipworm*[tiab] OR whip worm*[tiab] OR trichuris[tiab] OR ascaris[tiab] OR enterobi*[tiab] OR strongyloide*[tiab] OR mansonell*[tiab] OR taenia[tiab] schistosom*[tiab] OR necator*[tiab] OR paragonim*[tiab] OR hymenolepis[tiab] OR fasciol*[tiab] OR filariasis[tiab] OR trichostrongl*[tiab] OR microfilaria[tiab] OR parasitic diseases[mh:noexp] OR helminthiasis[mh] OR intestinal diseases, parasitic[mh])

#4

Search (benzimidazoles OR albendazole OR mebendazole OR ivermectin OR praziquantel OR diethylcarbamazine OR bithionol OR oxamniquine OR pyrantel OR nitazoxanide OR anthelmintic OR anthelmintics OR anthelminthic OR anthelminthics OR “anti helminthic” OR “anti helminthics” OR “anti helmintic” OR “anti helmintics” OR antihelminthic OR antihelminthics OR antihelmintic OR antihelmintics)

#5

Search (#1 AND #2 AND #3 AND #4)

#6

Search (((#1 AND #2 AND #3 AND #4))) AND ("1980/01/01"[Date ‐ Publication] : "2015/09/29"[Date ‐ Publication])

Figuras y tablas -
Table 1. Cochrane librarian search strategy for MEDLINE
Table 2. Cochrane librarian search strategy for EMBASE

Number

Search terms

#1

'human immunodeficiency virus infection' exp OR 'human immunodeficiency virus infection':ab,ti OR 'hiv infection':ab,ti OR 'hiv infections':ab,ti OR 'human immunodeficiency virus'/exp OR 'human immunodeficiency virus':ab,ti OR hiv:ab,ti OR 'hiv 1':ab,ti OR 'hiv 2':ab,ti OR 'human immune deficiency virus':ab,ti OR 'human immuno deficiency virus':ab,ti OR 'acquired immunodeficiency syndrome':ab,ti OR 'acquired immuno deficiency syndrome':ab,ti OR 'acquired immune deficiency syndrome':ab,ti OR 'acquired immunedeficiency syndrome':ab,ti

#2

'randomized controlled trial'/de OR 'randomized controlled trial' OR random*:ab,ti OR trial:ti OR allocat*:ab,ti OR factorial*:ab,ti OR placebo*:ab,ti OR assign*:ab,ti OR volunteer*:ab,ti OR 'crossover procedure'/de OR 'crossover procedure'OR 'double‐blind procedure'/de OR 'double‐blind procedure' OR 'single‐blind procedure'/de OR 'single‐blind procedure' OR (doubl*NEAR/3 blind*):ab,ti OR (singl*:ab,ti AND blind*:ab,ti) OR crossover*:ab,ti OR cross+over*:ab,ti OR (crossNEXT/1 over*):ab,ti

#3

'animal'/de OR 'animal experiment'/de OR 'invertebrate'/de OR 'animal tissue'/de OR 'animal cell'/de OR 'nonhuman'/de

#4

'human'/de OR 'normal human'/de OR 'human cell'/de

#5

#3 AND #4

#6

#3 NOT #5

#7

#2 NOT #6

#8

'helminth'/exp OR helminth*:ab,ti OR nematode*:ab,ti OR worm*:ab,ti OR 'parasite'/exp OR parasit*:ab,ti OR roundAND worm*:ab,ti OR roundworm*:ab,ti OR hookworm*:ab,ti OR hookAND worm*:ab,ti OR ancylostoma*:ab,ti OR cestode*:ab,ti OR tapeworm*:ab,ti OR tapeAND worm*:ab,ti OR trematode*:ab,ti OR fluke*:ab,ti OR whipworm*:ab,ti OR whipAND worm*:ab,ti OR trichuris:ab,ti OR ascaris:ab,ti OR enterobi*:ab,ti OR strongyloide*:ab,ti OR mansonell*:ab,ti OR taenia:ab,ti AND schistosom*:ab,ti OR necator*:ab,ti OR paragonim*:ab,ti OR hymenolepis:ab,ti OR fasciol*:ab,ti OR filariasis:ab,ti OR trichostrongl*:ab,ti OR microfilaria:ab,ti OR 'parasitic diseases'/exp OR 'helminthiasis'/de OR 'intestine infection'/de

