Scolaris Content Display Scolaris Content Display

Inmunoterapia para el carcinoma metastásico de células renales

Collapse all Expand all

References

References to studies included in this review

Amato 2010 {published data only}

Amato RJ, Hawkins RE, Kaufman HL, Thompson JA, Tomczak P, Szczylik C, et al. Vaccination of metastatic renal cancer patients with MVA‐5T4: a randomized, double‐blind, placebo‐controlled phase III study. Clinical Cancer Research 2010;16(22):5539‐47. [DOI: 10.1158/1078‐0432.CCR‐10‐2082]CENTRAL

Escudier 2007 {published data only}

Escudier B, Pluzanska A, Koralewski P, Ravaud A, Bracarda S, Szczylik C, et al. Bevacizumab plus interferon alfa‐2a for treatment of metastatic renal cell carcinoma: a randomised, double‐blind phase III trial. Lancet 2007;370(9605):2103‐11. [DOI: 10.1200/JCO.2009.26.7849]CENTRAL

Hudes 2007 {published data only}

Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, et al. Temsirolimus, interferon alfa, or both for advanced renal‐cell carcinoma. New England Journal of Medicine2007; Vol. 356, issue 22:2271‐81. CENTRAL

Motzer 2007 {published data only}

Motzer RJ, Hutson TE, Tomczak P, Michaelson D, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal‐cell carcinoma. New England Journal of Medicine 2007;356(2):115‐24. CENTRAL

Motzer 2015a {published data only}

Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus everolimus in advanced renal‐cell carcinoma. New England Journal of Medicine 2015;373(19):1803‐13. [DOI: 10.1056/NEJMoa1510665]CENTRAL

Negrier 2011 {published data only}

Negrier S, Gravis G, Perol D, Chevreau C, Delva R, Bay JO, et al. Temsirolimus and bevacizumab, or sunitinib, or interferon alfa and bevacizumab for patients with advanced renal cell carcinoma (TORAVA): a randomised phase 2 trial. Lancet Oncology 2011;12(7):673‐80. CENTRAL

Rini 2010 {published data only}

Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Archer L, et al. Phase III trial of bevacizumab plus interferon alfa versus interferon alfa monotherapy in patients with metastatic renal cell carcinoma: final results of CALGB 90206. Journal of Clinical Oncology 2010;28(13):2137‐43. [DOI: 10.1200/JCO.2009.26.5561]CENTRAL

Rini 2015 {published and unpublished data}

Rini B, Stenzl A, Zdrojowy R, Kogan M, Shkolnik M, Oudard S, et al. Results from an open‐label, randomized, controlled Phase 3 study investigating IMA901 multipeptide cancer vaccine in patients receiving sunitinib as first‐line therapy for advanced/metastatic RCC. European Cancer Congress. 2015:17LBA. CENTRAL

References to studies excluded from this review

Aass 2005 {published data only}

Aass N, Mulder PH, Mickisch GH, Mulders P, Oosterom AT, Poppel H, et al. Randomized phase II/III trial of interferon Alfa‐2a with and without 13‐cis‐retinoic acid in patients with progressive metastatic renal cell Carcinoma: the European Organisation for Research and Treatment of Cancer Genito‐Urinary Tract Cancer Group (EORTC 30951). Journal of Clinical Oncology2005; Vol. 23, issue 18:4172‐8. CENTRAL

Adler 1987 {published data only}

Adler A, Gillon G, Lurie H, Shaham J, Loven D, Shachter Y, et al. Active specific immunotherapy of renal cell carcinoma patients: a prospective randomized study of hormono‐immuno versus hormonotherapy. Preliminary report of immunological and clinical aspects. Journal of Biological Response Modifiers 1987;6(6):610‐24. CENTRAL

Amato 2009 {published data only}

Amato RJ, Shingler W, Goonewardena M, Belin J, Naylor S, Jac J, et al. Vaccination of renal cell cancer patients with modified vaccinia ankara delivering the tumor antigen 5T4 (TroVax) alone or administered in combination with interferon‐alpha (IFN‐alpha): a phase 2 trial. Journal of Immunotherapy2009; Vol. 32, issue 7:765‐72. CENTRAL

Amin 2015 {published data only}

Amin A, Dudek AZ, Logan TF, Lance RS, Holzbeierlein JM, Knox JJ, et al. Survival with AGS‐003, an autologous dendritic cell‐based immunotherapy, in combination with sunitinib in unfavourable risk patients with advanced renal cell carcinoma (RCC): phase 2 study results. Journal for Immunotherapy of Cancer 2015;3:14. [DOI: 10.1186/s40425‐015‐0055‐3]CENTRAL

Atkins 1993 {published data only}

Atkins MB, Sparano J, Fisher RI, Weiss GR, Margolin KA, Fink KI, et al. Randomized phase II trial of high‐dose interleukin‐2 either alone or in combination with interferon alfa‐2b in advanced renal cell carcinoma. Journal of Clinical Oncology1993; Vol. 11, issue 4:661‐70. CENTRAL

Atzpodien 2001 {published data only}

Atzpodien J, Kirchner H, Illiger HJ, Metzner B, Ukena D, Schott H, et al. IL‐2 in combination with IFN‐alpha and 5‐FU versus tamoxifen in metastatic renal cell carcinoma: long‐term results of a controlled randomized clinical trial. British Journal of Cancer 2001;85(8):1130‐6. CENTRAL

Atzpodien 2004 {published data only}

Atzpodien J, Kirchner H, Jonas U, Bergmann L, Schott H, Heynemann H, et al. Interleukin‐2‐ and interferon alfa‐2a‐based immunochemotherapy in advanced renal cell carcinoma: a prospectively randomized trial of the German Cooperative Renal Carcinoma Chemoimmunotherapy Group (DGCIN). Journal of Clinical Oncology2004; Vol. 22, issue 7:1188‐94. CENTRAL

Atzpodien 2005 {published data only}

Atzpodien J, Schmitt E, Gertenbach U, Fornara P, Heynemann H, Maskow A, et al. Adjuvant treatment with interleukin‐2‐ and interferon‐alpha2a‐based chemoimmunotherapy in renal cell carcinoma post tumour nephrectomy: results of a prospectively randomised trial of the German Cooperative Renal Carcinoma Chemoimmunotherapy Group (DGCIN). British Journal of Cancer 2005;92(5):843‐6. CENTRAL

Atzpodien 2006 {published data only}

Atzpodien J, Kirchner H, Rebmann U, Soder M, Gertenbach U, Siebels M, et al. Interleukin‐2/interferon‐alpha2a/13‐retinoic acid‐based chemoimmunotherapy in advanced renal cell carcinoma: results of a prospectively randomised trial of the German Cooperative Renal Carcinoma Chemoimmunotherapy Group (DGCIN). British Journal of Cancer 2006;95(4):463‐9. CENTRAL

Bellmunt 2008 {published data only}

Bellmunt J, Szczylik C, Feingold J, Strahs A, Berkenblit A. Temsirolimus safety profile and management of toxic effects in patients with advanced renal cell carcinoma and poor prognostic features. Annals of Oncology 2008;19(8):1387‐92. CENTRAL

Boccardo 1998 {published data only}

Boccardo F, Rubagotti A, Canobbio L, Galligioni E, Sorio R, Lucenti A, et al. Interleukin‐2, interferon‐alpha and interleukin‐2 plus interferon‐alpha in renal cell carcinoma. A randomized phase II trial. Tumori 1998;84(5):534‐9. CENTRAL

Borden 1990 {published data only}

Borden EC, Rinehart JJ, Storer BE, Trump DL, Paulnock DM, Teitelbaum AP. Biological and clinical effects of interferon‐beta ser at two doses. Journal of Interferon Research1990; Vol. 10, issue 6:559‐70. CENTRAL

Bracarda 2010 {published data only}

Bracarda S, Bellmunt J, Melichar B, Negrier S, Bajetta E, Ravaud A, et al. Overall survival in patients with metastatic renal cell carcinoma initially treated with bevacizumab plus interferon‐α2a and subsequent therapy with tyrosine kinase inhibitors: a retrospective analysis of the phase III AVOREN trial. BJU International 2010;107:214‐9. [DOI: 10.1111/j.1464‐410X.2010.09707.x]CENTRAL

Bracarda 2013 {published data only}

Bracarda S, Porta C, Boni C, Santoro A, Mucciarini C, Pazzola A, et al. Could interferon still play a role in metastatic renal cell carcinoma? A randomized study of two schedules of sorafenib plus interferon‐alpha 2a (RAPSODY). European Urology2013; Vol. 63, issue 2:254‐61. CENTRAL

Brinkmann 2004 {published data only}

Brinkmann OA, Hertle L. Combined cytokine therapy vs mistletoe treatment in metastatic renal cell cancer. Clinical comparison of therapy success with combined administration of interferon‐alpha2b, interleukin‐2, and 5‐fluorouracil compared to treatment with mistletoe lectin [in German]. Onkologe 2004;10(9):978‐85. CENTRAL

Bromwich 2002 {published data only}

Bromwich E, Hendry D, Aitchison M. Cytoreductive nephrectomy: is it a realistic option in patients with renal cancer?. BJU International 2002;89(6):523‐5. CENTRAL

