Scolaris Content Display Scolaris Content Display

Intervenciones para la micosis fungoide

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

Child 2004 {published data only (unpublished sought but not used)}

Child FJ, Mitchell TJ, Whittaker SJ, Scarisbrick JJ, Seed PT, Russel‐Jones R. A randomized cross‐over study to compare PUVA and extracorporeal photopheresis in the treatment of plaque stage (T2) mycosis fungoides. Clinical & Experimental Dermatology 2004;29(3):231‐6. [PUBMED: 15115499]
Child FJ, Mitchell TJ, Whittaker SJ, Watkins P, Seed P, Russell‐Jones R. A randomised cross‐over study to compare Puva and extracorporeal photopheresis (ECP) in the treatment of plaque stage (T2) mycosis fungoides. British Journal of Dermatology 2001;145(Suppl 59):16.

Chong 2004 {published data only (unpublished sought but not used)}

Chong A, Loo WJ, Banney L, Grant JW, Norris PG. Imiquimod 5% cream in the treatment of mycosis fungoides ‐ a pilot study. Journal of Dermatological Treatment 2004;15(2):118‐9. [PUBMED: 15204164]

Duvic 2001 {published data only}

Duvic M, Martin AG, Kim Y, Olsen E, Wood GS, Crowley CA, et al. Phase 2 and 3 clinical trial of oral bexarotene (Targretin capsules) for the treatment of refractory or persistent early‐stage cutaneous T‐cell lymphoma. Archives of Dermatology 2001;137(5):581‐93. [PUBMED: 11346336]
Prince HM, McCormack C, Ryan G, Baker C, Rotstein H, Davison J, et al. Bexarotene capsules and gel for previously treated patients with cutaneous T‐cell lymphoma: results of the Australian patients treated on phase II trials. Australasian Journal of Dermatology 2001;42(2):91‐7. [PUBMED: 11309029]

Duvic 2001a {published data only}

Duvic M, Olsen EA, Omura GA, Maize JC, Vonderheid EC, et al. A phase III, randomized, double‐blind, placebo‐controlled study of peldesine (BCX‐34) cream as topical therapy for cutaneous T‐cell lymphoma. Journal of the American Academy of Dermatology 2001;44(6):940‐7. [PUBMED: 11369904]

Guitart 2002 {published and unpublished data}

Guitart J, Tucker R, Stevens V. Low Dose Bexarotene (Targretin®) Capsules and Phototherapy for Early Stage Cutaneous T‐Cell Lymphoma. [Abstract 199]. The 63rd Annual Meeting of the Society for Investigative Dermatology, 15‐18 May, Los Angeles, USA. Journal of Investigative Dermatology 2002;119(1):241.
NCT00030589. A Muliticenter, Dose‐Reandomized Evaluation Of Targretin Capsules Plus PUVA In Patients With Stage IB ‐ IIA Cutaneous T‐Cell Lymphoma. clinicaltrials.gov/ct/show/NCT00030589 (accessed 26 October 2011).

Kaye 1989 {published data only}

Kaye F, Eddy J, Ihde DC, Steinberg S, Fischmann AB, Glatstein E, et al. Conservative vs. aggressive therapy in mycosis fungoides [Abstract 1002]. Proceedings of the American Society of Clinical Oncology 1989;8:257.
Kaye F, Ihde D, Fischmann A, Glatstein E, Minna J, Schechter G, et al. A randomized trial comparing conservative and aggressive therapy in mycosis fungoides (MF) [Abstract 765]. Proceedings of the American Society of Clinical Oncology 1986;5:195.
Kaye FJ, Bunn PA, Steinberg SM, Stocker JL, Ihde DC, Fischmann AB, et al. A randomized trial comparing combination electron‐beam radiation and chemotherapy with topical therapy in the initial treatment of mycosis fungoides. New England Journal of Medicine 1989;321(26):1784‐90. [PUBMED: 2594037]

Olsen 2001 {published and unpublished data}

Duvic M, Kuzel TM, Olsen EA, Martin AG, Foss FM, Kim YH, et al. Quality‐of‐life improvements in cutaneous T‐cell lymphoma patients treated with denileukin diftitox (ONTAK). Clinical Lymphoma 2002;2(4):222‐8. [PUBMED: 11970761]
Kuzel T, Olsen E, Martin A, Kim Y, Duvic M, Frankel A, et al. Pivotal phase III trial of two dose levels of DAB389IL‐2 (Ontak®) for the treatment of mycosis fungoides (MF). Blood 1997;90(10 Suppl 1 (Pt 1)):586a.
NTC00050999. Study of ONTAK (Denileukin Diftitox) in Cutaneous T‐Cell Lymphoma (CTCL) Patients. clinicaltrials.gov/ct2/show/NCT00050999 (accessed 26 October 2011).
Olsen E, Duvic M, Frankel A, Kim Y, Martin A, Vonderheid E, et al. Pivotal phase III trial of two dose levels of denileukin diftitox for the treatment of cutaneous T‐cell lymphoma. Journal of Clinical Oncology 2001;19(2):376‐88. [PUBMED: 11208829]
Olsen E, Duvic M, Martin A. Pivotal phase III trial of two dose levels of DAB 389 IL‐2 (ONTAK) for the treatment of cutaneous T‐cell lymphoma (CTCL) [Abstract 1234]. Journal of Investigative Dermatology 1998;110(4):678.

Rook 2010 {published and unpublished data}

Rook AH, Wood GS, Duvic M, Vonderheid EC, Tobia A, Cabana B. A phase II placebo‐controlled study of photodynamic therapy with topical hypericin and visible light irradiation in the treatment of cutaneous T‐cell lymphoma and psoriasis. Journal of the American Academy of Dermatology 2010;63(6):984‐90. [PUBMED: 20889234]

Stadler 1998 {published and unpublished data}

Otte HG, Kuhl S, Stadler R, Luger T, Henz B, Sterry W. Treatment of cutaneous T‐cell lymphoma with Interferon alpha and PUVA versus Interferon alpha and Acitretin ‐ results of a prospective randomised multi‐centre study [Behandlung des kutanen T‐Zell‐Lymphoms mit Interferon alpha und PUVA versus Interferon alpha und Acirentin ‐ Ergebnisse einer prospektiv‐randomisierten Multicenter‐Studie]. Der Hautarzt 1997;48(Suppl):S97.
Otte HG, Stadier R, Luger T, Henz B, Sterry W. Open controlled clinical trial on the use of interferon plus acitretin versus interferon plus PUVA in patients with cutaneous T‐cell lymphoma (CTCL) [Poster P232]. Annals of Oncology 1996;7(Suppl 3):66.
Stadler R, Otte HG, Luger T, Henz BM, Kuhl P, Zwingers T, et al. Prospective randomized multicenter clinical trial on the use of interferon ‐2a plus acitretin versus interferon ‐2a plus PUVA in patients with cutaneous T‐cell lymphoma stages I and II. Blood 1998;92(10):3578‐81. [PUBMED: 9808550]

Stadler 2006 {published and unpublished data}

Bohmeyer J, Otte HG, Stadler R, Luger T, Sterry W. Therapy of cutaneous T‐cell lymphoma with PUVA versus interferon alfa plus PUVA: first results [Abstract P‐447]. Journal of the European Academy of Dermatology & Venereology 1999;12(Suppl 2):S268.
Bohmeyer J, Stadler R, Luger T, Sterry W. Prospective randomized multicentre therapy optimizing protocol for therapy of cutaneous t‐cell‐lymphoma with PUVA versus Interferon α + PUVA [Prospectiv randomisiertes, multizentrisches Therapieoptimierungsprotokoll zur Therapie kutaner T‐Zell‐Lymphome mit PUVA versus Interferon α + PUVA]. Zeitschrift für Hautkrankheiten 2001;76(Suppl 1):S55.
Kremer A, Bohmeyer J, Stadler R, Luger T, Sterry W. Prospective randomized multicentre therapy optimizing protocol for therapy of cutaneous t‐cell‐lymphoma with PUVA versus Interferon α + PUVA [Prospectiv randomisiertes, multizentrisches Therapieoptimierungsprotokoll zur Therapie kutaner T‐Zell‐Lymphome mit PUVA versus Interferon α + PUVA]. Journal der Deutschen Dermatologischen Gesellschaft 2003;1(Suppl 1):S99.
Otte HG, Stadler R. combination therapy of cutaneous t‐cell‐lymphoma with interferon alfa 2a and PUVA [Kombinationstherapie kutaner T‐Zell‐Lymphome mit Interferon Alfa 2a und PUVA]. Zentralblatt Haut‐ und Geschlechtskrankheiten 1993;162(Suppl):P2.03.08.
Otte HG, Stadler R, Luger T, Sterry W. Therapy of cutaneous t‐cell‐lymphoma with PUVA versus Interferon α plus PUVA: first results of a prospective randomized multicentre therapy optimizing protocol [Therapie kutaner T‐Zell‐Lymphome mit PUVA versus Interferon α plus PUVA: Erste Ergebnisse des prospektiven randomisierten multizentrischen Therapieoptimierungsprotokolls]. Der Hautarzt 1999;50(Suppl 1):S2.
Stadler M, Kremer A, Luger T, Sterry W. Prospective, randomized, multicentre clinical trial on the use of interferon a 2a plus PUVA versus PUVA monotherapy in patients with cutaneous T‐cell lymphoma, stages I and II [Abstract 7541]. ASCO annual meeting proceedings. Journal of Clinical Oncology 2006;24(18S Pt 1):432.

Thestrup‐Pedersen 1982 {published and unpublished data}

Thestrup‐Pedersen K, Grunnet E, Zachariae H. Transfer factor therapy in mycosis fungoides: a double‐blind study. Acta Dermato‐Venereologica 1982;62(1):47‐53. [PUBMED: 6175137]

Vonderheid 1987 {published data only (unpublished sought but not used)}

Vonderheid EC, Thompson R, Smiles KA, Lattanand A. Recombinant interferon alfa‐2b in plaque‐phase mycosis fungoides. Intralesional and low‐dose intramuscular therapy. Archives of Dermatology 1987;123(6):757‐63. [3579357]

Wolff 1985 {published data only (unpublished sought but not used)}

Wolff JM, Zitelli JA, Rabin BS, Smiles KA, Abell E. Intralesional interferon in the treatment of early mycosis fungoides. Journal of the American Academy of Dermatology 1985;13(4):604‐12. [PUBMED: 2934438]

Wozniak 2008 {published data only (unpublished sought but not used)}

Wozniak MB, Tracey L, Ortiz‐Romero PL, Montes S, Alvarez M, Fraga J, et al. Psoralen plus ultraviolet A +/‐ interferon‐alpha treatment resistance in mycosis fungoides: the role of tumour microenvironment, nuclear transcription factor‐kappaB and T‐cell receptor pathways. British Journal of Dermatology 2008;160(1):92‐102. [PUBMED: 18945306]

References to studies excluded from this review

Argyropoulos 1979 {published data only}

Argyropoulos CL, Lamberg SI, Clendenning WE, Fischmann AB, Jegasothy B, Zackheim H, et al. Preliminary evaluation of 15 chemotherapeutic agents applied topically in the treatment of mycosis fungoides. Cancer Treatment Reports 1979;63(4):619‐21.

Breneman 1991 {published data only}

Breneman DL, Nartker AL, Ballman EA, Pruemer JM, Blumsack RF, Davis M, et al. Topical mechlorethamine in the treatment of mycosis fungoides. Uniformity of application and potential for environmental contamination. Journal of the American Academy of Dermatology 1991;25(6 Pt 1):1059‐64.

Cooper 1994 {published data only}

Cooper IA, Wolf MM, Robertson TI, Fox RM, Matthews JP, Stone JM, et al. Randomized comparison of MACOP‐B with CHOP in patients with intermediate‐grade non‐Hodgkin's lymphoma. The Australian and New Zealand Lymphoma Group. Journal of Clinical Oncology 1994;12(4):769‐78.

Currie 1980 {published data only}

Currie VE, Kempin SJ, Young CW. Phase I trial of metoprine in patients with advanced cancer. Cancer Treatment Reports 1980;64(8‐9):951‐6.

Dang 2007 {published data only}

Dang NH, Pro B, Hagemeister FB, Samaniego F, Jones D, Samuels BI, et al. Phase II Study of Denileukin Diftitox for Relapsed/Refractory B‐Cell Non‐Hodgkin's Lymphoma. British Journal of Haematology 2007;136(3):439‐47. [PUBMED: 17233846]

Doan 1958 {published data only}

Doan CA, Wiseman BK, Bouroncle BA. Clinical evaluation of CB 1348 in leukemias and lymphomas. Annals of the New York Academy of Sciences 1958;68(3):979‐95.

Dueck 2010 {published data only}

Dueck G, Chua N, Prasad A, Finch D, Stewart D, White D, et al. Interim report of a phase 2 clinical trial of lenalidomide for T‐cell non‐Hodgkin lymphoma. Cancer 2010;116(19):4541‐8.

Fawzi 2010 {published data only}

Fawzi M, El Mofty M, Ramadan S, Hegazy R, Sayed S. Broadband UVA versus PUVA in the treatment of early stage mycosis fungoides: A comparative study. [Abstract P99]. 6th Congress of the European Association of Dermatologic Oncology Athens Greece. 16‐19 June 2010. Melanoma Research 2010;20:e85.

Fisher 1993 {published data only}

Fisher RI, Gaynor ER, Dahlberg S, Oken MM, Grogan TM, et al. Comparison of a standard regimen (CHOP) with three intensive chemotherapy regimens for advanced non‐Hodgkin's lymphoma. New England Journal of Medicine 1993;328(14):1002‐6.

JapicCTI‐050041 {published data only}

JapicCTI‐050041. Post‐marketing clinical study of Ogamma 100 in patients with mycosis fungoides. www.clinicaltrials.jp/user/showCteDetailE.jsp?japicId=JapicCTI‐050041 (accessed 27 October 2011).

Kaung 1969 {published data only}

Kaung DT, Wittington RM, Spencer H, Patno ME. Comparison of chlorambucil and streptonigrin (NSC‐45383) in the treatment of malignant lymphomas. Cancer 1969;23(6):1280‐3.

Kujawska 2003 {published data only}

Kujawska A, Abrou A, Fivenson DP, Lim HW. PUVA vs. PUVA and interferon alpha for treatment of mycosis fungoides. [Abstract 1185]. International Investigative Dermatology. The 4th Joint Meeting of the ESDR, Japanese SID & SID, 30th April‐4thMay 2003, Florida, USA. Journal of Investigative Dermatology 2003;121(1).

Kuzel 2010 {published data only}

Kuzel T. Correlates of capillary leak syndrome (CLS) in patients with cutaneous T‐cell lymphoma (CTCL) during treatment with denileukin diftitox (DD) in a placebo (PBO)‐controlled phase III trial. [Abstract e18509 ]. 4‐8 June 2010 Annual Meeting of the American Society of Clinical Oncology, ASCO Chicago, IL United States. Journal of Clinical Oncology 2010;28(15 Suppl May).

Lansigan 2010 {published data only}

Lansigan F, Foss FM. Rate of infection and immunosuppression in two phase III studies of denileukin diftitox (DD) in cutaneous T‐cell lymphoma (CTCL). [Abstract e18515]. 4‐8 June 2010 Annual Meeting of the American Society of Clinical Oncology, ASCO Chicago, IL United States. Journal of Clinical Oncology 2010;28(15 Suppl May).

NCT00054171 {published data only}

NCT00054171. Phase I Randomized Pilot Study of Photodynamic Therapy Using Aminolevulinic Acid in Patients With Cutaneous T‐Cell or B‐Cell Lymphoma or Early Chronic Lymphocytic Leukemia With Cutaneous Infiltrates. clinicaltrials.gov/ct/show/NCT00054171 (accessed 27 October 2011).

Neering 1972 {published data only}

Neering H, Kroon HV, van der Roeleveld CG. Treatment of localized skin lesions with betamethasone 17‐valerate and triamcinolone acetonide in alcoholic solution under occlusive dressing. A double blind comparative study. Dermatologica 1972;145(6):395‐9.

Negro‐Vilar 2007 {published data only}

Negro‐Vilar A, Dziewanowska Z, Groves ES, Stevens V, Zhang JK, Prince M, et al. Efficacy and safety of denileukin diftitox (Dd) in a phase III, double‐blind, placebo‐controlled study of CD25+ patients with cutaneous T‐cell lymphoma (CTCL) [Abstract 8026]. ASCO annual meeting proceedings. Journal of Clinical Oncology 2007;25(18S Suppl June):447.

No authors listed 1982 {published data only}

No authors listed. Comparison of the use of teniposide and vincristine in combination chemotherapy for non‐Hodgkin's lymphoma. Cancer Treatment Reports 1982;66(1):49‐55.

Olsen 1986 {published and unpublished data}

Olsen EA, Diab NG. Interferon alfa‐2A in the treatment of cutaneous T cell lymphoma [Abstract]. 47th Annual Meeting of The Society for Investigative Dermatology, Inc, Washington, D.C. 1‐4 May 1986. Journal of Investigative Dermatology 86;4:498. [DOI: 10.1111/1523‐1747.ep12285810]

Pan 2007 {published data only}

Pan Y, Prince HM, Ellis L, Culver K. LBH589, a Novel Deacetylase Inhibitor (DACi), in the Treatment of Cutaneous T‐Cell Lymphoma (CTCL): Changes in tumor Gene Expression Profiles Related to Clinical Response after Therapy. [Abstract 00172]. The 11th World Congress on Cancers of the Skin. Amsterdam, The Netherlands. 8th‐11th June 2007. 2007.

Peugeot 1995 {published data only}

Peugeot RL. Clinical efficacy of a novel purine nucleoside phosphorylase inhibitor (BCX‐34) in the treatment of cutaneous T‐cell lymphoma. [Abstract 54]. 1995 Annual Meeting of the Society for Investigative Dermatology. Journal of Investigative Dermatology 1995;104(4):563.

Plettenberg 2001 {published data only}

Plettenberg H, Stege H, Megahed T, Ruzicka T, Hosokawa Y, Tsuji T, et al. UVA1‐Phototherapie versus PUVA‐Photochemotherapie in der Behandlung von Patienten mit kutanem T‐Zell‐Lymphom (CTCL). Zeitschrift für Hautkrankheiten 2001;76(Suppl 1):S96.

Prince 2010 {published data only}

Prince HM, Duvic M, Martin A, Sterry W, Assaf C, Sun Y, et al. Phase III placebo‐controlled trial of denileukin diftitox for patients with cutaneous T‐cell lymphoma. Journal of Clinical Oncology 2010;28(11):1870‐7. [PUBMED: 20212249]

Schrag 1997 {published data only}

Schrag H‐J, Dippel E, Goerdt S, Orfanos C E. Extracorporal photophoresis (ECP) + Interferon‐alpha 2a vs. ECP in patients with cutaneous T‐cell lymphoma. Evaluation using a new score (CTCL‐SI) (German). Der Hautarzt 1997;48(Suppl):S96.