#9

'benzimidazoles'/de OR benzimidazolesOR 'albendazole'/de OR albendazoleOR 'mebendazole'/de OR mebendazoleOR 'ivermectin'/de OR ivermectinOR 'praziquantel'/de OR praziquantelOR 'diethylcarbamazine'/de OR diethylcarbamazineOR 'bithionol'/de OR bithionolOR 'oxamniquine'/de OR oxamniquineOR 'pyrantel'/de OR pyrantelOR 'nitazoxanide'/de OR nitazoxanideOR 'anthelmintic'/de OR anthelminticOR 'anthelmintics'/de OR anthelminticsOR 'anthelminthic'/de OR anthelminthicOR anthelminthicsOR 'anti helminthic'OR 'anti helminthics'OR 'anti helmintic'OR 'anti helmintics'OR antihelminthicOR antihelminthicsOR 'antihelmintic'/de OR antihelminticOR 'antihelmintics'/de OR antihelmintics

#10

#1 AND #7 AND #8 AND #9

#11

#1 AND #7 AND #8 AND #9 AND [1‐1‐1980]/sd NOT [29‐09‐2015]/sd

Figuras y tablas -
Table 2. Cochrane librarian search strategy for EMBASE
Table 3. Cochrane librarian search strategy for CENTRAL

Number

Search terms

#1

MeSH descriptor: [HIV Infections] explode all trees

#2

MeSH descriptor: [HIV] explode all trees

#3

hiv or hiv‐1* or hiv‐2* or hiv1 or hiv2 or (hiv near infect*) or (human immunodeficiency virus) or (human immunedeficiency virus) or (human immune‐deficiency virus) or (human immuno‐deficiency virus) or (human immune deficiency virus) or (human immuno deficiency virus) or (acquired immunodeficiency syndrome) or (acquired immunedeficiency syndrome) or (acquired immuno‐deficiency syndrome) or (acquired immune‐deficiency syndrome) or (acquired immun* deficiency syndrome) (Word variations have been searched)

#4

MeSH descriptor: [Lymphoma, AIDS‐Related] this term only

#5

MeSH descriptor: [Sexually Transmitted Diseases, Viral] this term only

#6

#1 or #2 or #3 or #4 or #5

#7

[mh helminths] or helminth*:ti,ab,kw or meatode*:ti,ab,kw or worm*:ti,ab,kw or [mh parasites] or parasit*:ti,ab,kw or round worm:ti,ab,kw or roundworm:ti,ab,kw or hook worm*:ti,ab,kw or hookworm*:ti,ab,kw or ancylostoma*:ti,ab,kw or cestode*:ti,ab,kw or tapeworm*:ti,ab,kw or tape worm*:ti,ab,kw or trematode*:ti,ab,kw or fluke*:ti,ab,kw or whipworm*:ti,ab,kw or whip worm*:ti,ab,kw or trichuris:ti,ab,kw or ascaris:ti,ab,kw or enterobi*:ti,ab,kw or strongyloide*:ti,ab,kw or mansonell*:ti,ab,kw or taenia:ti,ab,kw schistosom*:ti,ab,kw or necator*:ti,ab,kw or paragonim*:ti,ab,kw or hymenolepis:ti,ab,kw or fasciol*:ti,ab,kw or filariasis:ti,ab,kw or trichostrongl*:ti,ab,kw or microfilaria:ti,ab,kw or [mh ^"parasitic diseases"] or [mh helminthiasis] or [mh ^"intestinal diseases, parasitic"] (Word variations have been searched)

#8

benzimidazoles or albendazole or mebendazole or ivermectin or praziquantel or diethylcarbamazine or bithionol or oxamniquine or pyrantel or nitazoxanide or anthelmintic or anthelmintics or anthelminthic or anthelminthics or "anti helminthic" or "anti helminthics" or "anti helmintic" or "anti helmintics" or antihelminthic or antihelminthics or antihelmintic or antihelmintics (Word variations have been searched)