Buzogany 2001 {published data only}

Buzogany I, Feher G, Kocsis J, Molnar I, Nagy G, Laszlo K, et al. Interferon alfa‐2b versus interferon alfa 2b + vinblastin treatment in advanced renal cell carcinoma; randomized multicenter trial [Intron‐A vs Intron‐A‐vinbalstin‐kezeles hatekonysaganak es bistonsagossaganak vizsgalata ketkaru, randomizalt multicentrikus vizsgalatban]. Magyar Urologia 2001;13(1):19‐31. CENTRAL

Castellano 2009 {published data only}

Castellano D, del Muro XG, Perez‐Gracia JL, Gonzalez‐Larriba JL, Abrio MV, Ruiz MA, et al. Patient‐reported outcomes in a phase III, randomized study of sunitinib versus interferon‐{alpha} as first‐line systemic therapy for patients with metastatic renal cell carcinoma in a European population. Annals of Oncology 2009;20(11):1803‐12. CENTRAL

Cella 2008 {published data only}

Cella D, Li JZ, Cappelleri JC, Bushmakin A, Charbonneau C, Kim ST, et al. Quality of life in patients with metastatic renal cell carcinoma treated with sunitinib or interferon alfa: results from a phase III randomized trial. Journal of Clinical Oncology 2008;26(22):3763‐9. CENTRAL

Cella 2010 {published data only}

Cella D, Michaelson MD, Bushmakin AG, Cappelleri JC, Charbonneau C, Kim ST, et al. Health‐related quality of life in patients with metastatic renal cell carcinoma treated with sunitinib vs interferon‐alpha in a phase III trial: final results and geographical analysis. British Journal of Cancer 2010;102(4):658‐64. CENTRAL

Cella 2016 {published data only}

Cella D, Grünwald V, Nathan P, Doan J, Dastani H, Taylor F, et al. Quality of life in patients with advanced renal cell carcinoma given nivolumab versus everolimus in CheckMate 025: a randomised, open‐label, phase 3 trial. Lancet Oncology 2016;17:994‐1003. CENTRAL

Choueiri 2014 {published data only}

Choueiri TK, Fishman MN, Escudier B, McDermott DF, Kluger H, Stadler WM, et al. Immunomodulatory activity of nivolumab in metastatic renal cell carcinoma (mRCC): association of biomarkers with clinical outcomes. Asia‐Pacific Journal of Clinical Oncology 2015;11:119. CENTRAL

Clark 2003 {published data only}

Clark JI, Atkins MB, Urba WJ, Creech S, Figlin RA, Dutcher JP, et al. Adjuvant high‐dose bolus interleukin‐2 for patients with high‐risk renal cell carcinoma: a cytokine working group randomized trial. Journal of Clinical Oncology : official journal of the American Society of Clinical Oncology 2003;21(16):3133‐40. CENTRAL

Creagan 1991 {published data only}

Creagan ET, Twito DI, Johansson SL, Schaid DJ, Johnson PS, Flaum MA, et al. A randomized prospective assessment of recombinant leukocyte a human interferon with or without aspirin in advanced renal adenocarcinoma. Journal of Clinical Oncology1991; Vol. 9, issue 12:2104‐9. CENTRAL

De Mulder 1995 {published data only}

De Mulder PH, Oosterhof G, Bouffioux C, van Oosterom AT, Vermeylen K, Sylvester R. EORTC (30885) randomised phase III study with recombinant interferon alpha and recombinant interferon alpha and gamma in patients with advanced renal cell carcinoma. The EORTC Genitourinary Group. British Journal of Cancer 1995;71(2):371‐5. CENTRAL

Dexeus 1989 {published data only}

Dexeus FH, Logothetis CJ, Sella A, Finn L. Interferon alternating with chemotherapy for patients with metastatic renal cell carcinoma. American Journal of Clinical Oncology1989; Vol. 12, issue 4:350‐4. CENTRAL

Dillman 2003 {published data only}

Dillman RO, Wiemann M, Nayak SK, DeLeon C, Hood K, DePriest C. Interferon‐gamma or granulocyte‐macrophage colony‐stimulating factor administered as adjuvants with a vaccine of irradiated autologous tumor cells from short‐term cell line cultures: a randomized phase 2 trial of the cancer biotherapy research group. Journal of Immunotherapy 2003;26(4):367‐73. CENTRAL

Donskov 2006 {published data only}

Donskov F, Hokland M, Marcussen N, Torp Madsen HH, von der Maase H. Monocytes and neutrophils as 'bad guys' for the outcome of interleukin‐2 with and without histamine in metastatic renal cell carcinoma ‐ results from a randomised phase II trial. British Journal of Cancer 2006;94(2):218‐26. CENTRAL

Du Bois 1997 {published data only}

Du Bois JS, Trehu EG, Mier JW, Shapiro L, Epstein M, Klempner M, et al. Randomized placebo‐controlled clinical trial of high‐dose interleukin‐2 in combination with a soluble p75 tumor necrosis factor receptor immunoglobulin G chimera in patients with advanced melanoma and renal cell carcinoma. Journal of Clinical Oncology 1997;15(3):1052‐62. CENTRAL

Dudek 2008 {published data only}

Dudek AZ, Mescher MF, Okazaki I, Math VT, Luo X, Curtsinger JM, et al. Autologous large multivalent immunogen vaccine in patients with metastatic melanoma and renal cell carcinoma. American Journal of Clinical Oncology 2008;31(2):173‐81. CENTRAL

Dutcher 2003 {published data only}

Dutcher JP, Fine JP, Krigel RL, Murphy BA, Schaefer PL, Ernstoff MS, et al. Stratification by risk factors predicts survival on the active treatment arm in a randomized phase II study of interferon‐gamma plus/minus interferon‐alpha in advanced renal cell carcinoma (E6890). Medical Oncology 2003;20(3):271‐81. CENTRAL

Dutcher 2009 {published data only}

Dutcher JP, de Souza P, McDermott D, Figlin RA, Berkenblit A, Thiele A, et al. Effect of temsirolimus versus interferon‐alpha on outcome of patients with advanced renal cell carcinoma of different tumor histologies. Medical Oncology 2009;26(2):202‐9. CENTRAL

Edsmyr 1985 {published data only}

Edsmyr F, Esposti PL, Andersson L. Interferon therapy in disseminated renal cell carcinoma. Radiotherapy and Oncology 1985;4(1):21‐6. CENTRAL

Elkord 2013 {published and unpublished data}

Elkord E, Burt DJ, Sundstedt A, Nordle Ö, Hedlund G, Hawkins RE. Immunological response and overall survival in a subset of advanced renal cell carcinoma patients from a randomized phase 2/3 study of naptumomab estafenox plus IFN‐a versus IFN‐a. Oncotarget 2015;6(6):4428‐39. CENTRAL

Escudier 2009 {published data only}

Escudier B, Szczylik C, Hutson TE, Demkow T, Staehler M, Rolland F, et al. Randomized phase II trial of first‐line treatment with sorafenib versus interferon alfa‐2a in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology 2009;27(8):1280‐9. CENTRAL

Escudier 2010 {published data only}

Escudier B, Bellmunt J, Negrier S, Bajetta E, Melichar B, Bracarda S, et al. Phase III trial of bevacizumab plus interferon alfa‐2a in patients with metastatic renal cell carcinoma (AVOREN): final analysis of overall survival. Journal of Clinical Oncology 2010;28(13):2144‐50. [DOI: 10.1200/JCO.2009.26.7849]CENTRAL

Escudier 2011 {published data only}

Escudier BJ, Negrier S, Perol D, Gravis G, Delva R, Bay J, et al. Prognostic factors for progression‐free survival (PFS) in patients with metastatic renal cell carcinoma (mRCC): results from the French randomized phase II study TORAVA. Journal of Clinical Oncology 2011;29(Suppl):15. CENTRAL

Fenton 1996 {published data only}

Fenton RG, Steis RG, Madara K, Zea AH, Ochoa AC, Janik JE, et al. A phase I randomized study of subcutaneous adjuvant IL‐2 in combination with an autologous tumor vaccine in patients with advanced renal cell carcinoma. Journal of Immunotherapy 1996;19(5):364‐74. CENTRAL

Figlin 1999 {published data only}

Figlin RA, Thompson JA, Bukowski RM, Vogelzang NJ, Novick AC, Lange P, et al. Multicenter, randomized, phase III trial of CD8(+) tumor‐infiltrating lymphocytes in combination with recombinant interleukin‐2 in metastatic renal cell carcinoma. Journal of Clinical Oncology1999; Vol. 17, issue 8:2521‐9. CENTRAL

Figlin 2009 {published data only}

Figlin RA, de Souza P, McDermott D, Dutcher JP, Berkenblit A, Thiele A, et al. Analysis of PTEN and HIF‐1alpha and correlation with efficacy in patients with advanced renal cell carcinoma treated with temsirolimus versus interferon‐alpha. Cancer 2009;115(16):3651‐60. CENTRAL