Simon 2010 {published data only}

Simon A, Peoch M, Casassus P, Deconinck E, Colombat P, Desablens B, et al. Upfront VIP‐reinforced‐ABVD (VIP‐rABVD) is not superior to CHOP/21 in newly diagnosed peripheral T cell lymphoma. Results of the randomized phase III trial GOELAMS‐LTP95. British Journal of Haematology 2010;151(2):159‐66. [PUBMED: 20738307]

Thomsen 1977 {published data only (unpublished sought but not used)}

Thomsen K. Scandinavian mycosis fungoides study group. Bulletin du Cancer 1977;64(2):287‐90. [PUBMED: 912132]

Wain 2005 {published data only}

Wain EM, Whittaker SJ, Russell‐Jones R. A randomized, open, crossover study to compare the efficacy of extracorporeal photopheresis with methotrexate in the treatment of erythrodermic primary cutaneous T‐cell lymphoma. [Abstract RF‐5]. The 85th BAD Annual Meeting 5‐8th July 2005, Glasgow, UK. British Journal of Dermatology 2005;153(Suppl 1):10.

Wiernik 1998 {published data only}

Wiernik PH, Moore DF, Bennett JM, Vogl SE, Harris JE, Luger S, et al. Phase II study of mitoguazone, cyclophosphamide, doxorubicin, vincristine and prednisone for patients with diffuse histologic subtypes of non‐Hodgkin's lymphoma: an Eastern Cooperative Oncology Group Study (PE481). Leukemia & Lymphoma 1998;30(5‐6):601‐7.

Zubrod 1960 {published data only}

Zubrod CG, Schneiderman M, Frei E, Brindley C, Gold GL, Shnider B, et al. Appraisal of methods for the study of chemotherapy of cancer in man: Comparative therapeutic trial of nitrogen mustard and triethylene thiophosphoramide. Journal of Chronic Diseases 1960;11(1):7‐33.

References to studies awaiting assessment

Foss 2011 {published data only}

Foss F, Duvic M, Olsen EA. Predictors of complete responses with denileukin diftitox in cutaneous T‐cell lymphoma. American Journal of Hematology 2011;86(7):627‐30.

Lessin 2011 {published data only}

Lessin S, Duvic M, Guitart J, Pandya A, Strober B, Olsen E, et al. Positive results of a pivotal phase ii trial testing a manufactured 0.02% Mechlorethamine gel in mycosis fungoides (mf) [Abstract 503]. Annual Meeting of the Society for Investigative Dermatology Phoenix, AZ United States, 4‐7 May 2011. Journal of Investigative Dermatology 2011;131(Suppl 1):S84.

NCT00056056 {published data only}

NCT00056056. Phase III Randomized Study of Ultraviolet A Light Therapy With Methoxsalen (PUVA) With or Without Bexarotene in Patients With Mycosis Fungoides. clinicaltrials.gov/ct/show/NCT00056056 (accessed 24 September 2011).

NCT00091208 {published data only}

NCT00091208. A Phase I/II Open Label, Multi‐Center Study For The Evaluation Of CPG 7909 In Patients With Stage IB To IVA Cutaneous T‐Cell Lymphoma. clinicaltrials.gov/ct/show/NCT00091208 (accessed 15 September 2011).

NCT00168064 {published data only}

NCT00168064. A Phase II Pivotal Trial to Evaluate the Safety and Efficacy of Nitrogen Mustard (NM) 0.02% Ointment Formulations in Patients With Stage I or IIA Mycosis Fungoides (MF). clinicaltrials.gov/ct/show/NCT000168064 (accessed 3 October 2011).

NCT01007448 {published data only}

NCT01007448. Phase IV Randomized Study Of Two Dose Levels Of Targretin Capsules In Patients With Refractory Cutaneous T‐Cell Lymphoma. clinicaltrials.gov/ct/show/NCT01007448 (accessed 15 September 2011).

NCT01098656 {published data only}

NCT01098656. A Phase III Study of Lenalidomide Maintenance After Debulking With Gemcitabine or Liposomal Doxorubicin +/‐ Radiotherapy in Patients With Advanced Cutaneous T‐Cell Lymphoma Not Previously Treated With Intravenous Chemotherapy. clinicaltrials.gov/ct/show/NCT01098656 (accessed 15 September 2011).

NCT01187446 {published data only}

NCT01187446. A Multicenter, Open‐label, Randomized, Phase I/II Study Evaluating the Safety and Efficacy of Low‐dose (12 Gy) Total Skin Electron Beam Therapy (TSEBT) Combined With Vorinostat Versus Low‐dose TSEBT Monotherapy in Mycosis Fungoides (MF). clinicaltrials.gov/ct/show/NCT01187446 (accessed 15 September 2011).

NCT01386398 {published data only}

NCT01386398. Vorinostat (Zolinza®) in Combination With (Velcade®) Versus Vorinostat Alone in Refractory or Recurrent Advanced CTCL: A Randomized Phase III Study. clinicaltrials.gov/ct/show/NCT01386398 (accessed 15 September 2011).

NCT01433731 {published data only}

NCT01433731. A Randomized, Double‐Blind, Placebo‐Controlled, Dose‐Escalating Phase 1b Study to Assessthe Safety, Pharmacodynamics and Pharmacokinetics of SHP 141, A Histone DeacetylaseInhibitor, Administered Topically Up to 28 Days to Patients With Stage IA, IB or IIA CutaneousT‐Cell Lymphoma. clinicaltrials.gov/ct/show/NCT01433731 (accessed 29 April 2012).

Agar 2010

Agar NS, Wedgeworth E, Crichton S, Mitchell TJ, Cox M, Ferreira S, et al. Survival outcomes and prognostic factors in mycosis fungoides/Sezary syndrome: validation of the revised International Society for Cutaneous Lymphomas/European Organisation for Research and Treatment of Cancer staging proposal. Journal of Clinical Oncology 2010;28(31):4730‐9. [PUBMED: 20855822]

Bernstein 1989

Bernstein L, Deapen D, Ross RK. Mycosis fungoides. JAMA 1989;261(13):1882. [PUBMED: 2784510]

Bradford 2009

Bradford PT, Devesa SS, Anderson WF, Toro JR. Cutaneous lymphoma incidence patterns in the United States: a population‐based study of 3884 cases. Blood 2009;113(21):5064‐73. [PUBMED: 19279331]

Bunn 1979

Bunn PA, Lamberg SI. Report of the committee on staging and classification of cutaneous T‐cell lymphomas. Cancer Treatment Reports 1979;63(4):725‐8. [PUBMED: 445521]

Cella 1993

Cella DF, Tulsky DS, Gray G, Sarafian B, Linn E, Bonomi A, et al. The Functional Assessment of Cancer Therapy scale: development and validation of the general measure. Journal of Clinical Oncology 1993;11(3):570‐9. [PUBMED: 8445433]

Chan 2004

Chan AW, Hróbjartsson A, Haahr MT, Gøtzsche PC, Altman DG. Empirical evidence for selective reporting of outcomes in randomized trials: comparison of protocols to published articles. JAMA 2004;291(20):2457‐65.

Criscione 2007

Criscione VD, Weinstock MA. Incidence of cutaneous T‐cell lymphoma in the United States, 1973‐2002. Archives of Dermatology 2007;143(7):854‐9. [PUBMED: 17638728]

Dekkers 2010

Dekkers OM, von Elm E, Algra A, Romijn JA, Vandenbroucke JP. How to assess the external validity of therapeutic trials: a conceptual approach. International Journal of Epidemiology 2010;39(1):89‐94. [PUBMED: 19376882]

Dickersin 1994

Dickersin K, Scherer R, Lefebvre C. Identifying relevant studies for systematic reviews. BMJ 1994;309(6964):1286‐91. [PUBMED: 7718048]

Dummer 2008

Dummer R, Dreyling M. Primary cutaneous lymphoma: ESMO clinical recommendations for diagnosis, treatment and follow‐up. Annals of Oncology 2008;19(Suppl 2):ii72‐6. [PUBMED: 18456777]

Egger 2001

Egger M, Smith GD, Sterne JA. Uses and abuses of meta‐analysis. Clinical Medicine 2001;1(6):478‐84. [PUBMED: 11792089]

Gardner 2009

Gardner JM, Evans KG, Musiek A, Rook AH, Kim EJ. Update on treatment of cutaneous T‐cell lymphoma. Current Opinion in Oncology 2009;21(2):131‐7. [PUBMED: 19532014]

Grant 2009

Grant B. Biotech Baddies. The Scientist 2009;23(4):48. [http://classic.the‐scientist.com/article/display/55525/]

Hanley 1983

Hanley JA, Lippman‐Hand A. If nothing goes wrong, is everything all right? Interpreting zero numerators. JAMA 1983;249(13):1743‐5. [PUBMED: 6827763]

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.

Hollis 1999

Hollis S, Campbell F. What is meant by intention to treat analysis? Survey of published randomised controlled trials. BMJ 1999;319(7211):670‐4. [PUBMED: 10480822]

Hwang 2008

Hwang ST, Janik JE, Jaffe ES, Wilson WH. Mycosis fungoides and Sézary syndrome. Lancet 2008;371(9616):945‐57. [PUBMED: 18342689]

Jaffe 2001

Jaffe ES, Harris NL, Stein H, Vardiman JW, editors. Pathology and Genetics: Tumours of Hematopoetic and Lymphoid Tissues (World Health Organization Classification of Tumours). Lyon: IARC Press, 2001.

Jüni 2001

Jüni P, Altman DG, Egger M. Systematic reviews in health care: Assessing the quality of controlled clinical trials. BMJ 2001;323(7303):42‐6. [PUBMED: 11440947]

Jüni 2002

Jüni P, Holenstein F, Sterne J, Bartlett C, Egger M. Direction and impact of language bias in meta‐analyses of controlled trials: Empirical study. International Journal of Epidemiology 2002;31(1):115‐23. [PUBMED: 11914306]

Kim 1996

Kim YH, Jensen RA, Watanabe GL, Varghese A, Hoppe RT. Clinical stage IA (limited patch and plaque) mycosis fungoides. A long‐term outcome analysis. Archives of Dermatology 1996;132(11):1309‐13. [PUBMED: 8915308]

Kim 2003

Kim YH, Liu HL, Mraz‐Gernhard S, Varghese A, Hoppe RT. Long‐term outcome of 525 patients with mycosis fungoides and Sézary syndrome: Clinical prognostic factors and risk for disease progression. Archives of Dermatology 2003;139(7):857‐66. [PUBMED: 12873880]

Lorincz 1996

Lorincz AL. Cutaneous T‐cell lymphoma (mycosis fungoides). Lancet 1996;347(9005):871‐6. [PUBMED: 8622396]

Miettinen 1985

Miettinen O, Nurminen M. Comparative analysis of two rates. Statistics in medicine 1985;4(2):213‐26. [PUBMED: 4023479]

Moher 1995

Moher D, Jadad AR, Nichol G, Penman M, Tugwell P, Walsh S. Assessing the quality of randomized controlled trials: An annotated bibliography of scales and checklists. Controlled Clinical Trials 1995;16(1):62‐73. [PUBMED: 7743790]

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. Preferred reporting items for systematic reviews and meta‐analyses: the PRISMA statement. Journal of Clinical Epidemiology 2009;62(10):1006‐12. [PUBMED: 19631508]

Molin 1979

Molin L, Thomsen K, Volden G. Mycosis fungoides plaque stage treated with topical nitrogen mustard with and without attempts at tolerance induction: report from the Scandinavian mycosis fungoides study group. Acta Dermato‐Venereologica 1979;59(1):64‐8. [PUBMED: 84470]

Morales 2000

Morales Suárez‐Varela MM, Llopis González A, Marquina Vila A, Bell J. Mycosis fungoides: Review of epidemiological observations. Dermatology 2000;201(1):21‐8. [PUBMED: 10971054]

Olsen 2007

Olsen E, Vonderheid E, Pimpinelli N, Willemze R, Kim Y, Knobler R, et al. Revisions to the staging and classification of mycosis fungoides and Sézary syndrome: A proposal of the International Society for Cutaneous Lymphomas (ISCL) and the cutaneous lymphoma task force of the European Organization of Research and Treatment of Cancer (EORTC). Blood 2007;110(6):1713‐22. [PUBMED: 17540844]

Olsen 2011

Olsen EA, Whittaker S, Kim YH, Duvic M, Prince HM, Lessin SR, et al. Clinical end points and response criteria in mycosis fungoides and Sezary syndrome: a consensus statement of the International Society for Cutaneous Lymphomas, the United States Cutaneous Lymphoma Consortium, and the Cutaneous Lymphoma Task Force of the European Organisation for Research and Treatment of Cancer. Journal of Clinical Oncology 2011;29(18):2598‐607. [PUBMED: 21576639]

Sausville 1988

Sausville EA, Eddy JL, Makuch RW, Fischmann AB, Schechter GP, Matthews M, et al. Histopathologic staging at initial diagnosis of mycosis fungoides and the Sézary syndrome. Definition of three distinctive prognostic groups. Annals of Internal Medicine 1988;109(5):372‐82. [PUBMED: 3408055]

Scheffer 1980

Scheffer E, Meijer CJ, Van Vloten WA. Dermatopathic lymphadenopathy and lymph node involvement in mycosis fungoides. Cancer 1980;45(1):137‐48. [PUBMED: 7350998]

Schlaak 2012

Schlaak M, Pickenhain J, Theurich S, Skoetz N, von Bergwelt‐Baildon M, Kurschat P. Allogeneic stem cell transplantation versus conventional therapy for advanced primary cutaneous T‐cell lymphoma. Cochrane Database of Systematic Reviews 2012, Issue 1. [DOI: 10.1002/14651858.CD008908.pub2]

Spitzer 1981

Spitzer WO, Dobson AJ, Hall J, Chesterman E, Levi J, Shepherd R, Battista RN, Catchlove BR. Measuring the quality of life of cancer patients: a concise QL‐index for use by physicians. Journal of Chronic Diseases 1981;34(12):585‐97. [PUBMED: 7309824]

Trautinger 2006

Trautinger F, Knobler R, Willemze R, Peris K, Stadler R, Laroche L, et al. EORTC consensus recommendations for the treatment of mycosis fungoides/Sézary syndrome. European Journal of Cancer 2006;42(8):1014‐30. [PUBMED: 16574401]

van Doorn 2000

van Doorn R, Van Haselen CW, van Voorst Vader PC, Geerts ML, Heule F, de Rie M, et al. Mycosis fungoides: Disease evolution and prognosis of 309 Dutch patients. Archives of Dermatology 2000;136(4):504‐10. [PUBMED: 10768649]

van Scott 1973

Van Scott EJ, Kalmanson JD. Complete remissions of mycosis fungoides lymphoma induced by topical nitrogen mustard (HN2). Control of delayed hypersensitivity to HN2 by desensitization and by induction of specific immunologic tolerance. Cancer 1973;32(1):18‐30. [PUBMED: 4577503]

Verhagen 1998

Verhagen AP, de Vet HC, de Bie RA, Kessels AG, Boers M, Bouter LM, et al. The Delphi list: A criteria list for quality assessment of randomized clinical trials for conducting systematic reviews developed by Delphi consensus. Journal of Clinical Epidemiology 1998;51(12):1235‐41. [PUBMED: 10086815]

Vonderheid 2006

Vonderheid EC, Pena J, Nowell P. Sézary cell counts in erythrodermic cutaneous T‐cell lymphoma: Implications for prognosis and staging. Leukemia & Lymphoma 2006;47(9):1841‐56. [PUBMED: 17064997]

Wain 2003

Wain EM, Orchard GE, Whittaker SJ, Spittle MF, Russell‐Jones R. Outcome in 34 patients with juvenile‐onset mycosis fungoides: A clinical, immunophenotypic, and molecular study. Cancer 2003;98(10):2282‐90. [PUBMED: 14601100]

Weinstock 1988

Weinstock MA, Horm JW. Mycosis fungoides in the United States. Increasing incidence and descriptive epidemiology. JAMA 1988;260(1):42‐6. [PUBMED: 3379722]

Weinstock 1999

Weinstock MA, Reynes JF. The changing survival of patients with mycosis fungoides: A population‐based assessment of trends in the United States. Cancer 1999;85(1):208‐12. [PUBMED: 9921994]

Whittaker 2003

Whittaker SJ, Marsden JR, Spittle M, Russell Jones R. Joint British Association of Dermatologists and U.K. Cutaneous Lymphoma Group guidelines for the management of primary cutaneous T‐cell lymphomas. British Journal of Dermatology 2003;149(6):1095‐1107. [PUBMED: 14696593]

Willemze 1997

Willemze R, Kerl H, Sterry W, Berti E, Cerroni L, Chimenti S, et al. EORTC classification for primary cutaneous lymphomas: A proposal from the Cutaneous Lymphoma Study Group of the European Organization for Research and Treatment of Cancer. Blood 1997;90(1):354‐71. [PUBMED: 9207472]

Willemze 2005

Willemze R, Jaffe ES, Burg G, Cerroni L, Berti E, Swerdlow SH, et al. WHO‐EORTC classification for cutaneous lymphomas. Blood 2005;105(10):3768‐85. [PUBMED: 15692063]

Wu 2009

Wu PA, Kim YH, Lavori PW, Hoppe RT, Stockerl‐Goldstein KE. A meta‐analysis of patients receiving allogeneic or autologous hematopoietic stem cell transplant in mycosis fungoides and Sezary syndrome. Biology of Blood & Marrow Transplantation 2009;15(8):982‐90.

Zackheim 1999

Zackheim HS, Amin S, Kashani‐Sabet M, McMillan A. Prognosis in cutaneous T‐cell lymphoma by skin stage: Long‐term survival in 489 patients. Journal of the American Academy of Dermatology 1999;40(3):418‐25. [PUBMED: 10071312]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Child 2004

Methods

This was a randomised, open‐label, cross‐over trial, which lasted 12 months.

Participants

The study recruited 16 participants (10 were in the PUVA‐first group; 6 were in the ECP‐first group) with plaque‐stage (1B ⁄ T2, Bunn Lamberg 1B) MF and a peripheral blood T‐cell clone (detected by polymerase chain reaction (PCR)‐single‐strand conformational polymorphism (SSCP) methodology), but with no evidence of lymph node involvement.

Demographics of the included participants

  • 12 men and 4 women

  • Mean age (range) = 65.1 years (37 to 80 years)

  • 8 participants were lost to follow up (3/10 = 30% in the PUVA‐first group; 5/6 = 83% in the ECP‐first group), resulting in 8 participants evaluated (7 in the PUVA‐first group and 1 in the ECP‐first group)

Exclusion criteria of the trial

  • Haemoglobin < 11 g ⁄dL

  • Cardiac, liver, or renal impairment or positive HTLV‐1 (human T‐lymphotropic virus type 1) serology

  • Pregnancy

  • Progressive disease

Interventions

  • The PUVA‐first group was given PUVA twice a week for 3 months followed by ECP once monthly for 6 months (doses not reported).

  • The ECP‐first group was given ECP once monthly for 6 months followed by PUVA twice a week for 3 months (doses not reported).

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

  3. Survival rates (assessed 2 to 21 months after the end of the intervention)

  4. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size

Notes

No funding body was declared.