#9

#6 and #7 and #8 Publication Year from 1980 to 2015, in Trials

Figuras y tablas -
Table 3. Cochrane librarian search strategy for CENTRAL
Table 4. Cochrane librarian search strategy for the WHO ICTRP

Number

Search terms

#1

HIV AND HELMINTH

Figuras y tablas -
Table 4. Cochrane librarian search strategy for the WHO ICTRP
Table 5. Cochrane librarian search strategy for clinicaltrials.gov

Number

Search terms

#1

HIV AND HELMINTH | Interventional Studies | received from 01/01/1980 to 09/29/2015

Figuras y tablas -
Table 5. Cochrane librarian search strategy for clinicaltrials.gov
Table 6. Search terms used in 2009 newly applied in MEDLINE

Number

Search terms

#1

“HIV Infections”[MeSH] OR “HIV”[MeSH] OR hiv [tw] OR hiv‐1*[tw] OR hiv‐2*[tw] OR hiv1[tw] OR hiv2[tw] OR hiv infect*[tw] OR human immunodeficiency virus[tw] OR human immunedeficiency virus[tw] OR human immunodeficiency virus[tw] OR human immune‐deficiency virus[tw] OR ((human immun*) AND (deficiency virus[tw])) OR acquired immunodeficiency syndrome[tw] OR acquired immunedeficiency syndrome[tw] OR acquired immunodeficiency syndrome[tw] OR acquired immune‐deficiency syndrome[tw] OR ((acquired immun*) AND (deficiency syndrome[tw])) OR “Sexually Transmitted Diseases, Viral”[MeSH:NoExp]

#2

randomized controlled trial [pt] OR controlled clinical trial [pt] OR randomized controlled trials [mh] OR random allocation [mh] OR double‐blind method [mh] OR single‐blind method [mh] OR clinical trial [pt] OR clinical trials [mh] OR (“clinical trial” [tw]) OR ((singl* [tw] OR doubl* [tw] OR trebl* [tw] OR tripl* [tw]) AND (mask* [tw] OR blind* [tw])) OR (placebos [mh] OR placebo* [tw] OR random* [tw] OR research design [mh:noexp] OR comparative study [mh] OR evaluation studies [mh] OR follow‐up studies [mh] OR prospective studies [mh] OR control* [tw] OR prospectiv* [tw] OR volunteer* [tw]) NOT (animals [mh] NOT human [mh])

#3

HELMINTHS OR ROUNDWORM OR ROUND WORM OR ROUND‐WORM OR ROUNDWORMS OR ROUND WORMS OR ROUND‐WORMS OR NEMATODES OR NEMATODE OR CESTODE OR CESTODES OR TAPEWORM OR TAPE WORM OR TAPE‐WORM OR TAPEWORMS OR TAPE WORMS OR TAPE‐WORMS OR TREMATODE OR TREMATODES OR FLUKE OR FLUKES OR WORM OR WORMS OR PARASITE OR PARASITES OR ASCARIS OR TRICHURIS OR ENTEROBIUS OR STRONGYLOIDE OR STRONGYLOIDES OR ANCYLOSTOMA OR ANCYLOSTOMAS OR NECATOR OR NECATORS OR HYMENOLEPIS OR PARAGONIMUS OR FASCIOLA OR TAENIA OR HOOKWORM OR HOOK WORM OR HOOK‐WORM OR HOOKWORMS OR HOOK WORMS OR HOOK‐WORMS OR WHIPWORM OR WHIP WORM OR WHIP‐WORM OR WHIPWORMS OR WHIP WORMS OR WHIP‐WORMS OR SCHISTOSOMIASIS OR MANSONELLA OR FILARIASIS OR MICROFILARIA OR TRICHOSTRONGYLUS OR TRICHOSTRONGYLOSIS OR STRONGYLOIDEA OR PARAGONIMIASIS

#4

BENZIMIDAZOLES OR ALBENDAZOLE OR MEBENDAZOLE OR IVERMECTIN OR PRAZIQUANTEL OR DIETHYLCARBAMAZINE OR BITHIONOL OR OXAMNIQUINE OR PYRANTEL OR NITAZOXANIDE