Flanigan 2001 {published data only}

Flanigan RC, Salmon SE, Blumenstein BA, Bearman SI, Roy V, McGrath PC, et al. Nephrectomy followed by interferon alfa‐2b compared with interferon alfa‐2b alone for metastatic renal‐cell cancer. New England Journal of Medicine 2001;345(23):1655‐9. CENTRAL

Foon 1988 {published data only}

Foon K, Doroshow J, Bonnem E, Fefer A, Graham S, Grosh B, et al. A prospective randomized trial of alpha 2B‐interferon/gamma‐interferon or the combination in advanced metastatic renal cell carcinoma. Journal of Biological Response Modifiers1988; Vol. 7, issue 6:540‐5. CENTRAL

Fosså 1992 {published data only}

Fosså SD, Martinelli G, Otto U, Schneider G, Wander H, Oberling F, et al. Recombinant interferon alfa‐2a with or without vinblastine in metastatic renal cell carcinoma: results of a European multi‐center phase III study. Annals of Oncology1992; Vol. 3, issue 4:301‐5. CENTRAL

Fosså 2004 {published data only}

Fosså SD, Mickisch GH, De Mulder PH, Horenblas S, van Oosterom AT, van Poppel H, et al. Interferon‐alpha‐2a with or without 13‐cis retinoic acid in patients with progressive, measurable metastatic renal cell carcinoma. Cancer 2004;101(3):533‐40. CENTRAL

Fujita 1992 {published data only}

Fujita T, Fukushima M. Interferon therapy for advanced renal cell carcinoma [in Japanese]. Hinyokika Kiyo ‐ Acta Urologica Japonica 1992;38(11):1293‐8. [MEDLINE: 93135084]CENTRAL
Fujita T, Inagaki J, Asano H, Naide Y, Ono Y, Ohshima S, et al. Effects of low‐dose interferon‐alpha (Hlbi) following nephrectomy in metastatic renal cell carcinoma. 28th Annual Meeting of the American Society of Clinical Oncology; 1992 May 17‐19; San Diego (CA); A685. [MEDLINE: 92681414]CENTRAL

Galligioni 1996 {published data only}

Galligioni E, Quaia M, Merlo A, Carbone A, Spada A, Favaro D, et al. Adjuvant immunotherapy treatment of renal carcinoma patients with autologous tumor cells and bacillus Calmette‐Guerin: five‐year results of a prospective randomized study. Cancer 1996;77(12):2560‐6. CENTRAL

Gleave 1998 {published data only}

Gleave ME, Elhilali M, Fradet Y, Davis I, Venner P, Saad F, et al. Interferon gamma‐1b compared with placebo in metastatic renal‐cell carcinoma. Canadian Urologic Oncology Group. New England Journal of Medicine1998; Vol. 338, issue 18:1265‐71. CENTRAL

Gore 2010 {published data only}

Gore ME, Griffin CL, Hancock B, Patel PM, Pyle L, Aitchison M, et al. Interferon alfa‐2a versus combination therapy with interferon alfa‐2a, interleukin‐2, and fluorouracil in patients with untreated metastatic renal cell carcinoma (MRC RE04/EORTC GU 30012): an open‐label randomised trial. Lancet 2010;375(9715):641‐8. CENTRAL

Harlin 2004 {published data only}

Harlin H, Artz AS, Mahowald M, Rini BI, Zimmerman T, Vogelzang NJ, et al. Clinical responses following nonmyeloablative allogeneic stem cell transplantation for renal cell carcinoma are associated with expansion of CD8+ IFN‐gamma‐producing T cells. Bone Marrow Transplantation 2004;33(5):491‐7. CENTRAL

Henriksson 1998 {published data only}

Henriksson R, Nilsson S, Colleen S, Wersall P, Helsing M, Zimmerman R, et al. Survival in renal cell carcinoma‐a randomized evaluation of tamoxifen vs interleukin 2, alpha‐interferon (leucocyte) and tamoxifen. British Journal of Cancer 1998;77(8):1311‐7. CENTRAL

Jayson 1998 {published data only}

Jayson GC, Middleton M, Lee SM, Ashcroft L, Thatcher N. A randomized phase II trial of interleukin 2 and interleukin 2‐interferon alpha in advanced renal cancer. British Journal of Cancer 1998;78(3):366‐9. CENTRAL

Jocham 2004 {published data only}

Jocham D, Richter A, Hoffmann L, Iwig K, Fahlenkamp D, Zakrzewski G, et al. Adjuvant autologous renal tumour cell vaccine and risk of tumour progression in patients with renal‐cell carcinoma after radical nephrectomy: phase III, randomised controlled trial. Lancet 2004;363(9409):594‐9. CENTRAL

Jonasch 2010 {published data only}

Jonasch E, Corn P, Pagliaro LC, Warneke CL, Johnson MM, Tamboli P, et al. Upfront, randomized, phase 2 trial of sorafenib versus sorafenib and low‐dose interferon alfa in patients with advanced renal cell carcinoma ‐ clinical and biomarker analysis. Cancer 2010;116(1):57‐65. CENTRAL

Keefe 2015 {published data only}

Keefe SM, Hoffmann‐Censits JH, Mamtani R, Walicki M, Robinson J, Smith A, et al. HIF inhibition in metastatic renal cell carcinoma (mRCC): final results of a phase Ib/IIa clinical trial evaluating the nanoparticle drug conjugate (NDC), CRLX101, in combination with bevacizumab (bev). Journal of Clinical Oncology 2015;33(Suppl 1):15. CENTRAL

Kempf 1986 {published data only}

Kempf RA, Grunberg SM, Daniels JR, Skinner DG, Venturi CL, Spiegel R, et al. Recombinant interferon alpha‐2 (INTRON A) in a phase II study of renal cell carcinoma. Journal of Biological Response Modifiers 1986;5(1):27‐35. CENTRAL

Kim 2015 {published data only}

Kim H, Halabi S, Li P, Mayhew G, Simko J, Nixon A, et al. A prognostic model for overall survival in patients with metastatic clear cell renal carcinoma: results from CALGB 90206 (alliance). Journal of Urology 2015;193(Suppl):e526. CENTRAL

Kinouchi 2004 {published data only}

Kinouchi T, Sakamoto J, Tsukamoto T, et al. Prospective randomized trial of natural interferon‐alpha (IFN) versus IFN + cimetidine in advanced renal cell carcinoma with pulmonary metastasis. 40th Annual Meeting of the American Society of Clinical Oncology; 2004 June 5‐8; New Orleans (LA); A4676. CENTRAL

Kirkwood 1985 {published data only}

Kirkwood JM, Harris JE, Vera R. A randomized study of low and high doses of leukocyte alpha‐interferon in metastatic renal cell carcinoma: the American Cancer Society Collaborative Trial. Cancer Research 1985;45(2):863‐71. CENTRAL

Koretz 1991 {published data only}

Koretz MJ, Lawson DH, York RM, Graham SD, Murray DR, Gillespie TM, et al. Randomized study of interleukin 2 (IL‐2) alone vs IL‐2 plus lymphokine‐activated killer cells for treatment of melanoma and renal cell cancer. Archives of Surgery 1991;126(7):898‐903. CENTRAL

Kriegmair 1995 {published data only}

Kriegmair M, Oberneder R, Hofstetter A. Interferon alfa and vinblastine versus medroxyprogesterone acetate in the treatment of metastatic renal cell carcinoma. Urology 1995;45(5):758‐62. CENTRAL

Kwitkowski 2010 {published data only}

Kwitkowski VE, Prowell TM, Ibrahim A, Farrell AT, Justice R, Mitchell SS, et al. FDA approval summary: temsirolimus as treatment for advanced renal cell carcinoma. Oncologist 2010;15(4):428‐35. CENTRAL

Law 1995 {published data only}

Law TM, Motzer RJ, Mazumdar M, Sell KW, Walther PJ, O'Connell M, et al. Phase III randomized trial of interleukin‐2 with or without lymphokine‐activated killer cells in the treatment of patients with advanced renal cell carcinoma. Cancer 1995;76(5):824‐32. CENTRAL

Lissoni 1993 {published data only}

Lissoni P, Barni S, Ardizzoia A, Andres M, Scardino E, Cardellini P, et al. A randomized study of low‐dose interleukin‐2 subcutaneous immunotherapy versus interleukin‐2 plus interferon‐alpha as first line therapy for metastatic renal cell carcinoma. Tumori 1993;79(6):397‐400. CENTRAL

Lissoni 2000 {published data only}

Lissoni P, Mandala, Brivio F. Abrogation of the negative influence of opioids on IL‐2 immunotherapy of renal cell cancer by melatonin. European Urology 2000;38:115‐8. CENTRAL

Lissoni 2003 {published data only}

Lissoni P, Mengo S, Bucovec R, Brivio F, Fumagalli L, Tancini G, et al. Clinical and biological effects of interleukin‐2 with or without a concomitant administration of granulocyte‐macrophage colony‐stimulating factor in metastatic cancer patients. In Vivo 2003;17(1):73‐5. CENTRAL