This study was conducted at Skin Tumour Clinic, St John's Institute of Dermatology, United Kingdom.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A statistician generated the randomisation.

Allocation concealment (selection bias)

Unclear risk

It was unclear whether used envelopes were sealed and opaque.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

This was not possible because of different types of interventions.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

There were insufficient information to permit judgement.

Incomplete outcome data (attrition bias)
All outcomes

High risk

8/16 participants (50%) were lost to follow up: 3/10 participants in the PUVA‐first group and 5/6 participants in the ECP‐first group.

Selective reporting (reporting bias)

Unclear risk

This was unknown. We contacted the corresponding author for additional outcome data, but we received no reply within 4 weeks.

Other bias

High risk

It was unclear if concomitant medication was permitted.

Chong 2004

Methods

This was a randomised, double‐blind, parallel‐group trial, which lasted 4 months.

Participants

The study recruited 4 participants (3 in the intervention group and 1 in the control group) with histologically‐proven MF plaque stage 1B MF (T2N0M0).

Demographics of the included participants

  • 4 men and 0 women

  • Mean age (range) = 54 years (39 to 61 years)

  • 0 participants were lost to follow up

Exclusion criteria of the trial

These were not reported.

Interventions

  • The intervention group was given imiquimod 5% applied daily; the contact time was 8 hours for 16 weeks.

  • The control group was given placebo cream applied daily; the contact time was 8 hours for 16 weeks.

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size) (assessed 16 weeks after the end of the intervention)

  3. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size (assessed 16 weeks after end of intervention)

  4. Rare adverse effects

Notes

The funding body was 3M Health Care Limited supplied Aldara.

This study was conducted in the United Kindom (1 centre).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study did not provide information about this.

Allocation concealment (selection bias)

Unclear risk

The study did not provide information about this.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was described as double‐blind.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

There were insufficient information to permit judgement.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts were reported.

Selective reporting (reporting bias)

Unclear risk

This was unknown. We contacted the corresponding author for additional outcome data, but we received no reply within 4 weeks.

Other bias

High risk

The study had a very small sample size (n = 4): 3 participants in the intervention group, and 1 participant in the placebo group.

Duvic 2001

Methods

This was a randomised, open‐label, parallel‐group trial, which lasted 16 weeks.

Participants

The study recruited 58 participants (15 in the low‐dose group with 6.5 mg/m² daily versus 43 in the high‐dose group, which consisted of 28 participants who had 300 mg/m² daily and 15 participants who had 650 mg/m² daily) with histologically‐confirmed mycosis fungoides:

  • CTCL stage I through IIA refractory to therapy;

  • the participant was intolerant to therapy; or

  • the participant had reached a 6‐month or greater response plateau under at least 2 of the following qualifying prior therapies: phototherapy (psoralen‐UVA or UVB), total body skin electron beam irradiation therapy, topical chemotherapy (mechlorethamine [nitrogen mustard] or carmustine therapy), or interferon, or systemic cytotoxic chemotherapy.

Demographics of the included participants

  • 40 men and 18 women

  • Mean age (range) = 64 years (24 to 88 years)

  • Stages of disease: IA: 17, IB: 34, IIA: 6, IIB: ?

  • 142 participants (72.4%) were lost in total to follow up

Exclusion criteria of the trial

  • < 18 years

  • Systemic antibiotic or topical therapy (for 2 weeks prior)

  • Phototherapy (for 3 weeks prior)

  • Systemic cancer therapy, electron beam, or other experimental therapy (for 30 days prior)

  • Etretinate therapy (for 1 year prior)

  • Other oral retinoid therapies (for 3 months prior)

Interventions

  • The low‐dose group received bexarotene 6.5 mg/m²/d capsules (10 mg or 75 mg) once daily with their evening meal.

  • The high‐dose group received bexarotene 650 (reduced to 500) mg/m²/d capsules (10 mg or 75 mg) once daily with their evening meal or bexarotene 300 mg/m²/d capsules (10 mg or 75 mg) once daily with their evening meal after adjusting the dose during the trial.

Outcomes

Outcomes of the trial

  1. Quality of life measured by the Spitzer quality of life questionnaire and a non‐validated CTCL quality of life questionnaire

  2. Common adverse effects of the treatments

  3. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

  4. Relapse defined as the time period after remission when the eruption reappears after short‐term clearance

  5. Survival rates (assessed 4 weeks after the end of the intervention)

  6. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size

  7. Rare adverse effects

Notes

The high‐dose was reduced twice (from 650 to > 500, and from 500 to > 300) during the trial; there was separate assessments of the high‐dose and "optimal" dose groups. 11/15 participants in the low‐dose group crossed over to high‐dose therapy after 8 weeks of treatment. Randomisation discontinued during the trial after interim analysis and was reinstalled after consideration by the U.S. Food and Drug Administration (FDA).

The dropout rate for withdrawals was 72.4%.

Dr Duvic was funded by research grants from Ligand Pharmaceuticals, San Diego California, USA (R21‐CA74117); from the National Institutes of Health, Bethesda, Maryland; and from the MD Anderson Cancer Centre (CA16672‐22).

This study was conducted in 18 CTCL clinics at academic referral centres in the USA, Canada, Australia, and Europe.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study did not provide information about this. We sought information, but received no response.

Allocation concealment (selection bias)

Unclear risk

The study did not provide information about this. We sought information, but received no response.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Blinding was not possible because of the number of capsules given."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The physician was blinded to CA response because it was calculated from the case report form."

Incomplete outcome data (attrition bias)
All outcomes

High risk

The dropout rate for withdrawals was 72.4%.

Selective reporting (reporting bias)

Unclear risk

This was unknown. We contacted the corresponding author for additional outcome data, but we received no reply within 4 weeks.

Other bias

High risk

The initial dose in the intervention group was reduced from 650 mg/m²/day to 500 mg/m²/day to 300 mg/m²/day due to adverse reactions. The study discontinued randomisation.

Duvic 2001a

Methods

This was a randomised, double‐blind, parallel‐group trial, which lasted 24 weeks.

Participants

The study recruited 89 participants (43 in the intervention group and 46 in the control) with histologically‐confirmed MF manifested as patches with or without plaques (stage I), but without enlarged nodes, visceral involvement, or generalised erythroderma.

Demographics of the included participants

  • 45 men and 44 women

  • Mean age in the intervention group = 60.2 years

  • Mean age in the control group = 58.1 years

  • Stages of disease: IA: 45, IB: 44

  • 25 participants were lost to follow up (14/43 = 32.6% in the intervention group and 11/46 = 23.9% in the control group; all participants were evaluated (last observation carried forward)

Exclusion criteria of the trial

  • Pregnancy/lactation

  • Age < 18 years

  • PUVA treatment within 2 weeks prior to enrolment

  • Electron beam therapy within 4 weeks prior to enrolment

  • Karnofsky Performance Status < 70%

  • Life expectancy >12 months

  • Systemic cytotoxic chemotherapy other than methotrexate for CTCL prior to enrolment (however, interferon‐alpha and interleukin‐2 were allowed)

  • Hypersensitivity to any of the components of the topical formulation

  • Chronic eczema, including contact dermatitis or atopic dermatitis

  • Any other known or suspected immunodeficiency disorder

  • An acute systemic illness or chronic illness that would limit the ability to complete the protocol

  • Any baseline laboratory values outside normal ranges considered clinically significant

  • Participation in a study of any systemic experimental drug within the last 2 months

  • An intercurrent illness that intermittently or chronically required corticosteroid treatment

Interventions

  • The intervention group was given BCX‐34 dermal cream 1% twice daily, which was applied in a thin film to the entire skin surface and gently massaged into the skin.

  • The control group was given vehicle cream twice daily, which was applied in a thin film to the entire skin surface and gently massaged into the skin.

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

  3. Survival rates

  4. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size

Notes

Funding came from BioCryst Pharmaceuticals, Inc (Birmingham, Alabama, USA).

This study was conducted in 10 tertiary care centres in the USA.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

No specific information was given. The data were managed by a third party (Quintiles Inc).

Allocation concealment (selection bias)

Low risk

No specific information was given. The data were managed by a third party (Quintiles Inc).

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Only the sponsor was able to un‐blind in case of withdrawal.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Only the sponsor was able to un=‐blind in case of withdrawal.

Incomplete outcome data (attrition bias)
All outcomes

High risk

ITT analysis was carried out and last observation carried forward. 24/89 participants were lost to follow up: 14/43 (33%) in the intervention group and 11/46 (24%) in the placebo group.

Selective reporting (reporting bias)

Unclear risk

This was unknown. We contacted the corresponding author for additional outcome data, but we received no reply within 4 weeks.

Other bias

Low risk

None were found.

Guitart 2002

Methods

This was a randomised, open‐label, parallel‐group trial, which lasted 24 weeks.

Participants

The study recruited 43 participants (20 in the high‐dose group and 23 in the low‐dose group) with histologically‐proven mycosis fungoides stages IB and IIA, with lymph node biopsies negative for MF involvement.

Demographics of the included participants

  • 25 men and 18 women

  • Mean age (range) = 57.5 years

  • Stages of disease: IB: 36, IIA 7

  • 4 participants were lost to follow up (1/20 = 5% in the high‐dose group; 3/23 = 13% in the control group), resulting in 39 participants evaluated (19 in the high‐dose group and 20 in the low‐dose group)

Exclusion criteria of the trial

These were not reported.

Interventions

  • The high‐dose group was given bexarotene 300 mg/day (starting week 1) and PUVA (starting week 2) and fenofibrate 54 mg/day (starting week 0).

  • The low‐dose group was given bexarotene 150 mg/day (starting week 1) and PUVA (starting week 2) and fenofibrate 54 mg/day (starting week 0).

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

  3. Relapse defined as the time period after remission when the eruption reappears after short‐term clearance (assessed 6 months after the end of the intervention)

  4. Survival rates

  5. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size

Notes

Data were abstracted from the manuscript sent by the corresponding author; the sample size was smaller than planned according to the author. Dose reduction was necessary in 14/39 participants because of hyperlipidaemic side‐effects, although antilipidaemic therapy was prescribed for each participant.

This study was conducted in 12 tertiary care centres in the USA.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study did not provide information about this. We sought information but got no response.

Allocation concealment (selection bias)

Unclear risk

The study did not provide information about this. We sought information but got no response.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

This was an open‐label trial.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No separate outcome assessor was mentioned.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT analysis was carried out: 4 participants (10%) dropped out after randomisation without receiving a single treatment; 1 person (5%) in the 300 mg/day bexarotene group dropped out for "other" reason.

Selective reporting (reporting bias)

Low risk

We contacted the corresponding author for additional outcome data, and we had an email response. We were sent a manuscript of unpublished data, and we had further confirmation by email that all outcomes were reported.

Other bias

High risk

The study had a smaller sample size than planned; dose reduction was necessary in 14/39 participants due to hypertriglyceridaemia, although preventive antilipidaemic therapy was prescribed for each participant.

Kaye 1989

Methods

This was a randomised, open‐label, parallel‐group trial.

Participants

The study recruited 103 participants (52 in the combined‐therapy group and 51 in the conservative‐therapy group) with histologically‐proven MF of all stages.

Demographics of the included participants

  • 69 men and 34 women

  • Age < 60 years: 65 participants; ≥ 60 years: 38 participants

  • Stages of disease: IA: 6, IB: 16, IIA: 9, IIB: 12; III: 2, IVA: 42, IVB: 16

  • 8 participants were lost to follow up (6/52 = 11.5% in the combined‐therapy group; 2/51 = 3.9% in the conservative‐treatment group), resulting in 103 participants evaluated (last observation carried forward; participants were suspected to be still alive)

Exclusion criteria of the trial

  • Eastern Cooperative Oncology Group Performance Status > 3 (bedridden participant) related to other causes than MF

  • Prior systemic chemotherapy

  • Prior total‐skin electron‐beam therapy

Interventions

  • The combined‐therapy group was given electron‐beam radiation (3000 to 3200 cGy additional boost of 1000 to 1500 cGy to the top of the head, perineum, and soles of the feet) and parenteral chemotherapy (cyclophosphamide 500 mg/m² (day 1), doxorubicin 50 mg/m² (day 1), etoposide 100 mg/m² (day 1 to 3), vincristine 1.4 mg/m², with a maximum dose of 2 mg (day 1).

  • The conservative group was given topical treatment with 10 mg mechlorethamine applied to the entire skin alone or in combination with sequential escape therapies in case of visceral involvement or progressive disease:

a) oral methotrexate (20 mg/m² p.o. twice weekly for stage IVB participants)

b) PUVA (oral methoxsalen 0.6 mg/kg body weight followed by UVA light therapy 3 x/week)

c)  electron‐beam therapy (as described in the combined‐therapy group) combined with methotrexate (as described above)

d) systemic chemotherapy (as described in the combined‐therapy group)

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

  3. Relapse defined as the time period after remission when the eruption reappears after short‐term clearance

  4. Disease‐free interval

  5. Survival rates (assessed more than 5 years after the end of the intervention)

  6. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size

Notes

The funding body was not declared.

This study was conducted in 7 secondary/tertiary care centres in the USA.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Stratified block randomisation was undertaken.

Allocation concealment (selection bias)

Unclear risk

The study did not provide information about this. We sought information but got no response.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

This was not possible because of different interventions used.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study did not provide information about this. We sought information but got no response.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT analysis and last observation carried forward was carried out. There were 6/52 (12%) dropouts in the combined‐therapy group (2 refused to receive treatment; 1 withdrew because of congestive heart failure; 1 withdrew because of residual cutaneous disease; and 2 refused treatment after clinical response) and 2/51 (4%) in conservative‐treatment group (no reasons were stated).

Selective reporting (reporting bias)

Unclear risk

This was unknown. We contacted the corresponding author for additional outcome data, and the author requested original data from their former employer, but the data were not available so far.

Other bias

Unclear risk

It was unclear if previous treatment was stopped.

Olsen 2001

Methods

This was a randomised, parallel‐group trial, which lasted 6 months.

Participants

The study recruited 71 participants (35 in the low‐dose group and 36 in the high‐dose group) with histologically‐proven mycosis fungoides type with ≥ 20% of lymphocytes within the skin biopsy stain positively for CD25 by immunohistochemistry. Further inclusion criteria was as follows: stage Ib‐III CTCL (CTCL Cooperative Group staging) recurred or persisted after ≥ 4 previous treatments for CTCL (excluding topical or systemic corticosteroids) or stage IVa CTCL participants who failed at least 1 previous therapy study consideration,

and Eastern Cooperative Oncology Group Performance Status of 0, 1, or 2. Lymph node involvement was no greater than LN₂, and no CTCL involvement of bone marrow.

Demographics of the included participants

  • 37 men and 34 women

  • Mean age (range) = 61 years (26 to 90 years)

  • Stages of disease: IB:16, IIA: 10, IIB: 19, III: 11

  • 58% of the participants were lost to follow up in total, resulting in 30 participants evaluated

Exclusion criteria of the trial

  • Age < 18 years

  • Pregnancy/lactation

  • Disagreement to practice contraception

  • High‐grade large‐cell, poorly‐differentiated tumours, or both

  • Positive test for HIV, HTCLV‐1, or hepatitis B or C

  • Uncontrolled hypertension

  • Any signs of active systemic infection

  • Previous treatment with IL‐2 fusion proteins

Interventions

  • The intervention group was given 9 µg/kg/day denileukin diftitox intravenous infusion over 15 to 60 minutes for 5 consecutive days every 3 weeks.

  • The control group was given 18 µg/kg/day denileukin diftitox intravenous infusion over 15 to 60 minutes for 5 consecutive days every 3 weeks.

Outcomes

Outcomes of the trial

  1. Quality of life measured by the Functional Assessment of Cancer Therapy‐general (FACT‐G) questionnaire

  2. Common adverse effects of the treatments

  3. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

  4. Survival rates (assessed 90 days after the end of the intervention)

  5. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size

  6. Rare adverse effects

Notes

Only 42% of all randomised participants received 8 courses of treatment as planned. There was no comparison reported between both treatment groups regarding QoL from baseline to the end of the study (only subgroup analyses of responders vs non‐responders).

The funding body was Seragen, Inc (a wholly‐owned subsidiary of Ligand Pharmaceuticals Inc, San Diego, CA).

This study was conducted in 20 centres across the USA.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was stratified by stage of CTCL for multicentre trial. It was likely to have been carried out by a third party and concealed, although this was not formally stated.

Allocation concealment (selection bias)

Unclear risk

Randomisation was stratified by stage of CTCL for multicentre trial. However, it was unclear whether randomisation was concealed.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

This was unlikely to be blinded, since the drug was diluted to a certain minimum concentration in both arms and administered by a pump device for 15 to 60 minutes.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"All responses were verified by an independent panel of physicians [the Data End Point Review Committee]."

Incomplete outcome data (attrition bias)
All outcomes

High risk

41/71 (58%) participants dropped out. Discontinuation was due to adverse events (11/35 (31%) participants in the 9 µg/kg/day group vs 15/36 (42%) in the 18 µg/kg/day group) and treatment failure (6/35 (17%) in the 9 µg/kg/day group vs 2/36 (6%) in the 18 µg/kg/day group).

Selective reporting (reporting bias)

High risk

The quality of life assessment was compared between responders and non‐responders instead of comparing treatment groups. We contacted the corresponding author for additional outcome data, but we received no reply within 4 weeks.

Other bias

Unclear risk

There were insufficient information to permit judgement.

Rook 2010

Methods

This was a randomised, double‐blind (verified by author contact), within‐participant trial, which lasted 6 weeks.

Participants

The study recruited 12 participants (with 1 lesion per treatment): men or non‐pregnant women aged 18 to 70 with stable patch or plaque phase MF of at least 4 months' duration.

Demographics of the included participants

  • 8 men and 4 women

  • Mean age (SD) = 55 years (16.5 years)

  • 0 participants were lost to follow up, resulting in 12 lesions per treatment group

Exclusion criteria of the trial

These were not reported.

Interventions

  • The intervention group was given hypericin (0.05%, 0.1%, or 0.25%) applied twice a week for 24 hours before radiation with visible light (590 to 650 nm): 8 to 20 J/cm² up to 15 minutes, twice weekly, separated by at least 1 day.

  • The control group was given placebo cream applied twice a week for 24 hours before radiation with visible light (590 to 650 nm): 8 to 20 J/cm² up to 15 minutes, twice weekly, separated by at least 1 day.

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

  3. Survival rates

  4. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size

Notes

The study was supported in part by the USA's Department of Energy Merit Review.

Funding came from the Department of Veterans Affairs (Dr Wood) and Vimrx Inc. Disclosure: Dr Rook has been a consultant to Hy BioPharma Inc.

This study was conducted in 4 tertiary care centres in the USA.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study did not provide information about this. We sought information and had an email response: The corresponding author no longer had access to data from the former employer.

Allocation concealment (selection bias)

Unclear risk

The study did not provide information about this. We sought information and had an email response: The corresponding author no longer had access to data from the former employer.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was described in the publication as "double‐blind" and "open‐label". The email response from the author confirmed that the study was double‐blind.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study did not provide information about this. We sought information and had an email response: The corresponding author no longer had access to data from the former employer.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There were no dropouts.