#5

#3 AND #4

#6

#1 AND #2 AND #5

#7

#1 AND #2 AND #5 Limits: Publication Date from 1980 to 2015/09/29

Figuras y tablas -
Table 6. Search terms used in 2009 newly applied in MEDLINE
Table 7. Search terms used in 2009 newly applied in EMBASE

Number

Search terms

#1

((’human immunodeficiency virus infection’/exp OR ’human immunodeficiency virus infection’) OR (’human immunodeficiency virus infection’/exp OR ’human immunodeficiency virus infection’)) OR ((’human immunodeficiency virus’/exp OR ’human immunodeficiency virus’) OR (’human immunodeficiency virus’/exp OR ’human immunodeficiency virus’)) OR (hiv:ti OR hiv:ab) OR (’hiv‐1’:ti OR ’hiv‐1’:ab) OR (’hiv‐2’:ti OR ’hiv‐2’:ab) OR (’human immunodeficiency virus’:ti OR ’human immunodeficiency virus’:ab) OR (’human immuno‐deficiency virus’:ti OR ’human immuno‐deficiency virus’:ab) OR (’human immunedeficiency virus’:ti OR ’human immunedeficiency virus’:ab) OR (’human immune‐deficiency virus’:ti OR ’human immune‐deficiency virus’:ab) OR (’acquired immune‐deficiency syndrome’:ti OR ’acquired immune‐deficiency syndrome’:ab) OR (’acquired immunedeficiency syndrome’:ti OR ’acquired immunedeficiency syndrome’:ab) OR (’acquired immunodeficiency syndrome’:ti OR ’acquired immunodeficiency syndrome’:ab) OR (’acquired immuno‐deficiency syndrome’:ti OR ’acquired immuno‐deficiency syndrome’:ab)

#2

random:ti OR random:ab OR factorial:ti OR factorial*:ab OR 'cross over':ti OR 'cross over':ab OR crossover:ti OR crossover:ab OR placebo:ti OR placebo:ab OR (double:ti AND blind:ti) OR (doubl:ab AND blind*:ab) OR (single:ti AND blind:ti) OR (single:ab AND blind:ab) OR assign:ti OR assign:ab OR allocat:ti OR allocate:ab OR volunteer:ti OR volunteer:ab OR 'crossover procedure'/exp OR 'crossover procedure' OR 'double‐blind procedure'/exp OR 'double‐blind procedure' OR 'single‐blind procedure'/exp OR 'single‐blind procedure' OR 'randomized controlled trial'/exp OR 'randomized controlled trial'

#3

'helminths' OR 'roundworm' OR 'round worm' OR 'round‐worm' OR roundworms OR 'round worms' OR 'round‐worms' OR nematodes OR 'nematode' OR 'cestode' OR cestodes OR 'tapeworm' OR 'tape worm' OR 'tape‐worm' OR tapeworms OR 'tape worms' OR 'tape‐worms' OR 'trematode' OR trematodes OR 'fluke' OR flukes OR 'worm' OR worms OR 'parasite' OR 'parasites' OR 'ascaris' OR 'trichuris' OR 'enterobius' OR strongyloide OR stronglyloides OR 'ancylostoma' OR ancylostomas OR 'necator' OR necators OR 'hymenolepis' OR 'paragonimus' OR 'fasciola' OR 'taenia' OR 'hookworm' OR 'hook worm' OR 'hook‐worm' OR hookworms OR 'hook worms' OR 'hook‐worms' OR 'whipworm' OR 'whip worm' OR 'whip‐worm' OR whipworms OR 'whip worms' OR 'whip‐worms' OR shistosomiasis OR 'mansonella' OR 'filariasis' OR 'microfilaria' OR 'trichostrongylus' OR trichostronglylosis OR stronglyloidea OR pargonimiasis