Liu 2012 {published data only}

Liu L, Zhang W, Qi X, Li H, Yu J, Wei S, et al. Randomized study of autologous cytokine‐induced killer cell immunotherapy in metastatic renal carcinoma. Clinical Cancer Research 2012;18(6):1751‐9. CENTRAL

Lummen 1996 {published data only}

Lummen G, Goepel M, Mollhoff S, Hinke A, Otto T, Rubben H. Phase II study of interferon‐gamma versus interleukin‐2 and interferon‐alpha 2b in metastatic renal cell carcinoma. Journal of Urology 1996;155(2):455‐8. CENTRAL

Majhail 2006 {published data only}

Majhail NS, Wood L, Elson P, Finke J, Olencki T, Bukowski RM. Adjuvant subcutaneous interleukin‐2 in patients with resected renal cell carcinoma: a pilot study. Clinical Genitourinary Cancer 2006;5(1):50‐6. CENTRAL

Margolin 1997 {published data only}

Margolin K, Atkins M, Sparano J, Sosman J, Weiss G, Lotze M, et al. Prospective randomized trial of lisofylline for the prevention of toxicities of high‐dose interleukin 2 therapy in advanced renal cancer and malignant melanoma. Clinical Cancer Research 1997;3(4):565‐72. CENTRAL

McCabe 1991 {published data only}

McCabe MS, Stablein D, Hawkins MJ. The Modified Group C experience ‐ phase III randomized trials of IL‐2 vs IL‐2+LAK in advanced renal cell carcinoma and advanced melanoma. 35th Annual Meeting of the American Society of Clinical Oncology; 1999 May 15‐18; Atlanta, GA; A714. CENTRAL

McDermott 2005 {published data only}

McDermott DF, Regan MM, Clark JI, Flaherty LE, Weiss GR, Logan TF, et al. Randomized phase III trial of high‐dose interleukin‐2 versus subcutaneous interleukin‐2 and interferon in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology2005; Vol. 23, issue 1:133‐41. CENTRAL

Melichar 2008 {published data only}

Melichar B, Koralewski P, Ravaud A, Pluzanska A, Bracarda S, Szczylik C, et al. First‐line bevacizumab combined with reduced dose interferon‐alpha2a is active in patients with metastatic renal cell carcinoma. Annals of Oncology 2008;19(8):1470‐6. CENTRAL

Messing 2003 {published data only}

Messing EM, Manola J, Wilding G, Propert K, Fleischmann J, Crawford ED, et al. Phase III study of interferon alfa‐NL as adjuvant treatment for resectable renal cell carcinoma: an Eastern Cooperative Oncology Group/Intergroup trial. Journal of Clinical Oncology 2003;21(7):1214‐22. CENTRAL

Mickisch 2001 {published data only}

Mickisch GHJ, Garin A, van Poopel H, de Prijck L, Sylvester R, European Organisation for Research and Treatment of Cancer (EORTC) Genitourinary Group. Radical nephrectomy plus interferon‐alfa‐based immunotherapy compared with interferon alfa alone in metastatic renal‐cell carcinoma: a randomized trial. Lancet 2001;358:966‐70. CENTRAL

Motzer 2000 {published data only}

Motzer RJ, Murphy BA, Bacik J, Schwartz LH, Nanus DM, Mariani T, et al. Phase III trial of interferon alfa‐2a with or without 13‐cis‐retinoic acid for patients with advanced renal cell carcinoma. Journal of Clinical Oncology 2000;18(16):2972‐80. CENTRAL

Motzer 2001 {published data only}

Motzer RJ, Rakhit A, Thompson JA, Nemunaitis J, Murphy BA, Ellerhorst J, et al. Randomized multicenter phase II trial of subcutaneous recombinant human interleukin‐12 versus interferon‐alpha 2a for patients with advanced renal cell carcinoma. Journal of Interferon & Cytokine Research 2001;21(4):257‐63. CENTRAL

Motzer 2009 {published data only}

Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Oudard S, et al. Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology 2009;27(22):3584‐90. CENTRAL

Motzer 2015b {published data only}

Motzer RJ, Bono P, Hudes GR, Tomita Y, Ravaud A, Waxman I, et al. A phase III comparative study of nivolumab (anti‐PD‐1; BMS‐936558; ONO‐4538) versus everolimus in patients (pts) with advanced or metastatic renal cell carcinoma (mRCC) previously treated with antiangiogenic therapy. Journal of Clinical Oncology 2015;31(15 (suppl)):TP4592. CENTRAL

Motzer 2015c {published data only}

Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel T, Harrison MR, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. Journal of Clinical Oncology 2015;33(13):1430‐7. CENTRAL

MRCRCC 1999 {published data only}

Medical Research Council Renal Cancer Collaborators. Interferon‐alpha and survival in metastatic renal cell carcinoma: early results of a randomized controlled trial. Lancet 1999;353:14‐7. CENTRAL

Muss 1987 {published data only}

Muss HB, Costanzi JJ, Leavitt R. Recombinant alfa interferon in renal cell carcinoma: a randomized trial of two routes of administration. Journal of Clinical Oncology 1987;5(2):286‐91. CENTRAL

Naglieri 1998 {published data only}

Naglieri E, Gebbia V, Durini E, Lelli G, Abbate I, Selvaggi FP, et al. Standard interleukin‐2 (IL‐2) and interferon‐alpha immunotherapy versus an IL‐2 and 4‐epirubicin immuno‐chemotherapeutic association in metastatic renal cell carcinoma. Anticancer Research 1998;18(38):2021‐6. CENTRAL

NCT00352859 {published data only}

NCT00352859. A randomized discontinuation trial to determine the clinical benefit of continuation of sorafenib following disease progression in patients with advanced renal cell carcinoma, 2006. clinicaltrials.gov/ct2/show/NCT00352859 (assessed 30 May 2016). CENTRAL

NCT00678288 {published data only}

NCT00678288. A phase II, randomized, open‐label, multicenter, study evaluating the efficacy of sorafenib alone and sorafenib in combination with low dose interferon alpha‐2a as second‐line treatment of sunitinib failure in patients with metastatic renal cell carcinoma, 2008. clinicaltrials.gov/ct2/show/NCT00678288 (assessed 30 May 2016). CENTRAL

Negrier 1998 {published data only}

Escudier B, Chevreau C, Lasset C, Douillard JY, Ravaud A, Fabbro M, et al. Cytokines in metastatic renal cell carcinoma: is it useful to switch to interleukin‐2 or interferon after failure of a first treatment? Groupe Francais d'Immunotherape. Journal of Clinical Oncology 1999;17(7):2039‐43. CENTRAL
Negrier S, Escudier B, Lasset C, Douillard J Y, Savary J, Chevreau C, et al. Recombinant human interleukin‐2, recombinant human interferon alfa‐2a, or both in metastatic renal‐cell carcinoma. Groupe Français d'Immunothérapie. New England Journal of Medicine 1998;338(18):1272‐8. CENTRAL

Negrier 2000 {published data only}

Negrier S, Caty A, Lesimple T, Douillard JY, Escudier B, Rossi JF, et al. Treatment of patients with metastatic renal carcinoma with a combination of subcutaneous interleukin‐2 and interferon alfa with or without fluorouracil. Groupe Français d'Immunothérapie, Fédération Nationale des Centres de Lutte Contre le Cancer. Journal of Clinical Oncology2000; Vol. 18, issue 24:4009‐15. CENTRAL

Negrier 2007 {published data only}

Negrier S, Perol D, Ravaud A, Chevreau C, Bay JO, Delva R, et al. Medroxyprogesterone, interferon alfa‐2a, interleukin 2, or combination of both cytokines in patients with metastatic renal carcinoma of intermediate prognosis: results of a randomized controlled trial. Cancer 2007;110(11):2468‐77. CENTRAL

Negrier 2008 {published data only}

Negrier S, Perol D, Ravaud A, Bay JO, Oudard S, Chabaud S, et al. Randomized study of intravenous versus subcutaneous interleukin‐2, and IFNalpha in patients with good prognosis metastatic renal cancer. Clinical Cancer Research 2008;14(18):5907‐12. CENTRAL

Negrier 2010 {published data only}

Negrier S, Jager E, Porta C, McDermott D, Moore M, Bellmunt J, et al. Efficacy and safety of sorafenib in patients with advanced renal cell carcinoma with and without prior cytokine therapy, a subanalysis of TARGET. Medical Oncology 2010;27(3):899‐906. CENTRAL

Neidhart 1991 {published data only}

Neidhart JA, Anderson SA, Harris JE, Rinehart JJ, Laszlo J, Dexeus FH, et al. Vinblastine fails to improve response of renal cancer to interferon alfa‐n1: high response rate in patients with pulmonary metastases. Journal of Clinical Oncology 1991;9(5):832‐6. [MEDLINE: 91202210]CENTRAL

Osband 1990 {published data only}

Osband ME, Lavin PT, Babayan RK, Graham S, Lamm W, Parker B, et al. Effect of autolymphocyte therapy on survival and quality of life in patients with metastatic renal‐cell carcinoma. Lancet 1990;335(8696):994‐8. CENTRAL