Selective reporting (reporting bias)

Unclear risk

This was unknown. We sought information and had an email response: The corresponding author no longer had access to data from the former employer.

Other bias

Unclear risk

It was unclear if previous treatment was stopped and if concomitant medication was permitted.

Stadler 1998

Methods

This was a randomised, open‐label, parallel‐group trial, which lasted 48 weeks.

Participants

The study recruited 82 participants (40 in the IFN‐α + PUVA group and 42 in the IFN‐α + acitretin group) with small‐ to medium‐sized pleomorphic T‐cell lymphoma or mycosis fungoides stage I or II. The principle investigator (Stadler) stated on author contact that all participants had histologically‐proven mycosis fungoides.

Demographics of the included participants

  • 62 men and 20 women

  • Mean age (range) = 58 years (26 to 82 years)

  • Stages of disease: IA: 36; IB: 28; IIA: 10; IIB: 8

  • 16/98 (16.3%) participants were lost to follow up (distribution in groups not reported)

Exclusion criteria of the trial

These were not reported.

Interventions

  • The IFN‐α + PUVA group were given IFN‐α at a starting dose of 3‐6‐9 MU in week 1 followed by 3 x weekly 9 MU in weeks 2 to 48 and 8‐methoxypsoralen (0.6 mg/kg) 5 x weekly in weeks 1 to 4, 3 x weekly in weeks 5 to 23, 2 x weekly in weeks 24 to 48, with escalating doses beginning with 0.25 J/cm² until minimal erythema dose was reached.

  • The IFN‐α + acitretin group were given IFN‐α as described above and acitretin, 25 mg daily in week 1 and 50 mg daily in weeks 2 to 48.

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

  3. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size

Notes

The funding body was not declared.

This study was conducted in 21 tertiary care centres in Germany, Austria, and Switzerland.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was by a central institution/third party (Estimate GmbH, Augsburg/Germany) and stratified by pretreatment.

Allocation concealment (selection bias)

Low risk

Randomisation was by central institution/third party (Estimate GmbH, Augsburg/Germany) and stratified by pretreatment.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

This was not possible because of different interventions (PUVA vs capsules).

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study did not provide information about this. We sought information but got no response.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The primary analysis was per‐protocol. ITT analysis was also carried out for comparison between study groups regarding complete remission: 16/98 (16%) participants dropped out (6 participants did not receive any treatment; 6 participants had insufficient data monitored; and 4 participants had wrong staging at enrolment); there was no distribution between groups reported. 40/49 participants in the PUVA group and 42/49 participants in the acitretin group were evaluable.

Selective reporting (reporting bias)

Unclear risk

This was unknown. We contacted the corresponding author for additional outcome data, but we received no reply within 4 weeks.

Other bias

Unclear risk

It was unclear if previous treatment was stopped and if concomitant medication was permitted.

Stadler 2006

Methods

This was a randomised parallel‐group trial, which lasted 52 weeks.

Participants

The study recruited 124 participants with cutaneous T‐cell lymphoma (stages lA to IIA) ‐ type mycosis fungoides or small to medium cellular pleomorphic type. The principle investigator (Stadler) stated on author contact that all participants had histologically‐proven mycosis fungoides.

Demographics of the included participants

  • The male/female ratio was not reported

  • The mean age (range) was not reported

  • The stages of disease was not reported

  • 31/124 (25%) participants were lost to follow up, resulting in 93 participants evaluated (50 in the intervention group and 43 in the control group)

Exclusion criteria of the trial

These were not reported.

Interventions

  • The intervention group was given IFN‐α 3 x weekly 9 MU and PUVA: 8‐methoxypsoralen (0.6 mg/kg), 5 x weekly in weeks 1 to 4, 3 x weekly in weeks 5 to 23, 2 x weekly in weeks 24 to 48, with escalating doses beginning with 0.25 J/cm².

  • The control group was given PUVA as described above.

Outcomes

Outcomes of the trial

  1. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

Notes

The funding body was not declared. The disclosure in Stadler 2006 stated: "No significant financial relationships to disclose."

This study was conducted in 26 tertiary care centres in Germany and Switzerland.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study did not provide information about this. We sought information but got no response.

Allocation concealment (selection bias)

Unclear risk

The study did not provide information about this. We sought information but got no response.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Blinding was unlikely since no placebo injections were described.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study did not provide information about this. We sought information but got no response.

Incomplete outcome data (attrition bias)
All outcomes

High risk

31/124 (25%) randomised participants were not evaluable, and no reasons for this were stated.

Selective reporting (reporting bias)

High risk

The only outcome reported was complete remission; there was no report on adverse effects.

We contacted the corresponding author for additional outcome data, but we received no reply within 4 weeks.

Other bias

Unclear risk

It was unclear if previous treatment was stopped and if concomitant medication was permitted.

Thestrup‐Pedersen 1982

Methods

This was a randomised, double‐blind, parallel‐group trial, which lasted 12 months.

Participants

The study recruited 16 participants (8 in the intervention group and 8 in the control group) with histologically‐proven MF van Scott stage II to IV.

Demographics of the included participants

  • 8 men and 8 women

  • Mean age (range) in the intervention group = 67.4 years (53 to 83 years)

  • Mean age in the control group = 65.0 years (47 to 82 years)

  • Stages of disease: II: 14; III: 1; IV: 1

  • 0 participants were lost to follow up

Exclusion criteria of the trial

These were not reported.

Interventions

  • The intervention group was given 40 mg nitrogen mustard daily for 14 days followed by weekly/biweekly treatment 2 units transfer factor biweekly for 1 year. If participants had severe hypersensitivity towards HN₂ or relapse after previous HN₂ treatment, they were treated with PUVA.

  • The control group was given 40 mg nitrogen mustard daily for 14 days followed by weekly/biweekly treatment 2 units inactivated transfer factor biweekly for 1 year. If participants had severe hypersensitivity towards HN₂ or relapse after previous HN₂ treatment, they were treated with PUVA.

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size) (assessed 1 year after the end of the intervention)

  3. Survival rates (assessed 1 year after the end of the intervention)

  4. Improvement defined by clearance of at least 50% of all lesion surfaces, lesions, or tumour size (assessed 1 year after end of intervention)

Notes

The funding body was Landsforeningen til kraeftens bekaempelse (a grant came from the National Institution for Cancer Prevention of Danish Cancer Society).

This study was conducted in the Department of Dermatology, University of Aarhus, Denmark.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study did not provide information about this. We sought information but got no response.

Allocation concealment (selection bias)

High risk

The corresponding trial author confirmed in an email response that the randomisation list was open.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

This was a double‐blind study.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The corresponding trial author confirmed in an email response that the outcome assessors were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No losses to follow up were reported, but the number of participants randomised was not stated.

Selective reporting (reporting bias)

Low risk

This was unknown. We contacted the corresponding author for additional outcome data, and the author responded with a completed data extraction form.

Other bias

High risk

Concomitant treatment was permitted.

Vonderheid 1987

Methods

This was a randomised, double‐blind, within‐participant trial, which lasted 4 weeks.

Participants

The study recruited 6 participants (2 lesions per treatment) with plaque phase MF, MFCG stage nomenclature of 1979 stage IA (T1, Nx, T0, M0), stage IB (T2, Nx, T0, M0), or stage IIA (T2, N1, T0, M0)

Demographics of the included participants

  • 3 men and 3 women

  • Mean age (range) = 59.5 years (33 to 68 years)

  • Stages of disease: IA: 1, IB: 1, IIA: 4

  • 0 participants were lost to follow up

Exclusion criteria of the trial

  • Any topical therapy within 4 weeks prior to the study

  • Any previous systemic cytotoxic therapy

  • Any previous exposure to exogenous interferon or interferon‐inducer

  • History of cardiac disease, pulmonary embolism, or thrombophlebitis

  • History of exposure to radiation in areas of observation

Interventions

  • The intervention group was given IFN‐α 2b injections 106 units 3 times weekly at 2 representative sites.

  • The control group was given placebo injections with isotonic sterile water 3 times weekly at 2 representative sites.

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size) (assessed 4 weeks after the end of the intervention)

Notes

The funding body was not declared.

This study was conducted in a tertiary care centre in the USA.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Lesions were allocated by a random code; no further information was given; information was sought, but we received no response.

Allocation concealment (selection bias)

Unclear risk

Information was sought, but we received no response.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The trial was described as double‐blind for the first part, which we data extracted.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No separate outcome assessor was described.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT analysis was carried out.

There were no dropouts.

Selective reporting (reporting bias)

Unclear risk

This was unknown. We contacted the corresponding author for additional outcome data, but we received no reply within 4 weeks.

Other bias

Unclear risk

There were insufficient information to permit judgement.

Wolff 1985

Methods

This was a randomised, double‐blind, parallel‐group trial, which lasted 8 weeks.

Participants

The study recruited 12 participants (9 from the intervention group and 3 from the control group) with early plaque or patch stage MF (stage IA or IB), with no evidence of physical examination on lymphadenopathy or organomegaly.

Demographics of the included participants

  • 10 men and 2 women

  • Mean age (range) = 56.7 years (39 to 74 years)

  • Stages of disease: IA: 7; IB: 5

  • 0 participants were lost to follow up

Exclusion criteria of the trial

  • Any prior systemic chemotherapy or radiation therapy

  • Not been treated with any topical steroids, nitrogen mustard, or psoralens and UVA for 4 weeks prior to therapy with study medication

Interventions

  • The intervention group was given different interventions for 3 lesions for 4 weeks consisting of:

a) IFN‐α 2MU in superficial dermis 3 times weekly;

b) betamethasone dipropionate ointment 0.05% twice daily; or

c) no treatment.

  • The control group was given different interventions for 3 lesions consisting of:

a) placebo (buffered glycine serum human albumin) in superficial dermis 3 times weekly;

b) betamethasone dipropionate ointment 0.05% twice daily; or

c) no treatment.

Outcomes

Outcomes of the trial

  1. Common adverse effects of the treatments

  2. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

Notes

Partial improvement was reported as mean difference in size of lesions without the possibility to identify participants' improvement according to our defined secondary outcome. Lesions in the IFN‐α group generally improved better than in the placebo group, possibly due to a systemic effect of IFN‐α as discussed by the authors.

The IFN‐α was supplied by Schering Corp.

This study was conducted in a tertiary care centre in Pittsburgh, USA.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study did not provide information about this. We sought information, but received no response.

Allocation concealment (selection bias)

Unclear risk

The study did not provide information about this. We sought information, but received no response.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

This trial had a double‐blind setting.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Histopathologic features of biopsies were assessed without knowledge of the treatment or group.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT analysis was carried out.

There were no dropouts.

Selective reporting (reporting bias)

High risk

Only the mean difference in the decrease of the lesions were reported; no incidence of partial remission (i.e. > 50% reduction of disease) was reported. The corresponding author was contacted for additional outcome data but did not respond within 4 weeks.

Other bias

High risk

The groups were unequal: There was a higher proportion of stage 1B, more men, longer duration of skin disease, and longer time since diagnosis in intervention group

Wozniak 2008

Methods

This was a randomised, open‐label, parallel‐group trial, which lasted 24 weeks.

Participants

The study recruited 29 participants (12 in the intervention group and 17 in the control group) with mycosis fungoides stage IA to IIA.

Demographics of the included participants

  • 12 men and 17 women

  • Median age = 52 years

  • Stages of disease: IA: 14, IB: 6, IIA: 9

  • 0 participants were lost to follow up

Exclusion criteria of the trial

  • Pregnant or lactating women

  • Fertile women not accepting contraception

  • Medical history of melanoma or non‐melanoma skin cancer

  • Concomitant infections

  • Immunodeficiency states

  • Previous heart disease

  • Respiratory insufficiency

  • Chronic renal insufficiency

  • Chronic hepatopathy

  • Epilepsy

  • Depression

  • Leucocytes < 3000, or neutrophiles < 1000, or thrombocytes < 100000, or haemoglobin < 12 gr/dL, or ANA < 1/80

  • Treatment with systemic steroids

  • Altered thyroid hormones

  • Previous resistance to PUVA, IFN‐α, or both

  • Hypersensitivity to IFN‐α

  • Participants under treatment with theophylline, dicumarol, or both

  • Previous total skin electron beam

  • Wash‐up period less than 3 month for IFN‐α, PUVA, or both

  • Wash‐up period less than 1 month for topical treatments

Interventions

  • The intervention group was given PUVA in weeks 1 to 24, 0.6 mg/kg methoxsalen (8‐MOP) 3 times a week, with 2 hours pre UVA irradiation (1 to 2 Jul/cm² according to phototype, increasing to 10 Jul/cm², if tolerated) and IFN‐α week 1: 3, 6, and 9 MU (Monday, Wednesday, Friday), weeks 2 to 24: 9 MU 3 times a week).

  • The control group was given PUVA as described above.

Outcomes

Outcomes of the trial

  1. Percentage of participants demonstrating clearance (defined by clearance of at least 90% of all lesion surfaces, lesions, or tumour size)

  2. Relapse defined as the time period after remission when the eruption reappears after short‐term clearance

Notes

The funding body was Ministerio de Ciencia y Tecnologia (BIO2000‐0275‐C02 ⁄01‐⁄02, SAF2001‐0060, SAF2005‐00221), Comunidad Autonoma de Madrid (CAM 08.1 ⁄0011 ⁄2001.1), and the Ministerio de Sanidad y Consumo (FISP05 ⁄1710, FIS 01‐0035, G03 ⁄179, PI051623) RETICS, Spain.

The author, MBW, was supported by FISP05 ⁄1710, and LT was supported by grants from the CNIO and the Higher Education Authority of Ireland, St James Hospital, Dublin.

Participants were categorised to responders and non‐responders instead of treatment groups.

Some information was taken from the clinicaltrials.gov website (NCT00630903).

The main primary aim of the study was to examine the gene expression profiles of primary skin biopsies from these participants.

This study was conducted in 9 tertiary care hospitals in Madrid, Spain.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study did not provide information about this. We sought information, but received no response.

Allocation concealment (selection bias)

Unclear risk

The study did not provide information about this. We sought information, but received no response.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

This was taken from the previous version of the NCT00630903 protocol: The study was described as open‐label.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study did not provide information about this. We sought information, but received no response.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT analysis was carried out.

There were no dropouts.

Selective reporting (reporting bias)

High risk

Participants were characterised and divided into responders and non‐responders instead of treatment groups.

We contacted the corresponding author for additional outcome data, but we received no reply within 4 weeks.

Other bias

High risk

Some information was taken from protocol NCT00630903 (www.clinicaltrials.gov). The study was described as terminated due to accrual.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Argyropoulos 1979

There was no relevant end point according to the protocol report.

Breneman 1991

There was no relevant end point according to the protocol report.

Cooper 1994

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Currie 1980

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Dang 2007

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Doan 1958

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Dueck 2010

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Fawzi 2010

There was not enough information to confirm inclusion criteria, and we had no reaction when we attempted to contact the corresponding author.

Fisher 1993

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

JapicCTI‐050041

There was not enough information to confirm inclusion criteria, and we had no reply when we attempted to contact the corresponding author.

Kaung 1969

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Kujawska 2003

There was not enough information to confirm inclusion criteria, and we had no reply when we attempted to contact the corresponding author.

Kuzel 2010

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Lansigan 2010

There was not enough information to abstract data from the publication.

NCT00054171

An email response confirmed that the study was not completed. (The Principal Investigator passed away).

Neering 1972

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Negro‐Vilar 2007

There was not enough information to confirm inclusion criteria, and we had no reply when we attempted to contact the corresponding author.

No authors listed 1982

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Olsen 1986

This study was not a randomised controlled trial.

Pan 2007

There was not enough information to confirm inclusion criteria, and we had no reply when we attempted to contact the corresponding author.

Peugeot 1995

This was scientific fraud (see Grant 2009).

Plettenberg 2001

This was a report of an ongoing trial; there was not enough information to confirm inclusion criteria, and we had no reply when we attempted to contact the corresponding author.

Prince 2010

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Schrag 1997

There was no relevant end point according to the protocol report.

Simon 2010

This study explicitly excluded MF.

Thomsen 1977

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available. This was identified by Molin 1979 and retrieved as a reference in the adverse event search.

Wain 2005

There was not enough information to confirm inclusion criteria, and we had no reply when we attempted to contact the corresponding author.

Wiernik 1998

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Zubrod 1960

Less than 90% of enrolled participants had Alibert‐Bazin‐type MF, and no subgroup analysis was available.

Characteristics of studies awaiting assessment [ordered by study ID]

Foss 2011

Methods

This is a further report about data from Olsen 2001. This is a randomised, parallel‐group trial, which lasted 6 months.

Participants

Olsen 2001:

The study recruited 71 participants (35 in the low‐dose group and 36 in the high‐dose group) with histologically‐proven mycosis fungoides type with ≥ 20% of lymphocytes within the skin biopsy stain positively for CD25 by immunohistochemistry. Further inclusion criteria was as follows: stage Ib‐III CTCL (CTCL Co‐operative Group staging) recurred or persisted after ≥ 4 previous treatments for CTCL (excluding topical or systemic corticosteroids) or stage IVa CTCL participants who failed at least 1 previous therapy study consideration,

and Eastern Cooperative Oncology Group Performance Status of 0, 1, or 2. Lymph node involvement was no greater than LN₂, and no CTCL involvement of bone marrow.

Demographics of the included participants

  • 37 men and 34 women

  • Mean age (range) = 61 years (26 to 90 years)

  • Stages of disease: IB:16, IIA: 10, IIB: 19, III: 11

  • 58% of the participants were lost to follow up in total, resulting in 30 participants evaluated

Exclusion criteria of the trial

  • Age < 18 years

  • Pregnancy/lactation

  • Disagreement to practice contraception

  • High‐grade large‐cell, poorly‐differentiated tumours, or both

  • Positive test for HIV, HTCLV‐1, or hepatitis B or C

  • Uncontrolled hypertension

  • Any signs of active systemic infection

  • Previous treatment with IL‐2 fusion proteins

Interventions

  • The intervention group was given 9 µg/kg/day denileukin diftitox intravenous infusion over 15 to 60 minutes for 5 consecutive days every 3 weeks.

  • The control group was given 18 µg/kg/day denileukin diftitox intravenous infusion over 15 to 60 minutes for 5 consecutive days every 3 weeks.

Outcomes

Outcomes of the trial

  1. Interleukin‐2 receptor‐α (CD25) expression

  2. Overall response rate

  3. Complete response/complete clinical response

  4. Median response duration

Notes

This was a meeting abstract; the study also provides data for the excluded trial by Prince 2010.

This study was conducted in 20 centres across the USA.

Lessin 2011

Methods

This was a randomised non‐inferiority trial, which lasted 12 months.

Participants

This study recruited 260 stage I‐IIA mycosis fungoides participants.

Interventions

  • The intervention group was given manufactured 0.02% nitrogen mustard gel.

  • The control group was given compounded 0.02% nitrogen mustard petrolatum‐based ointment.