#4

((’benzimidazoles’/exp OR ’benzimidazoles’) OR (’benzimidazoles’/exp OR ’benzimidazoles’)) OR ((’albendazole’/exp OR ’albendazole’) OR (’albendazole’/exp OR ’albendazole’)) OR ((’mebendazole’/exp OR ’mebendazole’) OR (’mebendazole’/exp OR ’mebendazole’)) OR ((’ivermectin’/exp OR ’ivermectin’) OR (’ivermectin’/exp OR ’ivermectin’)) OR ((’praziquantel’/exp OR ’praziquantel’) OR (’praziquantel’/exp OR ’praziquantel’)) OR ((’diethylcarbamazine’/exp OR ’diethylcarbamazine’) OR (’diethylcarbamazine’/exp OR ’diethylcarbamazine’)) OR ((’bithionol’/exp OR ’bithionol’) OR (’bithionol’/exp OR ’bithionol’)) OR ((’oxamniquine’/exp OR ’oxamniquine’) OR (’oxamniquine’/exp OR ’oxamniquine’)) OR ((’pyrantel’/exp OR ’pyrantel’) OR (’pyrantel’/exp OR ’pyrantel’)) OR ((’nitazoxanide’/exp OR ’nitazoxanide’) OR (’nitazoxanide’/exp OR ’nitazoxanide’))

#5

#3 OR #4

#6

#1 AND #2 AND #5 [1‐1‐1980]/sd NOT [29‐09‐2015]/sd

Figuras y tablas -
Table 7. Search terms used in 2009 newly applied in EMBASE
Table 8. Search terms used in 2009 newly applied in CENTRAL

Number

Search terms

#1

(HIV INFECTIONS) OR HIV OR HIV OR HIV‐1* OR HIV‐2* OR HIV1 OR HIV2 OR (HIV INFECT*) OR (HUMAN IMMUNODEFICIENCY VIRUS) OR (HUMAN IMMUNEDEFICIENCY VIRUS) OR (HUMAN IMMUNO‐DEFICIENCY VIRUS) OR (HUMAN IMMUNE‐DEFICIENCY VIRUS) OR ((HUMAN IMMUN*) AND (DEFICIENCY VIRUS)) OR (ACQUIRED IMMUNODEFICIENCY SYNDROME) OR (ACQUIRED IMMUNEDEFICIENCY SYNDROME) OR (ACQUIRED IMMUNO‐DEFICIENCY SYNDROME) OR (ACQUIRED IMMUNE‐DEFICIENCY SYNDROME) OR ((ACQUIRED IMMUN*) AND (DEFICIENCY SYNDROME)) OR (VIRAL SEXUALLY TRANSMITTED DISEASES)

#2

HELMINTHS OR ROUNDWORM OR ROUND WORM OR ROUND‐WORM OR ROUNDWORMS OR ROUND WORMS OR ROUND‐WORMS OR NEMATODES OR NEMATODE OR CESTODE OR CES‐TODES OR TAPEWORM OR TAPE WORM OR TAPE‐WORM OR TAPEWORMS OR TAPE WORMS OR TAPE‐WORMS OR TREMATODE OR TREMATODES OR FLUKE OR FLUKES OR WORM OR WORMS OR PARASITE OR PARASITES OR ASCARIS OR TRICHURIS OR ENTEROBIUS OR STRONGYLOIDE OR STRONGYLOIDES OR ANCYLOSTOMA OR ANCYLOSTOMAS OR NECATOR OR NECATORS OR HYMENOLEPIS OR PARAGONIMUS OR FASCIOLA OR TAENIA OR HOOKWORM OR HOOK WORM OR HOOK‐WORM OR HOOKWORMS OR HOOK WORMS OR HOOK‐WORMS OR WHIPWORM OR WHIP WORM OR WHIP‐WORM OR WHIPWORMS OR WHIP WORMS OR WHIP‐WORMS OR SCHISTOSOMIASIS OR MANSONELLA OR FILARIASIS OR MICROFILARIA OR TRICHOSTRONGYLUS OR TRICHOSTRONGYLOSIS OR STRONGYLOIDEA OR PARAGONIMIASIS

#3

BENZIMIDAZOLES OR ALBENDAZOLE OR MEBENDAZOLE OR IVERMECTIN OR PRAZIQUANTEL OR DIETHYLCARBAMAZINE OR BITHIONOL OR OXAMNIQUINE OR PYRANTEL OR NITAZOXANIDE

#4

#2 OR #3

#5

#1 AND #4 from 1980 to 2015

Figuras y tablas -
Table 8. Search terms used in 2009 newly applied in CENTRAL
Table 9. Search terms used in 2009 newly applied in AIDSEARCH