Otto 1988 {published data only}

Otto U, Schneider A, Denkhaus H, Conrad S. Treatment of metastatic kidney cancer with recombinant alpha‐2 or gamma interferon. Results of 2 clinical phase II and III studies [Die Behandlung des metastasierenden Nierenkarzinoms mit rekombinantem alpha‐2‐ oder gamma‐Interferon. Ergebnisse zweier klinischer Phase‐II‐ bzw. Phase III‐Studien]. Onkologie 1988;11(4):185‐91. CENTRAL

Oudard 2011 {published data only}

Oudard S, Beuselinck B, Decoene J, Albers P. Sunitinib for the treatment of metastatic renal cell carcinoma. Cancer Treatment Reviews 2011;37(3):178‐84. CENTRAL

Passalacqua 2010 {published data only}

Passalacqua R, Buzio C, Buti S, Porta C, Labianca R, Pezzuolo D, et al. Phase III, randomised, multicentre trial of maintenance immunotherapy with low‐dose interleukin‐2 and interferon‐alpha for metastatic renal cell cancer. Cancer Immunology, Immunotherapy 2010;59(4):553‐61. CENTRAL

Passalacqua 2014 {published data only}

Passalacqua R, Caminiti C, Buti S, Porta C, Camisa R, Braglia L, et al. Adjuvant low‐dose interleukin‐2 (IL‐2) plus interferon‐alpha (IFN‐alpha) in operable renal cell carcinoma (RCC): a phase III, randomized, multicentre trial of the Italian oncology group for clinical research (GOIRC). Journal of Immunotherapy 2014;37(9):440‐7. CENTRAL

Patel 2008 {published data only}

Patel PM, Sim S, O'Donnell DO, Protheroe A, Beirne D, Stanley A, et al. An evaluation of a preparation of Mycobacterium vaccae (SRL172) as an immunotherapeutic agent in renal cancer. European Journal of Cancer 2008;44(2):216‐23. CENTRAL

Patil 2012 {published data only}

Patil S, Figlin RA, Hutson TE, Michaelson MD, Negrier S, Kim ST, et al. Q‐TWiST analysis to estimate overall benefit for patients with metastatic renal cell carcinoma treated in a phase III trial of sunitinib vs interferon‐alpha. British Journal of Cancer 2012;106(10):1587‐90. CENTRAL

Pedersen 1980 {published data only}

Pedersen F, Hippe E, Hvidt V, Francis D, Jensen HS, Dybkjaer E, et al. Treatment of metastasizing renal adenocarcinoma with specific plasma transfusion. A controlled trial of the effects on metastases and survival time [in Danish] [Behandling af metastaserende adenocarcinoma renis med specifik plasmatransfusion]. Ugeskrift for Laeger 1980;142(48):3167‐70. [MEDLINE: 81080479]CENTRAL

Pickering 2009 {published data only}

Pickering LM, Pyle L, Larkin JMG. Sunitinib is superior to interferon alfa with respect to quality of life for patients with renal cell carcinoma. Nature Review Clinical Oncology 2009;6(1):6‐7. CENTRAL

Pizzocaro 2001 {published data only}

Pizzocaro G, Piva L, Colavita M, Ferri S, Artusi R, Boracchi P, et al. Interferon adjuvant to radical nephrectomy in Robson stages II and III renal cell carcinoma: a multicentric randomized study. Journal of Clinical Oncology 2001;19(2):425‐31. CENTRAL

Porzsolt 1988 {published data only}

Porzsolt F, Messerer D, Hautmann R, Gottwald A, Sparwasser H, Stockamp K, et al. Treatment of advanced renal cell cancer with recombinant interferon alpha as a single agent and in combination with medroxyprogesterone acetate. A randomized multicenter trial. Journal of Cancer Research & Clinical Oncology 1988;114(1):95‐100. [MEDLINE: 88169678]CENTRAL

Powles 2015 {published data only}

Powles T, Wheater MJ, Din O, Geldart TR, Boleti E, Stockdale A, et al. A randomized phase II study of AZ2014 versus everolimus in patients with VEGF refractory metastatic clear cell renal cancer (mRCC). Journal of Clinical Oncology 2015;33(Suppl 1):7. CENTRAL

Procopio 2011 {published data only}

Procopio G, Verzoni E, Bracarda S, Ricci S, Sacco C, Ridolfi L, et al. Sorafenib with interleukin‐2 vs sorafenib alone in metastatic renal cell carcinoma: the ROSORC trial. British Journal of Cancer 2011;104(8):1256‐61. CENTRAL

Pyrhönen 1999 {published data only}

Pyrhönen S, Salminen E, Ruutu M, Lehtonen T, Nurmi M, Tammela T, et al. Prospective randomized trial of interferon alfa‐2a plus vinblastine versus vinblastine alone in patients with advanced renal cell cancer. Journal of Clinical Oncology1999; Vol. 17, issue 9:2859‐67. CENTRAL

Quesada 1985 {published data only}

Quesada JR, Rios A, Swanson D. Antitumor activity of recombinant‐derived interferon alpha in metastatic renal cell carcinoma. Journal of Clinical Oncology 1985;3(11):1522‐8. CENTRAL

Radosavljevic 2000 {published data only}

Radosavljevic D, Jelic S, Babovic N, Popov I, Kreacic M, Stamatovic L, et al. Addition of medroxyprogesterone‐acetate to interferon‐a ‐ vinblastine combination in advanced renal‐cell carcinoma ‐ is there any impact on quality of life?. Annals of Oncology 2000;11 Suppl 4:332P. CENTRAL

Ravaud 2015 {published data only}

Ravaud A, Barrios C, Alekseev B, Tay MH, Agarwala SS, Yalcin S, et al. RECORD‐2: phase II randomized study of everolimus and bevacizumab versus interferon α‐2a and bevacizumab as first‐line therapy in patients with metastatic renal cell carcinoma. Annals of Oncology 2015;26(7):1378‐84. [DOI: 110.1093/annonc/mdv170]CENTRAL

Reddy 2006 {published data only}

Reddy K. Phase III study of sunitinib malate (SU11248) versus interferon‐alpha as first‐line treatment in patients with metastatic renal cell carcinoma. Clinical Genitourinary Cancer 2006;5(1):23‐5. CENTRAL

Rini 2004 {published data only}

Rini BI, Halabi S, Taylor J, Small EJ, Schilsky RL. Cancer and Leukemia Group B 90206: a randomized phase III trial of interferon‐alpha or interferon‐alpha plus anti‐vascular endothelial growth factor antibody (bevacizumab) in metastatic renal cell carcinoma. Clinical Cancer Research 2004;10(8):2584‐6. CENTRAL

Rini 2008 {published data only}

Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Ou S‐S, et al. Bevacizumab plus interferon alfa compared with interferon alfa monotherapy in patients with metastatic renal cell carcinoma: CALGB 90206. Journal of Clinical Oncology 2008;26(33):5422‐8. CENTRAL

Rini 2012 {published data only}

Rini B, Szczylik C, Tannir NM, Koralewski P, Tomczak P, Deptala A, et al. AMG 386 in combination with sorafenib in patients with metastatic clear cell carcinoma of the kidney: a randomized, double‐blind, placebo‐controlled, phase 2 study. Cancer 2012;118(24):6152‐61. CENTRAL

Rini 2014 {published data only}

Rini BI, Bellmunt J, Clancy J, Wang K, Niethammer AG, Hariharan S, et al. Randomized phase III trial of temsirolimus and bevacizumab versus interferon alfa and bevacizumab in metastatic renal cell carcinoma: INTORACT trial. Journal of Clinical Oncology 2014;32(8):752‐9. CENTRAL

Rosenberg 1993 {published data only}

Rosenberg SA, Lotze MT, Yang JC, Topalian SL, Chang AE, Schwartzentruber DJ, et al. Prospective randomized trial of high‐dose interleukin‐2 alone or in conjunction with lymphokine‐activated killer cells for the treatment of patients with advanced cancer. Journal of the National Cancer Institute 1993;85(8):622‐32. CENTRAL

Rossi 2010 {published data only}

Rossi JF, Negrier S, James ND, Kocak I, Hawkins R, Davis H, et al. A phase I/II study of siltuximab (CNTO 328), an anti‐interleukin‐6 monoclonal antibody, in metastatic renal cell cancer. British Journal of Cancer 2010;103(8):1154‐62. CENTRAL

Sagaster 1995 {published data only}

Sagaster P, Micksche M, Flamm J, Ludwig H. Randomised study using IFN‐alpha versus IFN‐alpha plus coumarin and cimetidine for treatment of advanced renal cell cancer. Annals of Oncology 1995;6(10):999‐1003. [MEDLINE: 96343308]CENTRAL

Scardino 1997 {published data only}

Scardino E, Lissoni P, Andres M, Frea B, Favini P, Kocjancic E, et al. Preoperative subcutaneous immunotherapy with interleukin‐2 in renal cell carcinoma with synchronous metastases: a clinicobiological randomized study [in Italian]. Archivio Italiano di Urologia e Andrologia 1997;69(1):49‐54. CENTRAL