Outcomes

Outcomes of the trial

  1. Response rate according to the Composite Assessment of Index Lesion Severity (CAILS) and a Severity‐weighted assessment tool (SWAT)

  2. Adverse events

Notes

This was a meeting abstract; please see also NCT00168064 in the 'Characteristics of ongoing studies' section.

Characteristics of ongoing studies [ordered by study ID]

NCT00056056

Trial name or title

A Randomized, Open‐Label Phase III Trial to Evaluate the Efficacy and Safety of Bexarotene (Targretin) Capsules Combined With PUVA, Compared to PUVA Treatment Alone in Patients With Mycosis Fungoides

Methods

This is a randomised, open‐label, multicentre study. Participants are stratified according to participating centre, age (60 and under vs over 60), and stage of disease (IB vs IIA). Participants are randomised to 1 of 2 treatment arms.

Participants are followed every 8 weeks until the first documented progression or relapse.

Projected accrual

A total of 145 participants will be accrued for this study within 25 months.

Participants

Participants aged 18 years and older are eligible for this study.

Both genders are eligible for this study.

Healthy volunteers are not accepted.

Disease characteristics

  • Histologically‐confirmed mycosis fungoides stage IB or IIA confirmed by current or prior diagnostic lesion biopsy

Participant characteristics

  • Over 18 years of age

  • Performance Status: Karnofsky 60% to 100%

  • Life expectancy: not specified

  • Haematopoietic:

    • White blood cells (WBC) at least 2,000/mm^3

    • Haemoglobin at least 9 g/dl

  • Hepatic:

    • Bilirubin no greater than 1.5 times upper limit of normal (ULN)

    • AST (aspartate transaminase) and ALT (alanine transaminase) no greater than 2.5 times ULN

  • Renal:

    • Creatinine no greater than 2 times ULN

    • Calcium no greater than 11.5 mg/dL

  • Cardiovascular:

    • No New York Heart Association grade III or IV cardiac insufficiency

  • Other:

    • Not pregnant or nursing

    • Fertile participants must use effective contraception during and for at least 3 months after the study

    • Participation* NOTE: *Women using hormonal contraception must also use a non‐hormonal treatment

    • Fasting triglycerides normal (prior antilipaemic agents allowed to reach normalisation)

    • Willing and able to avoid prolonged exposure to the sun

    • Willing to limit sun exposure on day of PUVA therapy

    • No prior intolerance of or unresponsiveness to PUVA therapy

    • No other prior or concurrent malignant tumour except adequately treated carcinoma in situ of the cervix or basal cell or squamous cell skin cancer

    • No prior pancreatitis

    • No other concurrent serious illness or infection that would preclude study participation

    • No concurrent excessive alcohol consumption

    • No photosensitivity due to intrinsic (e.g. lupus) or extrinsic (e.g. photosensitive drugs) factors

    • No psychological, familial, sociological, or geographical condition that would preclude study compliance

    • No known contraindications to study drug

    • No known hypersensitivity to retinoids or hypervitaminosis A

    • No uncontrolled diabetes mellitus

    • No uncontrolled thyroid disease

Prior concurrent therapy

  • Biologic therapy:

    • At least 3 months since prior interferon therapy

  • Chemotherapy

    • No prior systemic combination chemotherapy

    • No prior participation in another study of bexarotene

    • At least 3 months since prior topical chemotherapy

  • Endocrine therapy

    • At least 1 month since prior topical corticosteroids

  • Radiotherapy

    • At least 6 months since prior total skin electron beam therapy

    • At least 1 month since prior superficial radiotherapy

  • Surgery

    • Not specified

  • Other

    • Ultraviolet light therapy using methoxsalen wth or without bexaroten

    • At least 30 days since prior participation in another investigational drug study

    • At least 3 months since prior photopheresis

    • At least 1 month since prior UVB/PUVA phototherapy

    • At least 1 month since prior retinoid class drugs

    • At least 1 month since prior beta‐carotene compounds

    • At least 1 month since other prior topical medications (e.g. tar baths)

    • No prior participation in this study

    • No other concurrent anticancer therapy

    • No other concurrent investigational drug therapy

    • No concurrent drugs associated with pancreatic toxicity or known to increase triglyceride concentration

Interventions

  • Arm I: Participants receive PUVA comprising oral methoxsalen given 2 hours before whole body ultraviolet A therapy. PUVA is given 3 times per week.

  • Arm II: Participants receive oral bexarotene once daily and PUVA as in arm I. In both arms, treatment repeats for up to 16 weeks in the absence of complete clinical response, disease progression, or unacceptable toxicity.

Outcomes

Primary outcomes of the trial

  • Overall response rate (complete clinical response [CCR) and partial response [PR]) [designated as safety issue: no]

Secondary outcomes of the trial

  • Cumulative dose of UVA required to achieve CCR [designated as safety issue: no]

  • Number of PUVA sessions necessary to achieve a CCR [designated as safety issue: no]

  • Duration of CCR as measured by Logrank every 4 weeks during treatment and then every 8 weeks until progression [designated as safety issue: no]

  • Time‐to‐relapse [designated as safety issue: no]

  • Safety as assessed by CTC v2.0 every 4 weeks during treatment, then every 8 weeks [designated as safety issue: no]

  • Percentage of dropouts as measured by the percentage of cases not completing treatment due to toxicity at the completion of treatment [designated as safety issue: no]

Starting date

January 2003

Contact information

Sponsors and collaborators

  • European Organization for Research and Treatment of Cancer

Investigators

  • Investigator: Sean J. Whittaker, MD, St. Thomas' Hospital

Notes

Other Study ID Numbers: CDR0000271933, EORTC‐21011

NCT00091208

Trial name or title

A Phase I/II Open Label, Multi‐Center Study For The Evaluation Of CPG 7909 In Patients With Stage IB To IVA Cutaneous T‐Cell Lymphoma

Methods

This is a phase I, open‐label, multicentre, dose‐escalation study followed by a randomised phase II study. Participants are followed every 4 weeks.

Projected accrual

A total of 3 to 56 participants (3 to 36 for phase I and 20 [10 per treatment
arm] for phase II) will be accrued for this study.

Participants

Participants aged 18 years and older are eligible for this study.

Both genders are eligible for this study.

Healthy volunteers are not accepted.

Disease characteristics

  • Histologically‐confirmed cutaneous T‐cell lymphoma (CTCL) (limited to mycosis fungoides)

    • Stage IB‐IVA disease

    • No other cutaneous lymphomas including, but not limited to, CD30‐positive large‐cell T‐cell lymphoma, lymphomatoid papulosis, and pagetoid reticulosis

    • No visceral disease (stage IVB CTCL)

  • Must have received 1 to 3 prior systemic regimen(s), including psoralen ultraviolet light therapy (PUVA)

  • No CNS disease

Participant characteristics

  • Aged 18 and over

  • Performance Status: Karnofsky 60% to 100%

  • Life expectancy more than 4 months

  • Haematopoietic:

    • Neutrophil count ≥ 1,000/mm^3

    • Platelet count > 100,000/mm^3

    • WBC > 4,000/mm^3

    • Heamoglobin ≥ 10 g/dL

  • Hepatic:

    • Bilirubin ≤ 1.5 mg/dL

    • SGOT or SGPT < 3 times upper limit of normal

    • PTT ≤ 40 seconds

    • No active hepatitis B or C

  • Renal:

    • Creatinine ≤ 2.0 mg/dL

  • Cardiovascular:

    • No unstable angina

    • No New York Heart Association class III‐IV congestive heart failure

    • No myocardial infarction within the past 6 months

    • No uncontrolled atrial or ventricular cardiac arrhythmias

    • No other significant cardiovascular disease

  • Immunologic:

    • HIV negative

    • No autoimmune or antibody‐mediated disease, including any of the following:

      • Systemic lupus erythematosus

      • Rheumatoid arthritis

      • Multiple sclerosis

      • Sjögren's syndrome

      • Autoimmune thrombocytopenia

    • Controlled thyroid disease allowed

    • Autoantibodies without clinical autoimmune disease allowed

    • No history of allergic reactions attributed to compounds of similar composition to CpG 7909

    • No fever ≥ 38.2°C within the past 24 hours

    • No serious, symptomatic, significant local or systemic infection, including urinary tract infection

  • Other:

    • Not pregnant or nursing

    • Negative pregnancy test

    • Fertile participants must use effective contraception during and for 4 weeks after study participation

    • No suspected or confirmed poor compliance, mental instability, or prior or current alcohol or drug abuse that would preclude study participation or giving informed consent

    • No other medical history, including laboratory results, that would preclude study participation

    • No other malignancy within the past 5 years except basal cell or completely excise non‐invasive squamous cell skin cancer or squamous cell carcinoma in situ of the cervix

Prior concurrent therapy

  • Biologic therapy

    • No concurrent denileukin diftitox

    • No other concurrent immunotherapy, including but not limited to, interleukin‐2, interferon‐alpha (IFN‐α) , or IFN‐gamma

  • Chemotherapy

    • At least 4 weeks since prior systemic chemotherapy for CTCL

    • No concurrent chemotherapy

  • Endocrine therapy

    • No concurrent topical or systemic corticosteroids

  • Radiotherapy

    • At least 4 weeks since prior electron beam therapy for CTCL

    • No concurrent radiotherapy

  • Surgery

    • More than 6 months since prior coronary angioplasty

  • Other

    • At least 2 weeks since prior topical therapy for CTCL

    • At least 3 weeks since prior phototherapy for CTCL

    • At least 4 weeks since prior photopheresis for CTCL

    • At least 4 weeks since other prior systemic therapy for CTCL

    • At least 4 weeks since prior daily systemic cholecalciferol (vitamin D) > 15,000 IU for CTCL

    • At least 4 weeks since prior oral retinoids, including bexarotene for CTCL

    • At least 4 weeks since prior investigational therapy for CTCL

    • No prior treatment for hepatitis B or C

    • More than 2 weeks since prior systemic antibiotics for CTCL

    • Participants receiving systemic antibiotics for CTCL must be on a stable regimen for at least 2 weeks before study entry

    • More than 30 days since prior participation in an investigational drug trial

    • No concurrent chloroquine phosphatase

    • No concurrent anticoagulant therapy except aspirin (≤ 325 mg/day)

    • No concurrent phototherapy

    • No concurrent photopheresis therapy

    • No concurrent bexarotene

    • No concurrent immunosuppressants

    • No other concurrent investigational drugs

Interventions

  • Phase I: Participants receive CpG 7909 subcutaneously (SC) once weekly on weeks 1 to 24 in the absence of disease progression or unacceptable toxicity. Cohorts of 3 to 6 participants receive escalating doses of CpG 7909 until the maximum tolerated dose (MTD) is determined. The MTD is defined as the dose preceding that at which 2 of 3 or 2 of 6 participants experience dose‐limiting toxicity.

  • Phase II: Participants are randomised to receive 1 of 2 doses of CpG 7909, administered as in phase I.

Outcomes

Primary outcomes of the trial

  • Tumor response rate (CCR and PR) as measured by the Composite Assessment of Index Lesion Disease Severity [designated as safety issue: no]

Secondary outcomes of the trial

  • Tumor response as measured by the Physician Global Assessment of Clinical Condition criteria [designated as safety issue: no]

  • Duration of overall response (CCR and PR) [designated as safety issue: no]

  • Duration of CCR [designated as safety issue: no]

  • Duration of PR [designated as safety issue: no]

  • Time‐to‐response [designated as safety issue: no]

  • Time to disease progression [designated as safety issue: no]

Starting date

July 2004

Contact information

Sponsors and collaborators

  • Jonsson Comprehensive Cancer Center

  • National Cancer Institute (NCI)

Investigators

  • Study Chair: Lauren C. Pinter‐Brown, MD, Jonsson Comprehensive Cancer Center

Notes

NCT00168064

Trial name or title

A Phase II Pivotal Trial to Evaluate the Safety and Efficacy of Nitrogen Mustard (NM) 0.02% Ointment Formulations in Patients With Stage I or IIA Mycosis Fungoides (MF)

Methods

This is a phase II, randomised, double‐blind, parallel‐group safety/efficacy study with an estimated enrolment of 250 participants.

Participants

Inclusion criteria of the trial

  • Participants with mycosis fungoides confirmed by a skin biopsy

  • Stage I or IIA participants must have been treated previously with prior topical therapies including PUVA, UVB, topical steroids, but not NM within the past 2 years, or topical carmustine (BCNU)

  • Participants must be otherwise healthy with acceptable organ function

  • Prior to initiating study therapy, participants must not have had topical therapy within 4 weeks

  • Lab values within normal range

  • Willing/able to give consent

  • Must use effective means of contraception if of childbearing potential

Exclusion criteria of the trial

  • Newly‐diagnosed mycosis fungoides with no prior therapy

  • A prior history of treatment with topical NM within the past 2 years or topical carmustine (BCNU)

  • Use of topical or systemic therapies for MF within 4 weeks of entry in the study

  • Participants with a diagnosis of stage IIB‐IV MF

  • Serious known concurrent medical illness or infection, which could potentially present a safety risk, prevent compliance, or both with the requirements of the treatment program

  • Pregnant or nursing women, or men and women of childbearing potential, not using an effective means of contraception

  • Participants who have had radiation therapy within 1 year of study start

  • Participants who have a history of a higher T score than T2 or a higher N score than N1

  • Participants who do not agree to do all labs at 1 site

Interventions

  • Drug: Nitrogen Mustard

"All affected areas (lesions) are to be treated once daily for 12 months with NM 0.02% PG or NM 0.02% AP ointment."

Outcomes

Primary outcomes of the trial

  • Skin response determined by the Composite Assessment of Index Lesion Severity (CAILS) following up to 12 months of treatment [time frame: assessment made at day 1 and every subsequent visit during treatment]

Secondary outcomes of the trial

  • Severity‐weighted assessment tool (SWAT) within up to 12 months by 2 or more consecutive observations over at least 4 weeks [time frame: assessment made at day 1 and every subsequent visit during treatment]

Starting date

May 2006

Contact information

Sponsors and collaborators

Yaupon Therapeutics

Investiogators

Study Director: Stuart Lessin, MD, Fox Chase Cancer Center

Notes

Please see also Lessin 2011 in the 'Studies awaiting classification' section.

NCT01007448

Trial name or title

Phase IV Randomized Study Of Two Dose Levels Of Targretin˜ Capsules In Patients With Refractory Cutaneous T‐Cell Lymphoma

Methods

This is a phase IV, randomised, open‐label, parallel‐group safety/efficacy study with an estimated enrolment of 60 participants.

Participants

Inclusion criteria of the trial

  • A clinical diagnosis of cutaneous T‐cell lymphoma (CTCL) without central nervous system (CNS) involvement, confirmed by biopsy to be histologically consistent with CTCL diagnosis by a dermatopathologist

  • Refractory to at least 1 systemic therapy for CTCL. (Refractory is defined as resistance to therapy due either to lack of response of at least 50% improvement or progression of disease while still on therapy after an initial response)

  • Systemic therapy for CTCL is indicated

  • A Karnofsky performance score ≥ 60%

  • Age ≥18 years

  • Women of childbearing potential must have a negative serum beta human chorionic gonadotropin (ß‐hCG) with a sensitivity of at least 50 mIU/L within 7 days prior to the initiation of treatment. Women of childbearing potential must have used simultaneously 2 highly effective methods of contraception (strongly recommended that 1 of the 2 forms of contraception be non‐hormonal, such as condom plus spermicide, condom plus diaphragm with spermicide, or have a vasectomised partner) or use an intrauterine device or must have been sexually abstinent for at least 4 weeks prior to or at least 1 menstrual cycle prior to (whichever is longer) the negative pregnancy test through entry in the study. Sexual abstinence or effective contraception must be used for at least 1 month prior to the initiation of therapy, during therapy, and for at least 1 month following discontinuation of therapy. Perimenopausal women must be amenorrhoeic for at least 12 months to be considered of non‐childbearing potential

  • Male participants with female partners of childbearing potential must agree to sexual abstinence or to practice 2 reliable forms of effective contraception used simultaneously (strongly recommended that 1 of the 2 forms of contraception be non‐hormonal, such as condom plus spermicide, condom plus diaphragm with spermicide, or partner with tubal ligation) or partner may use an intrauterine device, during the entire period of Targretin capsule treatment and for at least one month after treatment is discontinued. Male participants with female sexual partners who are pregnant, possibly pregnant or who could become pregnant during the study must agree to use condoms during sexual intercourse during the entire period of Targretin capsule treatment and for at least 1 month after the last dose of Targretin capsules

  • Must be willing and able to give informed consent, complete and understand, either oral or written, study procedures and assessments

  • Participant must be suitable for participation in the study in the investigator's opinion

  • Fasting serum triglyceride within normal limits (< 150 mg/dL) prior to study entry

  • Adequate renal function as evidenced by serum creatinine ≤ 2.0 mg/dL or calculated creatinine clearance ≥ 40 mL/min as per the Cockroft and Gault formula

  • Adequate hepatic function that is characterised by aspartate aminotransferase (SGOT [AST]), alanine aminotransferase (SGPT [ALT]), or serum bilirubin < 2.5 times the upper limit of normal

  • Adequate bone marrow function as evidenced by haemoglobin ≥ 8 g/dL, absolute neutrophil count (ANC) ≥ 1,000/mm3, and platelets ≥ 50,000/mm3

Exclusion criteria of the trial

  • Cutaneous T‐cell lymphoma involving the central nervous system

  • Participants with known human immunodeficiency virus (HIV) infection and active hepatitis B virus (HBV) or hepatitis C virus (HCV) infection (HBV/HCV or HIV testing is not required for the purpose of this study)

  • Participation in any other investigational drug study within 30 days of entry in this study

  • Within 5 years after the onset of menopause

  • Received systemic corticosteroids within 6 months of entry in the study

  • Known hypersensitivity to bexarotene or other component of Targretin capsules

  • Pregnancy, intent to become pregnant, or breast‐feeding

  • Received gemfibrozil within 1 day of starting the study

  • Prior therapy for the treatment of CTCL:

    • PUVA or UVB therapy within 3 weeks of study entry

    • EBT or photopheresis within 3 weeks of study entry

    • Topical retinoids, nitrogen mustard, BCNU, imiquimod, etc, within 2 weeks of study entry. If antipruritic medication cannot be avoided, antihistamine or antipruritic agents must be administered using a stable dose regimen for at least 1 week prior to initiation of study drug treatment and throughout the study, unless it is determined that a discontinuation or reduction in dose is indicated. Prior to the enrolment of any participant who will be taking systemic or dermatologically‐applied antihistamine or antipruritic agent, the investigator must contact Eisai to discuss the need for such agents. Mineral oil, baby oil, and simple moisturising lotions may be used as emollients. Low‐ to mid‐potency topical corticosteroids are allowed only for participants with erythroderma (stage III/IV CTCL) using a stable dose regimen for at least 4 weeks prior to study entry. High‐potency topical corticosteroids and tar baths are not permitted. NOTE: Prior to the enrolment of any participant who will be taking systemic or dermatologically‐applied antihistamine or antipruritic agent, the investigator must contact the sponsor to discuss the need for such agents

    • Anticancer therapy of any kind (e.g. methotrexate, cyclophosphamide, vorinostat, romidepsin, interferon, etc) within 30 days of entry to the study. Participant must recover from all signs of toxicity prior to entry in the study

    • Oral retinoid therapy for any indication within 3 months of study entry

    • Systemic therapy with Vitamin A in doses of greater than 15,000 IU (5,000 mcg) per day (equivalent to approximately three times RDA) within 30 days of entry in this study)

  • Systemic antibiotic therapy within 2 weeks of entry in the study. (Participants with infections requiring antibiotics or likely to require antibiotics should be appropriately treated with a course of antibiotics terminating at least 2 weeks prior to entry, or if indicated, a chronic suppressive or prophylactic dose of antibiotics stabilised at least 2 weeks prior to entry. Participants who require initiation of or changes in antibiotic therapy during the study will not be considered a violation of this protocol)

  • History of pancreatitis or significant risk factors for developing pancreatitis (e.g. prior pancreatitis, uncontrolled hyperlipidemia, excessive alcohol consumption, uncontrolled diabetes mellitus, biliary tract disease, and medications known to increase triglyceride levels or to be associated with pancreatic toxicity)

  • Unwillingness or inability to minimise exposure to sunlight and artificial ultraviolet light while receiving Targretin capsules

  • Systemic antibiotic therapy within 2 weeks of entry in the study. (Participants with infections requiring antibiotics or likely to require antibiotics should be appropriately treated with a course of antibiotics terminating at least 2 weeks prior to entry, or if indicated, a chronic suppressive or prophylactic dose of antibiotics stabilised at least 2 weeks prior to entry. Participants who require initiation of or changes in antibiotic therapy during the study will not be considered a violation of this protocol)

  • History of pancreatitis or significant risk factors for developing pancreatitis (e.g. prior pancreatitis, uncontrolled hyperlipidemia, excessive alcohol consumption, uncontrolled diabetes mellitus, biliary tract disease, and medications known to increase triglyceride levels or to be associated with pancreatic toxicity)

  • Unwillingness or inability to minimise exposure to sunlight and artificial ultraviolet light while receiving Targretin capsules

Interventions

  • Arm 1: drug: bexarotene 150 mg/m²/day oral bexarotene capsules, other name: Targretin

  • Arm 2: drug: bexarotene 300 mg/m²/day oral bexarotene capsules, other name: Targretin

Outcomes

Primary outcomes of the trial

  • Tumor responses (clinical complete and partial): Composite Assessment of Index Lesion Disease Severity (CA); Physician's Global Assessment of Clinical Condition (PGA); Percent Body Surface Area Involvement (BSA) [time frame: 24 weeks]

Secondary outcomes of the trial

  • Time to cutaneous tumour response

  • Response duration

  • Time to cutaneous tumour progression [time frame: 24 weeks]

Starting date

December 2009

Contact information

Sponsors and Collaborators

  • Eisai Inc

Investigators

  • Study Director: Chad McQueen, Eisai Inc.