Number

Search terms

#1

(HIV INFECTIONS) OR HIV OR HIV OR HIV‐1* OR HIV‐2* OR HIV1 OR HIV2 OR (HIV INFECT*) OR (HUMAN IMMUNODEFICIENCY VIRUS) OR (HUMAN IMMUNEDEFICIENCY VIRUS) OR (HUMAN IMMUNO‐DEFICIENCY VIRUS) OR (HUMAN IMMUNE‐DEFICIENCY VIRUS) OR ((HUMAN IMMUN*) AND (DEFICIENCY VIRUS)) OR (ACQUIRED IMMUNODEFICIENCY SYNDROME) OR (ACQUIRED IMMUNEDEFICIENCY SYNDROME) OR (ACQUIRED IMMUNO‐DEFICIENCY SYNDROME) OR (ACQUIRED IMMUNE‐DEFICIENCY SYNDROME) OR ((ACQUIRED IMMUN*) AND (DEFICIENCY SYNDROME)) OR (SEXUALLY TRANSMITTED DISEASES, VIRAL)

#2

((RANDOMIZED CONTROLLED TRIAL) OR (CONTROLLED CLINICAL TRIAL) OR (RANDOMIZED CONTROLLED TRIALS) OR (RANDOM ALLOCATION) OR (DOUBLE‐BLIND METHOD) OR (SINGLE‐ BLIND METHOD) OR (CLINICAL TRIAL) OR (CLINICAL TRIALS) OR (“CLINICAL TRIAL”) OR ((SINGL* OR DOUBL* OR TREBL* OR TRIPL* AND (MASK* OR BLIND*)) OR PLACEBOS OR PLACEBO* OR RANDOM* OR (COMPARATIVE STUDY) OR (EVALUATION STUDIES) OR (FOLLOW‐UP STUDIES) OR (PROSPECTIVE STUDIES) OR CONTROL* OR PROSPECTIV* OR VOLUNTEER*)) NOT (ANIMALS NOT HUMAN)

#3

#1 AND #2

#4

HELMINTHS OR ROUNDWORM OR ROUND WORM OR ROUND‐WORM OR ROUNDWORMS OR ROUND WORMS OR ROUND‐WORMS OR NEMATODES OR NEMATODE OR CESTODE OR CES‐TODES OR TAPEWORM OR TAPE WORM OR TAPE‐WORM OR TAPEWORMS OR TAPE WORMS OR TAPE‐WORMS OR TREMATODE OR TREMATODES OR FLUKE OR FLUKES OR WORM OR WORMS OR PARASITE OR PARASITES OR ASCARIS OR TRICHURIS OR ENTEROBIUS OR STRONGYLOIDE OR STRONGYLOIDES OR ANCYLOSTOMA OR ANCYLOSTOMAS OR NECATOR OR NECATORS

#5

HYMENOLEPIS OR PARAGONIMUS OR FASCIOLA OR TAENIA OR HOOKWORM OR HOOK WORM OR HOOK‐WORM OR HOOKWORMS OR HOOK WORMS OR HOOK‐WORMS OR WHIPWORM OR WHIP WORM OR WHIP‐WORM OR WHIPWORMS OR WHIP WORMS OR WHIP‐WORMS OR SCHISTOSOMIASIS OR MANSONELLA OR FILARIASIS OR MICROFILARIA OR TRICHOSTRONGYLUS OR TRICHOSTRONGYLOSIS OR STRONGYLOIDEA OR PARAGONIMIASIS

#6

BENZIMIDAZOLES OR ALBENDAZOLE OR MEBENDAZOLE OR IVERMECTIN OR PRAZIQUANTEL OR DIETHYLCARBAMAZINE OR BITHIONOL OR OXAMNIQUINE OR PYRANTEL OR NITAZOXANIDE

#7

#4 OR #5 OR #6

#8

#7 AND #3

Figuras y tablas -
Table 9. Search terms used in 2009 newly applied in AIDSEARCH
Table 10. Search strategy for the WHO Global Health Library