Schwaab 2000 {published data only}

Schwaab T, Heaney JA, Schned AR, Harris RD, Cole BF, Noelle RJ, et al. A randomized phase II trial comparing two different sequence combinations of autologous vaccine and human recombinant interferon gamma and human recombinant interferon alpha2B therapy in patients with metastatic renal cell carcinoma: clinical outcome and analysis of immunological parameters. Journal of Urology 2000;163(4):1322‐7. CENTRAL

Sharma 2015 {published data only}

Sharma P, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. CheckMate 025: a randomized, open‐label phase III study of nivolumab (NIVO) versus everolimus (EVE) in advanced renal cell carcinoma (RCC). European Journal Cancer 2015;51:S708. CENTRAL

Simons 1997 {published data only}

Simons JW, Jaffee EM, Weber CE, Levitsky HI, Nelson WG, Carducci MA, et al. Bioactivity of autologous irradiated renal cell carcinoma vaccines generated by ex vivo granulocyte‐macrophage colony‐stimulating factor gene transfer. Cancer Research 1997;57(8):1537‐46. CENTRAL

Smith 2003 {published data only}

Smith JW, Kurt RA, Baher AG, Denman S, Justice L, Doran T, et al. Immune effects of escalating doses of granulocyte‐macrophage colony‐stimulating factor added to a fixed, low‐dose, inpatient interleukin‐2 regimen: a randomized phase I trial in patients with metastatic melanoma and renal cell carcinoma. Journal of Immunotherapy 2003;26(2):130‐8. CENTRAL

Soret 1996 {published data only}

Soret JY, Escudier B. Adjuvant treatment with IL‐2 or interferon‐alpha in renal cell carcinoma: a French multicentric study. Cancer Biotherapy & Radiopharmaceuticals 1996;11(5):301‐2. CENTRAL

Steineck 1990 {published data only}

Steineck G, Strander H, Carbin BE, Borgström E, Wallin L, Achtnich U, et al. Recombinant leukocyte interferon alpha‐2a and medroxyprogesterone in advanced renal cell carcinoma. A randomized trial. Acta Oncology1990; Vol. 29, issue 2:155‐62. CENTRAL

Sternberg 2013 {published data only}

Sternberg CN, Hawkins RE, Wagstaff J, Salman P, Mardiak J, Barrios CH, et al. A randomised, double‐blind phase III study of pazopanib in patients with advanced and/or metastatic renal cell carcinoma: final overall survival results and safety update. European Journal of Cancer 2013;49(6):1287‐96. CENTRAL

Summers 2010 {published data only}

Summers J, Cohen MH, Keegan P, Pazdur R. FDA drug approval summary: bevacizumab plus interferon for advanced renal cell carcinoma. Oncologist 2010;15(1):104‐11. CENTRAL

Tannir 2006 {published data only}

Tannir NM, Cohen L, Wang X, Thall P, Mathew PF, Jonasch E, et al. Improved tolerability and quality of life with maintained efficacy using twice‐daily low‐dose interferon‐alpha‐2b: results of a randomized phase II trial of low‐dose versus intermediate‐dose interferon‐alpha‐2b in patients with metastatic renal cell carcinoma. Cancer 2006;107(9):2254‐61. CENTRAL

Tsavaris 2000 {published data only}

Tsavaris N, Skarlos D, Bacoyiannis C, Aravantinos G, Kosmas C, Retalis G, et al. Combined treatment with low‐dose interferon plus vinblastine is associated with less toxicity than conventional interferon monotherapy in patients with metastatic renal cell carcinoma. Journal of Interferon & Cytokine Research2000; Vol. 20, issue 8:685‐90. CENTRAL

Voss 2015 {published data only}

Voss MH, Plimack ER, Rini BI, Atkins MB, Alter R, Bhatt RS, et al. The DART Study: part 1 results from the dalantercept plus axitinib dose escalation and expansion cohorts in patients with advanced renal cell carcinoma (RCC). Journal of Clinical Oncology 2015;33(Suppl 1):15. CENTRAL

Walter 2012 {published data only}

Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, et al. Multipeptide immune response to cancer vaccine IMA901 after single‐dose cyclophosphamide associates with longer patient survival. Nature Medicine 2012;18(8):1254‐61. CENTRAL

Wang 2015 {published data only}

Wang H, Feng F, Zhu M, Wang R, Wang X, Wu Y, et al. Therapeutic efficacy of dendritic cells pulsed by autologous tumor cell lysate in combination with CIK cells on advanced renal cell carcinoma. Xibao Yu Fenzi Mianyixue Zazh 2015;31(1):61‐71. CENTRAL

Weiss 1992 {published data only}

Weiss GR, Margolin KA, Aronson FR, Sznol M, Atkins MB, Dutcher JP, et al. A randomized phase II trial of continuous infusion interleukin‐2 or bolus injection interleukin‐2 plus lymphokine‐activated killer cells for advanced renal cell carcinoma. Journal of Clinical Oncology1992; Vol. 10, issue 2:275‐81. CENTRAL

Witte 1995 {published data only}

Witte RS, Leong T, Emstoff MS, Krigel RL, Oken MM, Harris J, et al. A phase II study of interleukin‐2 with and without beta‐interferon in the treatment of advanced renal cell carcinoma. Investigational New Drugs 1995;13(3):241‐7. CENTRAL

Wood 2008 {published data only}

Wood C, Srivastava P, Bukowski R, Lacombe L, Gorelov AI, Gorelov S, et al. An adjuvant autologous therapeutic vaccine (HSPPC‐96; vitespen) versus observation alone for patients at high risk of recurrence after nephrectomy for renal cell carcinoma: a multicentre, open‐label, randomised phase III trial. Lancet 2008;372(9633):145‐54. CENTRAL

Yang 1995 {published data only}

Yang JC, Topalian SL, Schwartzentruber DJ, Parkinson DR, Marincola FM, Weber JS, et al. The use of polyethylene glycol‐modified interleukin‐2 (PEG‐IL‐2) in the treatment of patients with metastatic renal cell carcinoma and melanoma. A phase I study and a randomized prospective study comparing IL‐2 alone versus IL‐2 combined with PEG‐IL‐2. Cancer 1995;76(4):687‐94. CENTRAL

Yang 2003 {published data only}

Yang JC, Sherry RM, Steinberg SM, Topalian SL, Schwartzentruber DJ, Hwu P, et al. Randomized study of high‐dose and low‐dose interleukin‐2 in patients with metastatic renal cancer. Journal of Clinical Oncology 2003;21(4):3127‐32. CENTRAL

Yang 2007 {published data only}

Yang JC, Hughes M, Kammula U, Royal R, Sherry RM, Topalian SL, et al. Ipilimumab (anti‐CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. Journal of Immunotherapy 2007;30(8):825‐30. CENTRAL

Yang 2010 {published data only}

Yang S, De Souza P, Alemao E, Purvis J. Quality of life in patients with advanced renal cell carcinoma treated with temsirolimus or interferon‐alpha. British Journal of Cancer 2010;102(10):1456‐60. CENTRAL

Zhan 2012 {published data only}

Zhan HL, Gao X, Pu XY, Li W, Li ZJ, Zhou XF, et al. A randomized controlled trial of postoperative tumor lysate‐pulsed dendritic cells and cytokine‐induced killer cells immunotherapy in patients with localized and locally advanced renal cell carcinoma. Chinese Medical Journal 2012;125(21):3771‐7. CENTRAL

Zhao 2015 {published data only}

Zhao X, Zhang Z, Huang J, Yang S, Xie T, Yue D, et al. Clinical study of cytokine‐induced killer cells based immunotherapies in different stages of renal cell carcinoma. Cytotherapy 2015;17:S20‐21. CENTRAL

EudraCT2016‐002170‐13 {published data only}

AIO‐Studien‐gGmbH. NIVOSWITCH ‐ a randomized phase II study with NIVOlumab or continuation of therapy as an early SWITCH approach in patients with advanced or metastatic renal cell carcinoma (RCC) and disease control after 3 months of treatment with a tyrosine kinase inhibitor, 2016. www.clinicaltrialsregister.eu/ctr‐search/trial/2016‐002170‐13/DE (assessed 5 November 2016). CENTRAL

Figlin 2014 {published data only (unpublished sought but not used)}

Figlin RA, Wood CG. ADAPT: an ongoing international phase III randomized trial of autologous dendritic cell immunotherapy (AGS‐003) plus standard treatment in advanced renal cell carcinoma (RCC). Journal of Clinical Oncology 2014;32(Suppl 4):Abstract 449. CENTRAL

Hammers 2015 {published data only}

Hammers HJ, Plimack E, Sternberg C, McDermott DF, Larkin J, Ravaud A, Rini BI, et al. CheckMate 214: a phase III, randomized, open‐label study of nivolumab combined with ipilimumab versus sunitinib monotherapy in patients with previously untreated metastatic renal cell carcinoma. Journal of Clinical Oncology 2015;33(Suppl):Abstract TPS4578. CENTRAL

NCT00930033 {published data only}

NCT0930033. CARMENA: randomized phase III trial evaluating the importance of nephrectomy in patients presenting with metastatic renal cell carcinoma treated with sunitinib, 2009. clinicaltrials.gov/ct2/show/NCT0930033 (assessed 24 March 2016). CENTRAL