  • Contact: Lorraine Hughes, 410‐631‐8168, [email protected]

Notes

NCT01098656

Trial name or title

A Phase III Study of Lenalidomide Maintenance After Debulking With Gemcitabine or Liposomal Doxorubicin +/‐ Radiotherapy in Patients With Advanced Cutaneous T‐Cell Lymphoma Not Previously Treated With Intravenous Chemotherapy

Methods

This is a randomised, phase III, open‐label trial studying observation to see how well it works compared with lenalidomide in treating participants who are in complete or partial response after receiving previous gemcitabine hydrochloride or doxorubicin hydrochloride liposome for stage IIB, stage III, or stage IV cutaneous T‐cell lymphoma or stage IIB, stage III, or stage IV mycosis fungoides/Sézary syndrome with an estimated enrolment of 105 participants.

Participants

Disease characteristics

  • Diagnoses of advanced T‐cell cutaneous lymphoma or mycosis fungoides/Sézary syndrome

    • Stage IIB‐IV disease

  • Achieved complete or partial response after undergoing prior debulking therapy with 1 of the following recommended* regimens with or without radiotherapy**:

    • Gemcitabine hydrochloride IV over 30 minutes on days 1, 8, and 15 of a 28‐day course at a dose of 1,000 to 1,200 mg/m² for a total of 4 courses

    • Pegylated liposomal doxorubicin hydrochloride IV over 1 hour on days 1 and 15 of a 28‐day course at a dose of 20 mg/m² for a total of 4 courses

NOTE: *These recommended regimens can be altered according to local institutional policies. In case of drug intolerance, the study regimen can be switched from one regimen to the other.

NOTE: **Local low‐dose/energy‐ionizing radiation therapy allowed as part of the debulking process to treat lesions that do not respond after 3 courses of debulking chemotherapy.

  • Sézary cell burden must be decreased by at least 50% after debulking in participants with Sézary syndrome

  • Disease not appropriate for skin‐directed therapy per local institution standards

  • No disease progression between registration and randomisation

  • No CNS involvement

Participant characteristics

  • WHO Performance Status 0 to 2

  • Life expectancy > 12 months

  • Haemoglobin ≥ 10 g/dL

  • Absolute neutrophil count ≥ 1.5 x 10^9/L

  • Platelet count ≥ 60 x 10^9/L

  • Total bilirubin ≤ 1.5 times upper limit of normal (UNL)

  • Alkaline phosphatase ≤ 3 times UNL

  • ALT/AST ≤ 3 times UNL

  • Electrolytes (including sodium, potassium, and chloride) normal

  • Creatinine normal

  • Creatinine clearance ≥ 60 mL/min

  • Uric acid and calcium normal

  • Free T4 and TSH ≤ 1.5 times ULN

  • Participants with a buffer range from the normal values of +/‐ 10% for haematology and biochemistry are acceptable

  • Not pregnant or nursing

  • Negative pregnancy test

  • Fertile participants must use effective contraception 4 weeks prior to, during, and for 4 weeks after completion of study therapy

  • Men must agree not to donate semen during and for 1 week after completion of study therapy

  • Participants with high risk for or history of a thromboembolic event must agree to receive prophylactic anticoagulation therapy (e.g., vitamin K) to keep INR in the range of 2 to 3

  • No New York Heart Association class III‐IV disease

  • No blood donating during and for 1 week after completion of study therapy

  • No uncontrolled infectious disease, autoimmune disease, or immunodeficiency

  • No second malignancies within the past 3 years except surgically‐cured carcinoma in situ of the cervix, in situ breast cancer, incidental finding of stage T1a or T1b prostate cancer, and basal or squamous cell carcinoma of the skin

  • No psychological, familial, sociological, or geographical condition potentially hampering compliance with the study protocol and follow‐up schedule

  • No Lapp lactase deficiency or history of glucose‐galactose malabsorption

Prior concurrent therapy

  • See 'Disease characteristics'

  • No other prior intravenous chemotherapy for this cancer

    • For the purposes of this protocol, the definition of intravenous chemotherapy also includes denileukin diftitox, antibodies, or antibody conjugates

  • No prior splenectomy or splenic irradiation

  • No concurrent topical corticosteroids

    • Concurrent systemic corticosteroids allowed for treatment of tumour flare reactions

  • No radiation or drug‐based therapy (including steroids) between registration and randomisation

  • No other concurrent drugs (including steroids) during the debulking regimen

    • Low‐dose steroids as premedication allowed at the investigator's discretion

  • No other concurrent anticancer treatments

Interventions

  • Arm I: Beginning 4 to 6 weeks after completion of prior debulking therapy, participants undergo observation for 560 days.

  • Arm II: Beginning 4 to 6 weeks after completion of prior debulking therapy, participants receive oral lenalidomide once a day on days 1 to 21. Treatment repeats every 28 days for 20 courses in the absence of disease progression or unacceptable toxicity.

After completion of study treatment, participants are followed at 4 weeks and then every 12 weeks thereafter.

Outcomes

Primary outcomes of the trial

  • Progression‐free survival [designated as safety issue: no]

Secondary outcomes of the trial

  • Overall survival [designated as safety issue: no]

  • Progression‐free survival as assessed by haematogenous disease criteria

  • Acute and late toxicity

  • Conversion rate

  • Rate of occurrence of second cancers at any site

Starting date

July 2010

Contact information

Sponsors and collaborators

  • European Organization for Research and Treatment of Cancer

Investigators

  • Principal Investigator: Martine Bagot, Hopital Saint‐Louis

Notes

NCT01187446

Trial name or title

A Multicenter, Open‐label, Randomized, Phase I/II Study Evaluating the Safety and Efficacy of Low‐dose (12 Gy) Total Skin Electron Beam Therapy (TSEBT) Combined With Vorinostat Versus Low‐dose TSEBT Monotherapy in Mycosis Fungoides (MF)

Methods

This is a multicentre, open‐label, parallel‐group safety/efficacy study with an estimated enrolment of 60 participants.

Participants

Inclusion criteria of the trial

A participant will be eligible for inclusion only if all of the following criteria apply:

  • Biopsy‐confirmed mycosis fungoides, clinical stage IB‐IIIB

  • Participants must have failed or have been intolerant to at least 1 prior systemic or skin‐directed therapy. This may include topical steroids if used as primary therapy for MF

  • 18 years of age or older

  • Eastern Cooperative Oncology Group (ECOG) of < = 2

  • Adequate bone marrow function: WBC > 2000/uL; platelet count > 75,000/mm3; ANC > 1000. Participants cannot be using colony stimulating factors

  • Required wash‐out period for prior therapies

    • Topical therapy: 2 weeks

    • Systemic biologic, monoclonal antibody, or chemotherapy: 4 weeks

    • Phototherapy or radiotherapy (excluding TSEBT): 4 weeks

    • Other investigational therapy: 4 weeks

    • Note: participants with rapidly progressive disease may be treated earlier than required washout period; however, such circumstance must be discussed and approved by the protocol director at the primary site (Stanford)

  • Women of child‐bearing potential (WOCBP) must have negative serum pregnancy test

  • WOCBP must agree to use effective contraception, defined as oral contraceptives, intrauterine devices, or double barrier method (condom plus spermicide or diaphragm), or abstain from sexual intercourse. WOCBP includes any woman who has experienced menarche and who has not undergone successful surgical sterilisation or is not postmenopausal (defined as amenorrhoea for 12 consecutive months)

  • Male subjects must be willing to use an appropriate method of contraception (e.g. condoms) or abstain from sexual intercourse and inform any sexual partners that they must also use a reliable method of contraception (e.g. birth control pills) during the study.

  • Adequate hepatic function: bilirubin <= 1.5 x upper limit of normal (ULN), AST < = 2.5 x UNL, ALT < = 2.5 x UNL, alkaline phosphatase (liver fraction) < = 2.5 x ULN

  • Adequate renal function: creatinine < = 1.5 x UNL OR creatinine clearance < =60 mL/min for participants with creatinine levels > 1.5 X institutional ULN

  • Metabolic parameters: potassium level between 3.5 and 4.5, magnesium level between 1.5 and 2.5

  • Ability to understand and sign a written informed consent document.

  • Ability to comply with the treatment schedule

Exclusion criteria of the trial

A participant will not be eligible for inclusion if any of the following criteria apply:

  • Prior courses of TSEBT (Note: localised skin‐directed radiotherapy is allowed if administered at least 4 weeks prior to initiation on study)

  • Concomitant use of any anticancer therapy or immune modifier

  • Prior allogeneic or autologous transplant

  • Active infection or have received intravenous antibiotics, antiviral, or antifungal agents within 2 weeks prior to the start of the study drug

  • Known history of human immunodeficiency virus (HIV) or hepatitis B or C

  • History of prior malignancy with the exception of cervical intraepithelial neoplasia, non‐melanoma skin cancer, and adequately treated localised prostate carcinoma (PSA < 1.0). Participants with a history of other malignancies must have undergone potentially curative therapy and have no evidence of that disease for 5 years

  • Participant has uncontrolled intercurrent illness, condition, or circumstances that could limit compliance with the study, including, but not limited to the following: active infection, acute or chronic graft versus host disease, symptomatic congestive heart failure, unstable angina pectoris, medically‐significant cardiac arrhythmia, uncontrolled diabetes mellitus or hypertension, or psychiatric conditions

  • Recent (in the past 6 months) medically‐significant cardiac event (i.e. myocardial infarction, cardiac surgery)

  • Congenital long QT syndrome

  • QTc interval > 480 msec on screening ECG

  • Proven or suspected stage IV disease including participants with B2 (Sézary syndrome), N3 (frank LN disease), or M1 (visceral disease) categories; presence of reactive or dermatopathic lymphadenopathy (N1‐2) or limited blood involvement (B1) is permitted

  • ECOG Performance Status > 2

  • Pregnant or lactating

  • Unwilling to use reliable birth control methods

  • Any other medical issue, including laboratory abnormalities, deemed by the Investigator to be likely to interfere with participant participation

  • Unwilling or unable to provide informed consent

Interventions

  • TSEBT 12 Gy (other name: total skin electron beam therapy)

  • Drug: Vorinostat calculated per participant (other name: Zolinza)

Outcomes

Primary outcomes of the trial

  • Clinical response rate, especially complete response (CR) at week 8 as determined by an mSWAT score of 0 [time frame: at week 8 from the first treatment day]

  • Safety and tolerability as measured by severity and frequency of adverse events [time frame: at week 8 from the first treatment day]

Secondary outcomes of the trial

  • To determine duration of response and time to progression as measured by the mSWAT skin assessment [time frame: at week 8 from the first treatment day]

  • To determine pruritus response as measured by pruritus visual analogue scale (VAS) outcome time frame: at week 8 from the first treatment day. (Change of 3 points for more than 4 weeks will be determined to be statistically significant [time frame: at week 8 from the first treatment day]

Starting date

December 2010

Contact information

Sponsors and collaborators

  • Stanford University

Investigators

  • Principal Investigator: Youn H Kim, Stanford University

Contact: Cameron Harrison, (650) 721‐7186, [email protected]

Notes

NCT01386398

Trial name or title

Vorinostat (Zolinza®) in Combination With (Velcade®) Versus Vorinostat Alone in Refractory or Recurrent Advanced CTCL: A Randomized Phase III Study

Methods

This is a randomised, phase III open‐label trial studying how well vorinostat works when given alone compared with vorinostat given together with bortezomib in treating participants with refractory or recurrent stage IIB, stage III, or stage IV cutaneous T‐cell lymphoma with estimated enrolment of 189 participants.

Participants

Disease characteristics

  • Histologically‐confirmed advanced cutaneous T‐cell lymphoma (CTCL), including its variants mycosis fungoides and Sézary syndrome

    • Stage IIB‐IV disease

  • Relapsed or refractory disease, including any of the following:

    • Participants with clinical progression following EORTC‐21081 protocol treatment

    • Intolerant to ≥ 1 prior intravenous chemotherapy, including denileukin diftitox, antibodies or antibody conjugates, or any other systemic therapy

  • No CNS involvement

Participant characteristics

  • WHO Performance Status 0‐2

  • Absolute neutrophil count > 1.5 x 10^9/L*

  • Platelet count > 100 x 10^9/L*

  • Haemoglobin > 9 g/dL*

  • WBC > 3 x 10^9/L*

  • Bilirubin ≤ 1.5 times upper limit of normal (ULN)*

  • AST and ALT ≤ 3 times ULN (in case of liver infiltration ≤ 5 x ULN)*

  • Serum creatinine ≤ 2.0 mg/dL*

  • Calculated creatinine clearance ≥ 60 mL/min

  • Electrolytes (including potassium and magnesium) ≤ 1 times ULN*

  • Not pregnant or nursing prior to the first dose of study treatment and until 4 weeks after the last study treatment

  • Negative pregnancy test

  • Fertile participants must use effective contraception during and for 3 months after completion of study therapy

  • Able to swallow capsules and is able to take or tolerate oral medication on a continuous basis

  • No New York Heart Association class III‐IV disease

  • None of the following known conditions:

    • Infectious disease

    • Autoimmune disease

    • Immunodeficiency

  • No known or active HIV and/or hepatitis A, B, or C infection

  • No NCI CTC grade 1 peripheral sensory neuropathy with pain or peripheral sensory or motor neuropathy ≥ grade II

  • No other malignancy within the past 5 years

  • No psychological, familial, sociological, or geographical condition potentially hampering compliance with the study protocol and follow‐up schedule NOTE: Participants with a buffer range from the normal values of +/‐ 5% for haematology and +/‐ 10% for biochemistry are acceptable, except for renal function

Prior concurrent therapy

  • See Disease characteristics

  • Must have completely recovered from previous treatment toxicity

  • No prior splenectomy or splenic irradiation

  • No prior bortezomib, histone deacetylase inhibitors (including vorinostat [SAHA]), or both

  • More than 4 weeks since prior chemotherapy, immunotherapy, radiotherapy, or surgery

    • In case of clear progression during previous treatment, 2 weeks of wash‐out is enough

  • No concurrent chemotherapy, immunotherapy, radiotherapy, or surgery (except biopsies)

  • No concurrent steroid (prednisone or equivalent) dose > 20 mg/day

    • Prednisone ≤ 20 mg/day for treatment of disorders other than CTCL allowed

  • No concomitant use of other histone deacetylase inhibitors (e.g. valproic acid)

Interventions

  • Arm I: Participants receive oral vorinostat (SAHA) once daily in the absence of disease progression or unacceptable toxicity.

  • Arm II: Participants receive bortezomib IV on days 1, 4, 8, and 11 and oral vorinostat once daily on days 1 to 14. Treatment repeats every 21 days until progression or unacceptable toxicity.

Blood and tissue samples are collected periodically for translational research to provide insight into disease mechanism and identify biomarkers useful for prediction of treatment response.

After completion of study treatment, participants are followed up at 4 weeks and then every 3 months until disease progression.

Outcomes

Primary outcomes of the trial

  • Progression‐free survival [designated as safety issue: no]

Secondary outcomes of the trial

  • Overall survival

  • Response rate

  • Time to progression

  • Duration of response

  • Second cancers

  • Acute and late toxicity

Starting date

January 2012

Contact information

Sponsors and collaborators

  • European Organization for Research and Treatment of Cancer

Investigators

  • Investigator: Pablo Luis Ortiz‐Romero, Hospital Universitario

Notes

NCT01433731

Trial name or title

A Randomized, Double‐Blind, Placebo‐Controlled, Dose‐Escalating Phase 1b Study to Assess the Safety, Pharmacodynamics and Pharmacokinetics of SHP 141, A Histone Deacetylase Inhibitor, Administered Topically Up to 28 Days to Patients With Stage IA, IB or IIA Cutaneous T‐Cell Lymphoma

Methods

This is a phase I, randomised, placebo‐controlled, double‐blind, parallel‐group safety/efficacy study with an estimated enrolment of 48 participants.