Number

Search terms

#1

Helminth* AND "HIV Infections" received from 01/01/1980 to 09/29/2015

Figuras y tablas -
Table 10. Search strategy for the WHO Global Health Library
Table 11. Characteristics of trials in which participants had unknown helminth infection status

Trial

Country

Setting

Year enrolment completed

Inclusion criteria

Age

Total randomized

Number HIV‐positive participants

Taking ART

Number co‐infected with helminths at baseline

Kelly 1996 ZMB

Zambia

Urban

Not stated

HIV‐infected adults with persistent diarrhoea

> 18 years

174

174

Not stated (probably low)

Unknown

Webb 2012 UGA

Uganda

Urban

2005

HIV‐infected pregnant women

Not stated

264

264

< 3%

67% had at least 1 helminth species

Walson 2012 KEN

Kenya

Urban/rural

2011

HIV‐infected, not on ART

> 18 years

948

948

None at baseline

Unknown

Abbreviations: HIV: human immunodeficiency virus; ART: antiretroviral.

Figuras y tablas -
Table 11. Characteristics of trials in which participants had unknown helminth infection status
Table 12. Description of interventions

Trial

Intervention

Control

Outcomes

Timing

Drug

Dose

Frequency

Walson 2008 KEN

Albendazole

400 mg

Once daily for 3 days

Placebo

Viral load

CD4

Adverse events

Mortality events

12 weeks

Abate 2014 ETH

Albendazole

400 mg

Once daily for 3 days

Placebo

CD4

Adverse events

Mortality events

12 weeks

Kallestrup 2005 ZWE

Praziquantel

40 mg/kg

Once only

No intervention

Viral load

CD4

12 weeks

Nielsen 2007 TZA

Diethylcarbamazine

6 mg/kg

Once only

Placebo

Viral load

CD4

Adverse events

12 weeks

Kelly 1996 ZMB

Albendazole

800 mg

Twice daily for 14 days

Placebo

Adverse events

Mortality events

6 months

Suputtamongkol 2011 THA

Ivermectin

200 mg/kg

Single or double dose 2 weeks apart

Albendazole

Adverse events

1 year

Webb 2012 UGA

Albendazole

Praziquantel

400 mg

40 mg/kg

Once only

Once only

Placebo

Viral load

Adverse events

Mortality events

6 weeks

Walson 2012 KEN

Albendazole

Praziquantel

400 mg

25 mg/kg

Every 3 months

Annually

No intervention

Viral load

CD4

Adverse events

Mortality events

2 years

Figuras y tablas -
Table 12. Description of interventions
Table 13. Characteristics of trials in which participants had confirmed helminth infections

Trial

Country

Setting

Year enrolment completed

Inclusion criteria

Age

Total randomized

Number of HIV‐positive participants

Taking ART

Number co‐infected with helminths at baseline

Walson 2008 KEN

Kenya

Urban/rural

2007

HIV‐infected with at least one helminth co‐infection

> 18 years

234

234

None at baseline

All

Abate 2014 ETH

Ethiopia

Urban

2012

Newly diagnosed TB participants with helminth co‐infection

15 to 60 years

140

32

94% of intervention group, 100% of controls

All

Kallestrup 2005 ZWE

Zimbabwe

Rural

2003

Adults infected with schistosomiasis

> 18 years

287

130

Not stated

All

Nielsen 2007 TZA

Tanzania

Rural

2002

HIV‐infected adults

22 to 70 years

34

34

Not stated

18

Suputtamongkol 2011 THA

Thailand

Urban

2009

Adults with characteristic strongyloides infection

> 18 years

100

10

Not stated

All

Abbreviations: HIV: human immunodeficiency virus; ART: antiretroviral; TB: tuberculosis.