NCT01984242 {published data only}

NCT01984242. Phase 2 study of atezolizumab (an engineered anti‐PDL1 antibody) as monotherapy or in combination with avastin (bevacizumab) compared to sunitinib in patients with untreated advanced renal cell carcinoma [IMmotion150], 2013. clinicaltrials.gov/ct2/show/NCT01984242 (assessed 24 March 2016). CENTRAL

NCT02014636 {published data only}

NCT02014636. Safety and efficacy study of pazopanib and MK 3475 in advanced renal cell carcinoma (RCC), 2013. clinicaltrials.gov/ct2/show/NCT02014636 (assessed 24 March 2016). CENTRAL

NCT02089685 {published data only}

NCT02089685. Safety and tolerability of pembrolizumab (MK‐3475) + pegylated interferon alfa‐2b and pembrolizumab + ipilimumab in participants with advanced melanoma or renal cell carcinoma (MK‐3475‐029/KEYNOTE‐29), 2014. clinicaltrials.gov/ct2/show/NCT02089685 (assessed 24 March 2016). CENTRAL

NCT02210117 {published data only}

NCT02210117. Nivolumab vs nivolumab + bevacizumab vs nivolumab + ipilimumab in metastatic renal cell carcinoma (mRCC), 2014. clinicaltrials.gov/ct2/show/NCT02210117 (assessed 24 March 2016). CENTRAL

NCT02420821 {published data only (unpublished sought but not used)}

NCT02420821. A study of atezolizumab in combination with bevacizumab versus sunitinib in participants with untreated advanced renal cell carcinoma, 2015. clinicaltrials.gov/ct2/show/NCT02420821 (assessed 24 March 2016). CENTRAL

NCT02432846 {published data only}

NCT02432846. Intratumoral vaccination with intuvax pre‐nephrectomy followed by sunitinib post‐nephrectomy vs sunitinib post‐nephrectomy in newly diagnosed metastatic renal cell carcinoma (mRCC) (MERECA), 2015. clinicaltrials.gov/ct2/show/NCT02432846 (assessed 24 March 2016). CENTRAL

NCT02684006 {published data only}

NCT02684006. JAVELIN RENAL 101 ‐ a phase 3, multinational, randomized, open‐label, parallel‐arm study of avelumab (MSB0010718C) in combination with axitinib (Inlyta(Registered)) versus sunitinib (Sutent(Registered)) monotherapy in the first‐line treatment of patients with advanced renal cell carcinoma, 2016. clinicaltrials.gov/ct2/show/NCT02684006 (assessed 01 May 2016). CENTRAL

NCT02781506 {published data only}

NCT02781506. Nivolumab and stereotactic ablative radiation therapy versus nivolumab alone for metastatic renal cancer, 2016. clinicaltrials.gov/ct2/show/NCT02781506 (assessed 05 November 2016). CENTRAL

NCT02853331 {published data only}

NCT02853331. KEYNOTE‐426 ‐ a study to evaluate the efficacy and safety of pembrolizumab (MK‐3475) in combination with axitinib versus sunitinib monotherapy in participants with renal cell carcinoma, 2016. clinicaltrials.gov/ct2/show/NCT02853331 (assessed 05 November 2016). CENTRAL

Abe 2013

Abe H, Kamai T. Recent advances in the treatment of metastatic renal cell carcinoma. International Journal of Urology 2013;20(10):944‐55. [DOI: 10.1111/iju.12187]

Albiges 2015

Albiges L, Choueiri T, Escudier B, Galsky M, George D, Hofmann F, et al. A systematic review of sequencing and combinations of systemic therapy in metastatic renal cancer. European Urology 2015;67(1):100‐10. [DOI: 10.1016/j.eururo.2014.04.006]

Armstrong 2015

Armstrong AJ, Broderick S, Eisen T, Stadler WM, Jones RJ, Garcia JA, et al. Final clinical results of a randomized phase II international trial of everolimus vs. sunitinib in patients with metastatic non‐clear cell renal cell carcinoma (ASPEN). Journal of Clinical Oncology 2015;15(Suppl 1):Abstract 4507.

Athar 2008

Athar U, Gentile TC. Treatment options for metastatic renal cell carcinoma: a review. Canadian Journal of Urology 2008;15(2):3954‐66.

Bedke 2014

Bedke J, Gouttefangeas C, Singh‐Jasuja H, Stevanović S, Behnes C‐L, Stenzl A. Targeted therapy in renal cell carcinoma: moving from molecular agents to specific immunotherapy. World Journal of Urology 2014;32(1):31‐8. [DOI: 10.1007/s00345‐013‐1033‐3]

Bedke 2015

Bedke J, Kruck S, Gakis G, Stenzl A, Goebell PJ. Checkpoint modulation ‐ a new way to direct the immune system against renal cell carcinoma. Human Vaccines and Immunotherapeutics 2015;11(5):1201‐8. [DOI: 10.1080/21645515.2015.1016657]

Brookman‐May 2011

Brookman‐May S, Burger M, Wieland WF, Rössler W, May M, Denzinger S. Vaccination therapy in renal cell carcinoma: current position and future options in metastatic and localized disease. Expert Reviews of Vaccines 2011;10(6):837‐52. [DOI: 10.1586/erv.11.64]

Cella 1997

Cella D. Manual of the Functional Assessment of Chronic Illness Therapy (FACIT Scales), Version 4. Evanston (IL): Northwestern Healthcare & Northwestern University, Center on Outcomes, Research and Education, 1997.

Ciccarese 2016

Ciccarese C, Alfieri S, Santoni M, Santini D, Brunelli M, Bergamini C, et al. New toxicity profile for novel immunotherapy agents: focus on immune‐checkpoint inhibitors. Expert Opinion on Drug Metabolism and Toxicology 2016;12(1):57‐75. [DOI: 10.1517/17425255.2016.1120287]

Combe 2015

Combe P, de Guillebon E, Thibault C, Granier C, Tartour E, Oudard S. Trial Watch: therapeutic vaccines in metastatic renal cell carcinoma. Oncoimmunology 2015;4(5):e1001236.

Coppin 2008

Coppin C. Immunotherapy for renal cell cancer in the era of targeted therapy. Expert Review of Anticancer Therapy 2008;8(6):907‐19. [DOI: 10.1586/14737140.8.6.907]

Desrichard 2016

Desrichard A, Snyder A, Chan TA. Cancer Neoantigens and Applications for Immunotherapy. Clinical Cancer Research 2016;22(4):807‐12. [DOI: 10.1158/1078‐0432.CCR‐14‐3175]

Draube 2011

Draube A, Klein‐González N, Mattheus S, Brillant C, Hellmich M, Engert A, et al. Dendritic cell based tumor vaccination in prostate and renal cell cancer: a systematic review and meta‐analysis. PLoS One 2011;6(4):e18801. [PUBMED: 21533099]

Dutcher 2013

Dutcher JP. Recent developments in the treatment of renal cell carcinoma. Therapeutic Advances in Urology 2013;5(6):338‐53. [PUBMED: 24294292]

Eisenhauer 2009

Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). European Journal of Cancer 2009;45(2):228‐47.

Escudier 2014

Escudier B, Porta C, Schmidinger M, Algaba F, Patard JJ, Khoo V, et al. Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow‐up. Annals of Oncology 2014;25(Suppl 3):iii49‐56.

Ferlay 2013

Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, et al. GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11. Lyon, France: International Agency for Research on Cancer; 2013. globocan.iarc.fr (accessed 12 February 2015).

Figlin 2015

Figlin RA. Personalized immunotherapy (AGS‐003) when combined with sunitinib for the treatment of metastatic renal cell carcinoma. Expert Opinion on Biological Therapy 2015;15(8):1241‐8. [DOI: 10.1517/14712598.2015.1063610]

German Guideline Programme in Oncology 2015

The German Guideline Programme in Oncology (German Cancer Society, German Cancer Aid, AWMF). S3‐Guideline on Diagnostics, Therapy and Follow‐up of renal cell carcinoma, long version 1.0, AWMF registration 043/017OL, 2015 [Diagnostik, Therapie und Nachsorge des Nierenzellkarzinoms, Langversion 1.0, AWMF Registernummer: 043/017OL]. leitlinienprogramm‐onkologie.de/Leitlinien.7.0.html (assessed 31 May 2016).

Ghasemzadeh 2016

Ghasemzadeh A, Bivalacqua TJ, Hahn NM, Drake CG. New strategies in bladder cancer: a second coming for immunotherapy. Clinical Cancer Research 2016;22(4):793‐801. [DOI: 10.1158/1078‐0432.CCR‐15‐1135]

GRADEpro GDT [Computer program]

McMaster University. GRADEpro GDT. Version accessed 17 April 2015. McMaster University, 2014.