Participants

Inclusion criteria of the trial

  • Histopathologically‐confirmed CTCL; a documented verifiable biopsy report is required

  • Documented clinical Stage IA, IB, or IIA CTCL

  • Skin lesion involvement of at least 3% of BSA accessible for topical application of study drug and biopsy

  • ECOG Performance Status of 0 to 2

Exclusion criteria of the trial

  • CTCL with histologic evidence of folliculotropic variant or large cell transformed CTCL

  • Severe pruritus requiring systemic or topical treatment

  • Palpable lymph node ≥1.5 cm in diameter (unless the lymph node has been biopsied and has been designated as Stage IA‐IIA disease)

  • Coexistent second malignancy or history of prior solid organ malignancy within previous 5 years (excluding basal or squamous cell carcinoma of the skin, in situ carcinoma of the cervix (CIN 3), papillary or follicular thyroid cancer that has been treated curatively, or prostate cancer that has been treated curatively)

  • Any prior history of a haematologic malignancy (other than CTCL)

  • History of or current major renal, hepatic, gastrointestinal, pulmonary, cardiovascular, genito‐urinary or haematological disease, CNS disorders, infectious disease or coagulation disorders as determined by the Investigator.

  • Evidence of active hepatitis B or C or HIV

  • Circulating atypical cells > 5%

Interventions

  • Arm 1: drug: SHP‐141 topical gel

  • Arm 2: placebo SHP‐141 topical gel

Outcomes

Primary outcomes of the trial

  • Lesion severity using CAILS (Composite Assessment of Index Lesion Severity) [time frame: weekly through day 28 (days 1, 7, 14, 21, 28) and again day 42]

Starting date

September 2011

Contact information

Sponsors and Collaborators

  • Shape Pharmaceuticals, Inc

  • The Leukemia and Lymphoma Society

  • Therapeutics, Inc

  • Veristat, Inc

  • PPD

Investigators

  • Principal Investigator: Joan Guitart, MD Northwestern University

Notes

Data and analyses

Open in table viewer
Comparison 1. Topical peldesine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.1

Comparison 1 Topical peldesine versus placebo, Outcome 1 Adverse effects.

Comparison 1 Topical peldesine versus placebo, Outcome 1 Adverse effects.

1.1 Pruritus

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Topical peldesine versus placebo, Outcome 2 Clearance.

Comparison 1 Topical peldesine versus placebo, Outcome 2 Clearance.

3 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Topical peldesine versus placebo, Outcome 3 Improvement.

Comparison 1 Topical peldesine versus placebo, Outcome 3 Improvement.

Open in table viewer
Comparison 2. Topical imiquimod versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.1

Comparison 2 Topical imiquimod versus placebo, Outcome 1 Clearance.

Comparison 2 Topical imiquimod versus placebo, Outcome 1 Clearance.

2 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.2

Comparison 2 Topical imiquimod versus placebo, Outcome 2 Improvement.

Comparison 2 Topical imiquimod versus placebo, Outcome 2 Improvement.

Open in table viewer
Comparison 3. Topical hypericin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.1

Comparison 3 Topical hypericin versus placebo, Outcome 1 Improvement.

Comparison 3 Topical hypericin versus placebo, Outcome 1 Improvement.

Open in table viewer
Comparison 4. IFN‐α versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 4.1

Comparison 4 IFN‐α versus placebo, Outcome 1 Adverse effects.

Comparison 4 IFN‐α versus placebo, Outcome 1 Adverse effects.

1.1 Erythema

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Fever

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Myalgia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Chills or weakness

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Nausea, arthralgia, and malaise

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.2

Comparison 4 IFN‐α versus placebo, Outcome 2 Clearance.

Comparison 4 IFN‐α versus placebo, Outcome 2 Clearance.

Open in table viewer
Comparison 5. IFN‐α + PUVA versus PUVA alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clearance Show forest plot

2

122

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.87, 1.31]

Analysis 5.1

Comparison 5 IFN‐α + PUVA versus PUVA alone, Outcome 1 Clearance.

Comparison 5 IFN‐α + PUVA versus PUVA alone, Outcome 1 Clearance.

Open in table viewer
Comparison 6. Denileukin diftitox high versus low dose

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.1

Comparison 6 Denileukin diftitox high versus low dose, Outcome 1 Adverse effects.

Comparison 6 Denileukin diftitox high versus low dose, Outcome 1 Adverse effects.

1.1 Constitutional symptoms

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Grade 3 to 4 constitutional symptoms

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Infections

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Grade 3 to 4 infections

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Gastrointestinal syndromes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.6 Grade 3 to 4 gastrointestinal syndromes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.7 CNS syndromes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.8 Grade 3 to 4 CNS syndromes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.9 Rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.10 Grade 3 to 4 rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.11 Vascular leak syndrome

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.12 Grade 3 to 4 vascular leak syndrome

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.13 Thrombotic events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.14 Grade 3 to 4 thrombotic events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.15 Cardiopulmonary events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.16 Grade 3 to 4 cardiopulmonary events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.17 Acute infusion related events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.18 Grade 3 to 4 acute infusion related events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.19 Grade 3 to 4 laboratory abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 6.2

Comparison 6 Denileukin diftitox high versus low dose, Outcome 2 Clearance.

Comparison 6 Denileukin diftitox high versus low dose, Outcome 2 Clearance.

3 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 6.3

Comparison 6 Denileukin diftitox high versus low dose, Outcome 3 Improvement.

Comparison 6 Denileukin diftitox high versus low dose, Outcome 3 Improvement.

Open in table viewer
Comparison 7. Bexarotene high versus low dose

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.1

Comparison 7 Bexarotene high versus low dose, Outcome 1 Adverse effects.

Comparison 7 Bexarotene high versus low dose, Outcome 1 Adverse effects.

1.1 Photosensitivity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Constipation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Fatigue

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Pruritus

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.6 Arthralgia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.7 Nasopharyngitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.8 Headache

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.9 Insomnia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.10 Free T4 abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.11 Cholesterol abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.12 Triglycerid abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.13 SGOT/SGPT abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 7.2

Comparison 7 Bexarotene high versus low dose, Outcome 2 Clearance.

Comparison 7 Bexarotene high versus low dose, Outcome 2 Clearance.

3 Relapse Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 7.3

Comparison 7 Bexarotene high versus low dose, Outcome 3 Relapse.

Comparison 7 Bexarotene high versus low dose, Outcome 3 Relapse.

4 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 7.4

Comparison 7 Bexarotene high versus low dose, Outcome 4 Improvement.

Comparison 7 Bexarotene high versus low dose, Outcome 4 Improvement.

Open in table viewer
Comparison 8. Extracorporal photopheresis versus PUVA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 8.1

Comparison 8 Extracorporal photopheresis versus PUVA, Outcome 1 Clearance.

Comparison 8 Extracorporal photopheresis versus PUVA, Outcome 1 Clearance.

2 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 8.2

Comparison 8 Extracorporal photopheresis versus PUVA, Outcome 2 Improvement.

Comparison 8 Extracorporal photopheresis versus PUVA, Outcome 2 Improvement.

Open in table viewer
Comparison 9. Combined therapy versus conservative therapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 9.1

Comparison 9 Combined therapy versus conservative therapy, Outcome 1 Adverse effects.

Comparison 9 Combined therapy versus conservative therapy, Outcome 1 Adverse effects.

1.1 Hospitalization

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Fatal myocardial infarction

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Cutaneous toxicity from electron beam therapy

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Acute non‐lymphocytic leukaemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Non‐melanoma skin cancer

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.6 Unspecified

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 9.2

Comparison 9 Combined therapy versus conservative therapy, Outcome 2 Clearance.

Comparison 9 Combined therapy versus conservative therapy, Outcome 2 Clearance.

3 Relapse Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 9.3

Comparison 9 Combined therapy versus conservative therapy, Outcome 3 Relapse.

Comparison 9 Combined therapy versus conservative therapy, Outcome 3 Relapse.

4 Survival rate Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 9.4

Comparison 9 Combined therapy versus conservative therapy, Outcome 4 Survival rate.

Comparison 9 Combined therapy versus conservative therapy, Outcome 4 Survival rate.

5 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 9.5

Comparison 9 Combined therapy versus conservative therapy, Outcome 5 Improvement.

Comparison 9 Combined therapy versus conservative therapy, Outcome 5 Improvement.

Open in table viewer
Comparison 10. IFN‐α + PUVA versus IFN‐α + acitretin

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 10.1

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 1 Adverse effects.

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 1 Adverse effects.

1.1 Grade I to II adverse events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Grade III adverse events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Adverse events requiring treatment discontinuation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Flu‐like symptoms

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Dryness/redness of skin or hair loss

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.6 Neurological disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.7 Psychiatric disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.8 Gastrointestinal disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.9 Elevated liver or biliary tract enzymes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.10 Elevated triglycerides

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.11 Anemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.12 Leukopenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.13 Impotentia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.14 Redness and infiltration at application site

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 10.2

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 2 Clearance.

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 2 Clearance.

3 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 10.3

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 3 Improvement.

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 3 Improvement.

Open in table viewer
Comparison 11. Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 11.1

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 1 Clearance.

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 1 Clearance.

2 Survival rate Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 11.2

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 2 Survival rate.

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 2 Survival rate.

3 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 11.3

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 3 Improvement.

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 3 Improvement.

Study flow chart
Figuras y tablas -
Figure 1

Study flow chart

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

'Risk of bias' summary: review authors' judgements about each 'Risk of bias' item for each included study.
Figuras y tablas -
Figure 3

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

Forest plot of comparison: 5 IFN‐α + PUVA versus PUVA alone, outcome: 5.1 Clearance.
Figuras y tablas -
Figure 4

Forest plot of comparison: 5 IFN‐α + PUVA versus PUVA alone, outcome: 5.1 Clearance.

Comparison 1 Topical peldesine versus placebo, Outcome 1 Adverse effects.
Figuras y tablas -
Analysis 1.1

Comparison 1 Topical peldesine versus placebo, Outcome 1 Adverse effects.

Comparison 1 Topical peldesine versus placebo, Outcome 2 Clearance.
Figuras y tablas -
Analysis 1.2

Comparison 1 Topical peldesine versus placebo, Outcome 2 Clearance.

Comparison 1 Topical peldesine versus placebo, Outcome 3 Improvement.
Figuras y tablas -
Analysis 1.3

Comparison 1 Topical peldesine versus placebo, Outcome 3 Improvement.

Comparison 2 Topical imiquimod versus placebo, Outcome 1 Clearance.
Figuras y tablas -
Analysis 2.1

Comparison 2 Topical imiquimod versus placebo, Outcome 1 Clearance.

Comparison 2 Topical imiquimod versus placebo, Outcome 2 Improvement.
Figuras y tablas -
Analysis 2.2

Comparison 2 Topical imiquimod versus placebo, Outcome 2 Improvement.

Comparison 3 Topical hypericin versus placebo, Outcome 1 Improvement.
Figuras y tablas -
Analysis 3.1

Comparison 3 Topical hypericin versus placebo, Outcome 1 Improvement.

Comparison 4 IFN‐α versus placebo, Outcome 1 Adverse effects.
Figuras y tablas -
Analysis 4.1

Comparison 4 IFN‐α versus placebo, Outcome 1 Adverse effects.

Comparison 4 IFN‐α versus placebo, Outcome 2 Clearance.
Figuras y tablas -
Analysis 4.2

Comparison 4 IFN‐α versus placebo, Outcome 2 Clearance.

Comparison 5 IFN‐α + PUVA versus PUVA alone, Outcome 1 Clearance.
Figuras y tablas -
Analysis 5.1

Comparison 5 IFN‐α + PUVA versus PUVA alone, Outcome 1 Clearance.

Comparison 6 Denileukin diftitox high versus low dose, Outcome 1 Adverse effects.
Figuras y tablas -
Analysis 6.1

Comparison 6 Denileukin diftitox high versus low dose, Outcome 1 Adverse effects.

Comparison 6 Denileukin diftitox high versus low dose, Outcome 2 Clearance.
Figuras y tablas -
Analysis 6.2

Comparison 6 Denileukin diftitox high versus low dose, Outcome 2 Clearance.

Comparison 6 Denileukin diftitox high versus low dose, Outcome 3 Improvement.
Figuras y tablas -
Analysis 6.3

Comparison 6 Denileukin diftitox high versus low dose, Outcome 3 Improvement.

Comparison 7 Bexarotene high versus low dose, Outcome 1 Adverse effects.
Figuras y tablas -
Analysis 7.1

Comparison 7 Bexarotene high versus low dose, Outcome 1 Adverse effects.

Comparison 7 Bexarotene high versus low dose, Outcome 2 Clearance.
Figuras y tablas -
Analysis 7.2

Comparison 7 Bexarotene high versus low dose, Outcome 2 Clearance.

Comparison 7 Bexarotene high versus low dose, Outcome 3 Relapse.
Figuras y tablas -
Analysis 7.3

Comparison 7 Bexarotene high versus low dose, Outcome 3 Relapse.

Comparison 7 Bexarotene high versus low dose, Outcome 4 Improvement.
Figuras y tablas -
Analysis 7.4

Comparison 7 Bexarotene high versus low dose, Outcome 4 Improvement.

Comparison 8 Extracorporal photopheresis versus PUVA, Outcome 1 Clearance.
Figuras y tablas -
Analysis 8.1

Comparison 8 Extracorporal photopheresis versus PUVA, Outcome 1 Clearance.

Comparison 8 Extracorporal photopheresis versus PUVA, Outcome 2 Improvement.
Figuras y tablas -
Analysis 8.2

Comparison 8 Extracorporal photopheresis versus PUVA, Outcome 2 Improvement.

Comparison 9 Combined therapy versus conservative therapy, Outcome 1 Adverse effects.
Figuras y tablas -
Analysis 9.1

Comparison 9 Combined therapy versus conservative therapy, Outcome 1 Adverse effects.

Comparison 9 Combined therapy versus conservative therapy, Outcome 2 Clearance.
Figuras y tablas -
Analysis 9.2

Comparison 9 Combined therapy versus conservative therapy, Outcome 2 Clearance.

Comparison 9 Combined therapy versus conservative therapy, Outcome 3 Relapse.
Figuras y tablas -
Analysis 9.3

Comparison 9 Combined therapy versus conservative therapy, Outcome 3 Relapse.

Comparison 9 Combined therapy versus conservative therapy, Outcome 4 Survival rate.
Figuras y tablas -
Analysis 9.4

Comparison 9 Combined therapy versus conservative therapy, Outcome 4 Survival rate.

Comparison 9 Combined therapy versus conservative therapy, Outcome 5 Improvement.
Figuras y tablas -
Analysis 9.5

Comparison 9 Combined therapy versus conservative therapy, Outcome 5 Improvement.

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 1 Adverse effects.
Figuras y tablas -
Analysis 10.1

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 1 Adverse effects.

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 2 Clearance.
Figuras y tablas -
Analysis 10.2

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 2 Clearance.

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 3 Improvement.
Figuras y tablas -
Analysis 10.3

Comparison 10 IFN‐α + PUVA versus IFN‐α + acitretin, Outcome 3 Improvement.

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 1 Clearance.
Figuras y tablas -
Analysis 11.1

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 1 Clearance.

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 2 Survival rate.
Figuras y tablas -
Analysis 11.2

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 2 Survival rate.

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 3 Improvement.
Figuras y tablas -
Analysis 11.3

Comparison 11 Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor, Outcome 3 Improvement.

Table 1. Clinical staging system

2007

MF and Sézary syndrome

1979

CTCL

Disease‐specific survival rates in % (Agar 2010)

5 year

10 year

IA

T1

IA

T1

98

95

N0

N0

M0

M0

B0‐1

IB

T2

IB

T2

89

77

N0

N0

M0

M0

0‐1

IIA

T1‐2

IIA

T1‐2

89

67

N1‐2

N1

M0

M0

B0‐1

IIB

T3

IIB

T3

56

42

N0‐2

N0,1

M0

M0

B0‐1

III

T4

III

T4

N0‐2

N0,1

M0

M0

B0‐1

IIIA

T4

54

45

N0‐2

M0

B0

IIIB

T4

48

45

N0‐2

M0

B1

IVA1

T1‐4

IVA

T1‐4

41

20

N0‐2

M0

N2‐3

B2

IVA2

T1‐4

M0

23

20

N3

M0

B0‐2

IVB

T1‐4

IVB

T1‐4

18

not reached

N0‐3

N0‐3

M1

M1

B0‐2

Figuras y tablas -
Table 1. Clinical staging system
Table 2. Glossary of terms

General medical terms

Explanation

  • Plaques

A solid elevated area on the skin that is more broad than it is high

  • Neoplasm

Any new and abnormal growth

  • Primary cutaneous lymphoma

Cutaneous T‐ and B‐cell lymphoma that primarily affect the skin

  • Cutaneous T‐cell lymphoma

Group of skin‐directed T‐cell neoplasms with diverse clinical and histological features and prognosis

  • Cutaneous B‐cell lymphoma

Group of skin‐directed B‐cell neoplasms with diverse clinical and histological features and prognosis

  • NK‐cell lymphoma

Group of neoplasms derived from the natural killer cells (NK‐cells) with diverse clinical and histological features and prognosis

  • Precursor haematologic neoplasm

Clinically‐aggressive neoplasm with a high incidence of cutaneous involvement and risk of leukaemic dissemination

  • Lesional skin atrophy

Death of the cells in the damaged area of skin

  • T‐cell

A type of lymphocyte (white cell)

Adverse effects

  • Acute myocardial infarction

Death of myocardial tissue due to blocked blood supply

  • Acute non‐lymphatic leukaemia

Serious disease with high production of non‐lymphatic leucocytes

  • ALT

The alanine transaminase is a liver enzyme (SGPT)

  • Anaemia

Low count of red blood cells

  • Anaemia hypochromic

Low count of red blood cells with low amount of haemoglobin, the red molecule that transports oxygen within the blood vessels

  • Anaphylactoid reactions

Very acute generalsied reaction with flush/urticaria, bad breathing, lowered blood pressure, nausea

  • Arthralgia

Joint pain

  • AST

The aspartate transaminase is an enzyme mainly present in the liver but also in the blood, muscle cells, and bones (SGOT)

  • Asthenia

Lack of energy or physical weakness or both

  • Cardiomyopathy

Structural or functional disease of the cardiac muscle

  • Cardiopulmonary syndrome

Adverse effect where the heart and the lung are involved

  • Chill

Shivering attack

  • CNS syndrome

Adverse effect where the central nervous system (brain, spinal cord) is involved

  • Cutaneous hypersensitivity

Altered reactivity to a specific antigen leading to cutaneous alterations

  • Cutaneous toxicity

Cutaneous adverse effect of an agent used in therapeutic dosages

  • Constipation

No stool

  • Dermatitis exfoliation

Inflammation and detachment of the skin

  • Diarrhoea

Many fluid stools

  • Dyspnea

Bad breathing

  • Erythema

Red dot or area on the skin

  • Fatigue

To be exhausted

  • Flu‐like symptoms

Symptoms which are often seen with influenza, such as fever, chills, and muscular pain

  • Gastrointestinal syndromes

Adverse effects affecting the digestive system (oesophagus, stomach, bowel)

  • Hair loss

Pathological increased loss of hair

  • Hepatotoxicity

Capacity of a substance to have damaging effects on the liver

  • Hospitalisation

Confinement of a patient in a hospital

  • Hypercholestaeremia

Elevated levels of cholesterol in the blood

  • Hyperlipaemia

Elevated levels of lipids in the blood

  • Hypotension

Low blood pressure

  • Hypothyroidism

Low function of thyroid gland

  • Impotentia

Inability to engage in sexual intercourse

  • Insomnia

Not being able to sleep

  • LDH

The lactate dehydrogenase is an enzyme which helps to produce energy in the body when oxygen is absent

  • Leukopenia

Low count of white blood cells

  • Malaise

To feel ill

  • Mucositis

Inflammation of mucosa

  • Myalgia

Muscle pain

  • Nasopharyngitis

Inflammation of the inner nose and the throat

  • nausea

About to vomit while feeling sick

  • Neuropathy

Abnormal state of the nervous system or nerves

  • Non‐melanoma skin cancer

Skin cancer which does not originate from melanocytes

  • Photosensitivity

Enhanced responsibility to light or ultraviolet light

  • Pruritus

Itching of the skin

  • Radiodermatitis

Dermatitis resulting from overexposure to sources of radiant energy

  • Rash

A lot of red dots on the skin

  • SGOT/SGPT

Liver enzymes, see also AST and ALT

  • T4

Enzyme of the thyroid gland

  • Thrombopenia

Low count of thrombocytes

  • Thrombotic syndrome

Blood coagulates within blood vessels

  • Triglycerid

Lipid

  • Vascular leak syndrome

When the blood vessels are not tight anymore and parts of the blood leak out in the surrounding tissue

  • Vasodilatation

Blood vessels widening

Figuras y tablas -
Table 2. Glossary of terms
Table 3. List of abbreviations and acronyms

Acronym

Description (letters used for acronym in capitals)

ADF

Arbeitsgemeinschaft Dermatologische Forschung

BCNU

Carmustine, a nitrogen mustard related alkylating agent

CENTRAL

Cochrane Central Register of Controlled Trials

CI

Confidence interval

CTCL

Cutaneous T‐Cell Lymphoma

DDG

German Dermatologic Society

EORTC

European Organization of Research and Treatment of Cancer

ISCL

International Society for Cutaneous Lymphoma (ISCL)

ITT

Intention‐to‐treat

LILACS

Latin American and Caribbean Health Science Information database

MF

Mycosis Fungoides

PICOS

Participants, Interventions, Controls, Outcomes and Study

PRISMA

Preferred Reporting Items of Systematic Reviews and Meta‐Analyses

RCT

Randomised‐Controlled Trial

RR

Risk Ratio

TSEB

Total Skin Electron Beam

TNMB

Tumour, lymph Node, Metastasis and Blood

UK

United Kingdom

US

Unites States of America

USCLC

United States Cutaneous Lymphoma Consortium

WHO

World Health Organization

Figuras y tablas -
Table 3. List of abbreviations and acronyms
Table 4. TNMB classifications

2007

MF and Sézary syndrome

1979

CTCL

T: Skin

T0

N.E.