Figuras y tablas -
Table 13. Characteristics of trials in which participants had confirmed helminth infections
Table 14. Ongoing or planned RCTs

Trial name

Relevant intervention

Relevant outcome

Target population

Location(s)

Estimated completion date

Reduction of EArly mortaLITY in HIV‐infected Adults and Children Starting Antiretroviral Therapy (REALITY)

Immediate enhanced opportunistic infections (OI) prophylaxis with isoniazid/pyridoxine and cotrimoxazole, plus 12 weeks fluconazole, 5 days azithromycin, and a single dose of albendazole versus cotrimoxazole prophylaxis alone for the first 12 weeks followed by isoniazid and any prophylaxis and/or treatment prescribed at screening

• Change in CD4 count

• Adverse events

HIV‐infected individuals ages ≥ 5 years

Kenya, Malawi, Uganda, Zimbabwe

February 2016

Can Anthelminthic Treatment Delay the Progression of HIV? Randomised Open‐label Trial Testing Presumptive Anthelminthic Treatment on Progression of HIV in ART‐naïve HIV‐positive Patients in a Rural African Setting With Presumed High Prevalence of Helminth Infections

Standard HIV care with provision of praziquantel, albendazole, and ivermectin at baseline, after 6 months, and after 12 months versus standard HIV care with no anthelminthic treatment

• Change in viral load

• Change in CD4 count

• Adverse event

HIV‐infected individuals aged ≥ 18 years

Tanzania

Terminated prematurely due to recruitment difficulties

Abbreviations: RCT: randomized controlled trial; HIV: human immunodeficiency virus; ART: antiretroviral.

Figuras y tablas -
Table 14. Ongoing or planned RCTs
Comparison 1. Deworming drugs versus placebo in people with unknown helminth infection status

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Viral load: change in log10 HIV‐1 RNA Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

1.1 At 6 weeks after one dose

1

166

Mean Difference (IV, Random, 95% CI)

‐0.14 [‐0.35, 0.07]

1.2 At 2 years after multiple doses

1

917

Mean Difference (IV, Random, 95% CI)

0.01 [‐0.03, 0.05]

2 Change in CD4 count Show forest plot

1

917

Mean Difference (IV, Random, 95% CI)

2.60 [‐10.15, 15.35]

2.1 At 2 years after multiple doses

1

917

Mean Difference (IV, Random, 95% CI)

2.60 [‐10.15, 15.35]

Figuras y tablas -
Comparison 1. Deworming drugs versus placebo in people with unknown helminth infection status
Comparison 2. Deworming drugs versus placebo in people with confirmed helminth infection

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Viral load: change in log10 HIV‐1 RNA Show forest plot

4

445

Mean Difference (IV, Random, 95% CI)

‐0.13 [‐0.26, ‐0.00]

1.1 Soil‐transmitted helminths

2

273

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.31, 0.12]

1.2 Schistosomiasis

2

161

Mean Difference (IV, Random, 95% CI)

‐0.13 [‐0.36, 0.10]

1.3 Lymphatic filariasis

1

11

Mean Difference (IV, Random, 95% CI)

‐0.08 [‐0.93, 0.77]

2 Change in CD4 count Show forest plot

3

358

Mean Difference (IV, Random, 95% CI)

37.86 [7.36, 68.35]

2.1 Soil‐transmitted helminths

2

228

Mean Difference (IV, Random, 95% CI)

40.69 [1.51, 79.87]

2.2 Schistosomiasis

1

130

Mean Difference (IV, Random, 95% CI)

33.5 [‐15.06, 82.06]

Figuras y tablas -
Comparison 2. Deworming drugs versus placebo in people with confirmed helminth infection
Comparison 3. Deworming drugs versus placebo; all trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Change in log10 HIV RNA, by antihelminthic Show forest plot

5

1534

Mean Difference (IV, Random, 95% CI)

‐0.09 [‐0.19, 0.01]

1.1 Albendazole

2

302

Mean Difference (IV, Random, 95% CI)

‐0.18 [‐0.37, 0.02]

1.2 Praziquantel

2

213

Mean Difference (IV, Random, 95% CI)

‐0.17 [‐0.32, ‐0.03]

1.3 Diethylcarbamazine

1

11

Mean Difference (IV, Random, 95% CI)

‐0.08 [‐0.93, 0.77]

1.4 Albendazole and praziquantel

2

1008

Mean Difference (IV, Random, 95% CI)

0.01 [‐0.04, 0.05]

2 Difference in adverse events between treatment and no treatment groups Show forest plot

7

1649

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.53, 2.83]

3 Difference in mortality events between treatment and no treatment groups Show forest plot

5

1627

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.52, 1.14]

Figuras y tablas -
Comparison 3. Deworming drugs versus placebo; all trials