Clinically and/or histopathologically suspicious lesions

T1

Limited patches, papules, and/or plaques covering < 10% of the skin surface. May further stratify into T1a (patch only) versus T1b (plaque ± patch)

Limited plaques, papules, or eczematous patches
covering < 10% of the skin surface

T2

Patches, papules, or plaques covering ≥ 10% of the skin surface. May further stratify into T2a (patch only) versus T2b (plaque ± patch)

Generalised plaques, papules, or erythematous
patches covering ≥ 10% of the skin surface

T3

1 or more tumours ( ≥ 1 cm diameter)

Tumors, 1 or more

T4

Confluence of erythema covering ≥ 80% body surface area

Generalised erythroderma

N: Node

N0

No clinically abnormal peripheral lymph nodes; biopsy not required

No clinically abnormal peripheral lymph nodes palpable,
histopathology negative for CTCL

N1

Clinically abnormal peripheral lymph nodes,
histopathology Dutch grade 1 or NCI LN0‐2

Palpable Clinically abnormal peripheral lymph nodes, histopathology
negative for CTCL

N1a

Clone negative

N1b

Clone positive

N2

Clinically abnormal peripheral lymph nodes,
histopathology Dutch grade 2 or NCI LN3

No clinically abnormal peripheral lymph nodes,
histopathology positive for CTCL

N2a

Clone negative

N2b

Clone positive

N3

Clinically abnormal peripheral lymph nodes,
histopathology Dutch grades 3 to 4 or NCI LN4, clone positive or negative

Palpable clinically abnormal peripheral lymph nodes, pathology
positive for CTCL

N3x

Clinically abnormal peripheral lymph nodes, no histologic confirmation

M: Visceral

M0

No visceral organ involvement

No visceral organ involvement

M1

Visceral involvement (must have pathology confirmation and organ involved should be specified)

Visceral involvement (must have pathology
confirmation and organ involved should be
specified)

B: Blood

B0

Absence of significant blood involvement: Less than 5% of peripheral blood lymphocytes are atypical (Sézary) cells

Atypical circulating cells not present (less than 5%)

B0a

Clone negative

B0b

Clone positive

B1

Low blood tumour burden: More than 5% of peripheral blood lymphocytes are atypical (Sézary) cells but do not meet the criteria of B2

Atypical circulating cells present (more than 5%), record total
white blood count and total lymphocyte counts, and
number of atypical cells/100 lymphocytes

B1a

Clone negative

B1b

Clone positive

B2

High blood tumour burden: ≥ 1000/ µL Sézary cells with positive clone

For the 'N' category histopathologic grading is necessary with the new 2007 TNMB classification. It may be either done via the Dutch System or the NCI/VA classification system (Sausville 1988; Scheffer 1980).

Figuras y tablas -
Table 4. TNMB classifications
Table 5. Summary of effects of interventions

Study

Participants randomised

First intervention

Second intervention

Reported outcomes

baseline risk

Significant differences in outcome

Duvic 2001a

89 participants stage I

(IA: 45, IB:44)

topical peldesine 1%

given for 24 weeks

placebo cream

given for 24 weeks

common adverse effects

clearance

survival rates

improvement

common adverse effects (rash): 3% with placebo cream

common adverse effects (rash): The relative risk of 7.24 (95% CI 0.92 to 56.76)

showed a higher risk for peldesine (Fisher test P≤0.041).

Chong 2004

4 participants plaque stage IB

topical imiquimod 5%

given for 16 weeks

placebo cream

given for 16 weeks

common adverse effects

clearance (assessed 16 weeks after end of intervention)

improvement (assessed 16 weeks after end of intervention)

rare adverse effects

none

Rook 2010

12 participants patch or plaque stage, lesional comparison

hypericin 0.05‐0.25% in combination with visible light given for 6 weeks

placebo cream in combination with visible light

given for 6 weeks

common adverse effects

survival rates

improvement

improvement: 8% with placebo cream

improvement: The relative risk of 7.00 (95% CI 1.01 to 48.54) favoured hypericin (Fisher test P≤0.028).

Vonderheid 1987

6 participants stage IA to IIA

(IA: 1; IB: 1; IIA: 4), lesional comparison

injections of interferon‐α 2b

given for 4 weeks

injections of isotonic sterile water given for 4 weeks

common adverse effects (assessed after 3 weeks of intervention)

clearance (assessed 4 weeks after end of intervention)

common adverse effects (mild erythema): 0% with injections of sterile water

clearance: 8% injections of sterile water

common adverse effects (mild erythema):

The relative risk of 11.00 (95% CI 0.74 to 163.49) favoured interferon‐α (Fisher test P≤0.016).

clearance: The RR 10.00 (95% CI 1.51 to 66.43) favoured interferon‐α (Fisher test P≤0.0007).

Wolff 1985

12 participants stage IA (7) to IB (5)

injections of interferon‐α

given for 4 weeks

injections of buffered glycine serum human albumin

given for 4 weeks

common adverse effects

clearance

common adverse effects (mild fever): 0% with injections of buffered glycine serum human albumin

common adverse effects (mild fever): The relative risk of 11.00 (95% CI 0.70‐173.66) showed a higher risk for interferon‐α (Fisher test P≤0.03).

Stadler 2006

124 participants stage lA to IIA

injections of interferon‐α in combination with PUVA

given for up to 52 weeks

PUVA alone given for up to 52 weeks

clearance

none

Wozniak 2008

29 participants stage lA to IIA (IA: 14; IB: 6; IIA: 9)

injections of interferon‐α in combination with PUVA given for 24 weeks

PUVA alone given for 24 weeks

clearance

relapse

disease‐free interval

none

Olsen 2001

71 participants stage IB‐IVA

(IB:16, IIA: 10, IIB: 19, III: 11, IVA:15)

denileukin diftitox intravenous infusion 9µg/kg/day given for up to 6 months

denileukin diftitox intravenous infusion 18µg/kg/day given for up to 6 months

quality of life

common adverse effects

clearance

survival rates (assessed 90 days after end of intervention)

improvement

rare adverse effects

none

Duvic 2001

58 participants stage I to IIA

(IA: 17, IB: 34, IIA: 6, IIB: 1)

bexarotene capsules dosed 300 to 650mg/m²/day given for 16 weeks

bexarotene capsules dosed 6.5mg/m²/day given for 16 weeks

quality of life

common adverse effects

clearance

relapse

survival rates (assessed 4 weeks after end of intervention)

improvement

rare adverse effects

no calculations, due to methodological problems

Guitart 2002

43 participants stage IB (36) and IIA (7)

bexarotene 300mg/day in combination with PUVA and fenofibrate 54mg/day given for 24 weeks

bexarotene 150mg/day in combination with PUVA and fenofibrate 54mg/day given for 24 weeks

common adverse effects

clearance

relapse (assessed 6 months after end of intervention)

disease‐free interval

survival rates

improvement

common adverse effects (cholesterol levels): 15% with bexarotene 150mg/day

common adverse effects (triglyceride levels):25% with bexarotene 150mg/day

common adverse effects (cholesterol levels): The relative risk of 4.56 (5% CI 1.54 to 13.53) showed a higher risk for bexarotene 300mg/day (Fisher test P≤0.002).

common adverse effects (triglyceride levels): The RR 3.16 (95% CI 1.93 to 6.98) showed a higher risk for bexarotene 300mg/day (Fisher test P≤0.002).

Child 2004

8 participants plaque stage (Bunn Lamberg 1B)

extracorporeal photopheresis given for 6 months

PUVA

given for 3 months

common adverse effects

clearance

survival rates (assessed 2‐21 months after end of intervention)

improvement

improvement:0% with extracorporeal photopheresis

improvement: The relative risk of 0.07 (95% CI 0.00 to 1.00) favoured PUVA (Fisher test P≤0.002).

Kaye 1989

103 participants of all stages

(IA: 6, IB: 16, IIA: 9, IIB: 12; III: 2, IVA: 42, IVB: 16)

"combined therapy" (electron‐beam radiation and parenteral chemotherapy) given for 8 to 12 weeks

"conservative treatment"

(topical treatment with mechlorethamine supported by a stepwise escalation of the therapy according to stage of disease) duration of intervention not reported

common adverse effects

clearance

relapse

disease‐free interval

survival rates (assessed more than 5 years after end of intervention)

improvement

common adverse effects

(hospitalisation): 0% with "conservative treatment"

clearance: 18% with "conservative treatment"

improvement: 65% with "conservative treatment"

common adverse effects

(hospitalizations): The relative risk of 33.65 (95% CI 2.07 to 546.09) showed a higher risk for the "combined therapy" (Fisher test P≤0.0001).

clearance: The relative risk of 2.18 (95% CI 1.10 to 4.33) favoured the "combined therapy" (Fisher test P≤0.03).

improvement: The relative risk of 1.40 (95% CI 1.12 to 1.74) favoured the "combined therapy" (Fisher test P≤0.003).

Stadler 1998

82 participants stage I and II

(IA: 36; IB: 28; IIA: 10; IIB: 8)

injections of interferon‐α in combination with PUVA given for up to 48 weeks

injections of interferon‐α in combination with acitretin given for up to 48 weeks

common adverse effects

clearance

improvement

common adverse effects

(adverse effects grade III): 10% IFN‐α in combination with PUVA

common adverse effects (requiring discontinuation): 5% IFN‐α in combination with PUVA

common adverse effects (neurological disorders): 8% IFN‐α in combination with PUVA

clearance: 38% with IFN‐α in combination with acitretin

common adverse effects

(adverse effects grade III): The relative risk of 3.10 (95% CI 1.10 to 8.70) showed a higher risk for interferon‐α in combination with acitretin (Fisher test P≤0.03).

common adverse effects (requiring discontinuation): The relative risk of 4.29 (95% CI 0.99 to 18.63) showed a higher risk for interferon‐α in combination with acitretin (Fisher test P≤0.049).

common adverse effects (neurological disorders): The relative risk of 3.49 (95% CI 1.05 to 11.60) showed a higher risk for interferon‐α in combination with acitretin (Fisher test P≤0.04).

clearance: The relative risk of 0.54 (95% CI 0.35 to 0.84) favoured interferon‐α in combination with PUVA (Fisher test P≤0.005).

Thestrup‐Pedersen 1982

16 participants van Scott stage II‐IV

(II: 14; III: 1; IV: 1)

topically applied nitrogen mustard with active transfer factor given for 1 year

topically applied nitrogen mustard with inactivated transfer factor given for 1 year

common adverse effects

clearance (assessed 1 year after end of intervention)

survival rates (assessed 1 year after end of intervention)

improvement (assessed 1 year after end of intervention)

clearance: 0% with topically applied nitrogen mustard with active transfer factor

clearance: The relative risk of 0.09 (95% CI 0.01 to 1.41) favoured topically applied nitrogen mustard with inactivated transfer factor (Fisher test P≤0.03).

Figuras y tablas -
Table 5. Summary of effects of interventions
Table 6. Rare adverse effects detected by separate adverse event search

Intervention 

Rare severe adverse effects

PUVA

transient lymphomatoid papulosis (Aronsson 1982)
basal cell carcinoma/squamous cell carcinoma (Herrmann 1995)
squamous cell carcinoma (Molin 1981)
cataract (Rupoli 2005)

extracorporeal photopheresis

sarcoma (Korpusik 2008)

imiquimod

no reported SAEs found

electron beam

total epilation (Braverman 1987, Desai 1988),
nail dystrophy/edema of hands and feet/bullae dorsum and feet/conjunctivitis/hospitalisation due to skin ulcers (Desai 1988)
diffuse permanent telangiectasia/linear sclerosis/Ischaemic ulceration of finger tips (Micaily 1983)

cyclophosphamide, doxorubicin, etoposide, vincristine

haematologic toxicity: febrile neutropenia/staphylococcal bacteraemia/disseminated herpes infection/pneumocystis carinii pneumonia/neurologic toxicity grade 3/decreased left ventricular ejection fraction (Akpek 1999)

active transfer factor

no reported SAEs found

methotrexate

severe edematous erythroderma/denudation on the trunk and extremities/Interstitial pulmonary fibrosis (Zackheim 1996)

interferon‐alpha

acrocyanosis (Campo‐Voegeli 1998)
oropharyngeal lichen planus (Kutting 1997)
seizures (Legroux‐Crespel 2003)
fatal neutropenia and sepsis (Vegna 1990)
liver toxicity (Rupoli 2005, Simoni 1987, Vegna 1990)

bexarotene

neutropenia/non‐ST‐elevation myocardial infarction/elevated liver enzymes (Abbott 2009)
lethal sepsis (Bohmeyer 2003)

peldesine (BCX‐34)

no reported SAEs found

denileukin diftitox (ONTAK)

lethal vascular leak syndrome with rhabdomyolysis (Avarbock 2008)
grade 4 infusion event (Foss 2001)

nitrogen mustard

urticaria and anaphylactoid reaction (Daughters 1973, Grunnet 1976, Sanchez 1977)
local bullous reaction (Goday 1990)
perforating follicular mucinosis (Guilhou 1980)
Stevens‐Johnson‐Syndrome (Newman 1997)
cutaneous squamous/basal cell carcinoma (Hoppe 1987)

Figuras y tablas -
Table 6. Rare adverse effects detected by separate adverse event search
Comparison 1. Topical peldesine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 Pruritus

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 1. Topical peldesine versus placebo
Comparison 2. Topical imiquimod versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 2. Topical imiquimod versus placebo
Comparison 3. Topical hypericin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 3. Topical hypericin versus placebo
Comparison 4. IFN‐α versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 Erythema

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Fever

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Myalgia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Chills or weakness

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Nausea, arthralgia, and malaise

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 4. IFN‐α versus placebo
Comparison 5. IFN‐α + PUVA versus PUVA alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clearance Show forest plot

2

122

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.87, 1.31]

Figuras y tablas -
Comparison 5. IFN‐α + PUVA versus PUVA alone
Comparison 6. Denileukin diftitox high versus low dose

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 Constitutional symptoms

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Grade 3 to 4 constitutional symptoms

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Infections

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Grade 3 to 4 infections

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Gastrointestinal syndromes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.6 Grade 3 to 4 gastrointestinal syndromes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.7 CNS syndromes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.8 Grade 3 to 4 CNS syndromes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.9 Rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.10 Grade 3 to 4 rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.11 Vascular leak syndrome

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.12 Grade 3 to 4 vascular leak syndrome

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.13 Thrombotic events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.14 Grade 3 to 4 thrombotic events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.15 Cardiopulmonary events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.16 Grade 3 to 4 cardiopulmonary events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.17 Acute infusion related events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.18 Grade 3 to 4 acute infusion related events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.19 Grade 3 to 4 laboratory abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 6. Denileukin diftitox high versus low dose
Comparison 7. Bexarotene high versus low dose

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 Photosensitivity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Constipation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Fatigue

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Pruritus

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.6 Arthralgia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.7 Nasopharyngitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.8 Headache

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.9 Insomnia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.10 Free T4 abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.11 Cholesterol abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.12 Triglycerid abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.13 SGOT/SGPT abnormalities

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3 Relapse Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 7. Bexarotene high versus low dose
Comparison 8. Extracorporal photopheresis versus PUVA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 8. Extracorporal photopheresis versus PUVA
Comparison 9. Combined therapy versus conservative therapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 Hospitalization

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Fatal myocardial infarction

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Cutaneous toxicity from electron beam therapy

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Acute non‐lymphocytic leukaemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Non‐melanoma skin cancer

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.6 Unspecified

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3 Relapse Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4 Survival rate Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

5 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 9. Combined therapy versus conservative therapy
Comparison 10. IFN‐α + PUVA versus IFN‐α + acitretin

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse effects Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 Grade I to II adverse events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Grade III adverse events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Adverse events requiring treatment discontinuation

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Flu‐like symptoms

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.5 Dryness/redness of skin or hair loss

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.6 Neurological disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.7 Psychiatric disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.8 Gastrointestinal disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.9 Elevated liver or biliary tract enzymes

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.10 Elevated triglycerides

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.11 Anemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.12 Leukopenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.13 Impotentia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.14 Redness and infiltration at application site

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 10. IFN‐α + PUVA versus IFN‐α + acitretin
Comparison 11. Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clearance Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2 Survival rate Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3 Improvement Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Figuras y tablas -
Comparison 11. Topical nitrogen mustard with active transfer factor versus topical nitrogen mustard with inactivated transfer factor