Grünwald 2015

Grünwald V. T‐cell checkpoint inhibitors in metastatic renal cell carcinoma. Current Opinion in Urology 2015;25(5):411‐5. [DOI: 10.1097/MOU.0000000000000199]

Gupta 2008

Gupta K, Miller JD, Li JZ, Russell MW, Charbonneau C. Epidemiologic and socioeconomic burden of metastatic renal cell carcinoma (mRCC): a literature review. Cancer Treatment Reviews 2008;34(3):193‐205. [DOI: 10.1016/j.ctrv.2007.12.001]

Guyatt 2011a

Guyatt G, Oxman AD, Akl E, Kunz R, Vist G, Brozek J, et al. GRADE guidelines: 1. Introduction ‐ GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383‐94. [DOI: 10.1016/j.jclinepi.2010.04.026]

Guyatt 2011b

Guyatt GH, Oxman AD, Vist G, Kunz R, Brozek J, Alonso‐Coello P, et al. GRADE guidelines: 4. Rating the quality of evidence ‐ study limitations (risk of bias). Journal of Clinical Epidemiology 2011;64(4):407‐15. [DOI: 10.1016/j.jclinepi.2010.07.017]

Guyatt 2011c

Guyatt GH, Oxman AD, Montori V, Vist G, Kunz R, Brozek J, et al. GRADE guidelines: 5. Rating the quality of evidence ‐ publication bias. Journal of Clinical Epidemiology 2011;64(12):1277‐82. [DOI: 10.1016/j.jclinepi.2010.01.011]

Guyatt 2011d

Guyatt GH, Oxman AD, Kunz R, Brozek J, Alonso‐Coello P, Rind D, et al. GRADE guidelines 6. Rating the quality of evidence ‐ imprecision. Journal of Clinical Epidemiology 2011;64(12):1283‐93. [DOI: 10.1016.j.jclinepi.2011.01.012]

Guyatt 2011e

Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines: 7. Rating the quality of evidence ‐ inconsistency. Journal of Clinical Epidemiology 2011;64(12):1294‐302. [DOI: 10.1016/j.jclinepi.2011.03.017]

Guyatt 2011f

Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines: 8. Rating the quality of evidence ‐ indirectness. Journal of Clinical Epidemiology 2011;64(12):1303‐10. [DOI: 10.1016.j.jclinepi.2011.04.014]

Heng 2009

Heng DYC, Xie W, Regan MM, Warren MA, Golshayan AR, Sahi C, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor‐targeted agents: results from a large, multicenter study. Journal of Clinical Oncology 2009;27(34):5794‐9.

Heng 2013

Heng DY, Xie W, Regan MM, Harshman LC, Bjarnason GA, Vaishampayan UN, et al. External validation and comparison with other models of the International Metastatic Renal‐Cell Carcinoma Database Consortium prognostic model: a population‐based study. Lancet Oncology 2013;14(2):141‐8. [DOI: 10.1016/S1470‐2045(12)70559‐4]

Higgins 2002

Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta‐analysis. Statistics in Medicine 2002;21(11):1539‐58. [DOI: 10.1002/sim.1186]

Higgins 2003

Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ (Clinical Research Ed.) 2003;327(7414):557‐60. [DOI: 10.1136/bmj.327.7414.557]

Higgins 2011a

Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011b

Higgins JPT, Altman DG, Sterne JAC. Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011c

Higgins JPT, Deeks JJ, Altman DG. Chapter 16: Special topics in statistics. In: Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Howlader 2015

Howlader N, Noone AM, Krapcho M, Garshell J, Miller D, Altekruse SF, et al (editors). SEER cancer statistics review, 1975‐2012, National Cancer Institute. Bethesda, MD. seer.cancer.gov/csr/1975_2012/, based on November 2014 SEER data submission, posted to the SEER website on April 2015 (accessed 30 May 2016).

Johannsen 2007

Johannsen M, Brinkmann OA, Bergmann L, Heinzer H, Steiner T, Ringsdorf M, et al. The role of cytokine therapy in metastatic renal cell cancer. European Urology Supplements 2007;6:658‐64.

Jonasch 2014

Jonasch E, Gao J, Rathmell WK. Renal cell carcinoma. BMJ 2014;349:g4797. [DOI: 10.1136/bmj.g4797]

Le 2015

Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD‐1 Blockade in tumors with mismatch‐repair deficiency. New England Journal Medicine 2015;372(26):2509‐20. [DOI: 10.1056/NEJMoa1500596]

Lee 2016

Lee CH, Motzer RJ. Immune checkpoint therapy in renal cell carcinoma. Cancer Journal 2016;22(2):92‐5. [DOI: 10.1097/PPO.0000000000000177]

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville J. Chapter 6: Searching for studies. In: Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Liberati 2009

Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, et al. The PRISMA statement for reporting systematic reviews and meta‐analyses of studies that evaluate health care interventions: explanation and elaboration. PLoS Medicine 2009;6(7):e1000100. [DOI: 10.1371/journal.pmed.1000100]

Ljungberg 2011

Ljungberg B, Campbell SC, Cho HY, Jacqmin D, Lee JE, Weikert S, et al. The epidemiology of renal cell carcinoma. European Urology 2011;60:615‐21. [DOI: 10.1016/j.eururo.2011.06.049]

Ljungberg 2015

Ljungberg B, Bensalah K, Canfield S, Dabestani S, Hofmann F, Hora M, et al. EAU guidelines on renal cell carcinoma: 2014 update. European Urology 2015;67(5):913‐24.

Massari 2015

Massari F, Santoni M, Ciccarese C, Santini D, Alfieri S, Martignoni G, et al. PD‐1 blockade therapy in renal cell carcinoma: current studies and future promises. Cancer Treatment Reviews 2015;41(2):114‐21. [DOI: 10.1016/j.ctrv.2014.12.013]

McDermott 2015a

McDermott DF, Cheng SC, Signoretti S, Margolin KA, Clark JI, Sosman JA, et al. The high‐dose aldesleukin "select" trial: a trial to prospectively validate predictive models of response to treatment in patients with metastatic renal cell carcinoma. Clinical Cancer Research 2015;21(3):561‐8. [DOI: 10.1158/1078‐0432.CCR‐14‐1520]

McDermott 2015b

McDermott DF, Choueiri TK, Puzanov I, Hodi S, Drake CG, Brahmer JR, et al. Survival, durable response, and long‐term safety in patients with previously treated advanced renal cell carcinoma receiving nivolumab. Journal of Clinical Oncology 2015;33(18):2013‐20.

Michael 2003

Michael A, Pandha HS. Renal‐cell carcinoma: tumour markers, T‐cell epitopes, and potential for new therapies. Lancet Oncology 2003;4(4):215‐23. [PUBMED: 12681265]

Motzer 1996

Motzer RJ, Bander NH, Nanus DM. Renal‐cell carcinoma. New England Journal of Medicine 1996;335(12):865‐75. [DOI: 10.1056/NEJM199609193351207]

Motzer 2002

Motzer RJ, Bacik J, Murphy BA, Russo P, Mazumdar M. Interferon‐alfa as a comparative treatment for clinical trials of new therapies against renal cell carcinoma. Journal of Clinical Oncology 2002;20(1):289‐96.

Motzer 2016

Motzer RJ, Sharma P, McDermott DF, George S, Hammers HJ, Srinivas S, et al. CheckMate 025 phase III trial: Outcomes by key baseline factors and prior therapy for nivolumab (NIVO) versus everolimus (EVE) in advanced renal cell carcinoma (RCC). Journal Clinical Oncology 2016;34(Suppl 2S):Abstract 498.

Parmar 1998

Parmar MKB, Torri V, Stewart L. Extracting summary statistics to perform meta‐analyses of the published literature for survival endpoints. Statistics in Medicine 1998;17(24):2815‐34.

Pickard 2007

Pickard AS, Neary MP, Cella D. Estimation of minimally important differences in EQ‐5D utility and VAS scores in cancer. Health and Quality of Life Outcomes 2007;5:70. [DOI: 10.1186/1477‐7525‐5‐70]

Postow 2015

Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. Journal of Clinical Oncology 2015;33(17):1974‐82.

Powles 2016

Powles T, Staehler M, Ljungberg B, Bensalah K, Canfield SE, Dabestani S, et al. Updated EAU guidelines for clear cell renal cancer patients who fail VEGF targeted therapy. European Urology 2016;69(1):4‐6. [DOI: 10.1016/j.eururo.2015.10.017]

Rayman 2004

Rayman P, Wesa AK, Richmond AL, Das T, Biswas K, Raval G, et al. Effect of renal cell carcinomas on the development of type 1 T‐cell responses. Clinical Cancer Research 2004;10(18 Pt 2):6360S‐6S. [PUBMED: 15448031]

RevMan 2014 [Computer program]

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

Ringash 2007

Ringash J, O'Sullivan B, Bezjak A, Redelmeier DA. Interpreting clinically significant changes in patient‐reported outcomes. Cancer 2007;110(1):196‐202.

Rothermundt 2015

Rothermundt C, Bailey A, Cerbone L, Eisen T, Escudier B, Gillessen S, et al. Algorithms in the first‐line treatment of metastatic clear cell renal cell carcinoma ‐ analysis using diagnostic nodes. Oncologist 2015;20(9):1028‐35. [DOI: 10.1634/theoncologist.2015‐0145]

Schünemann 2011

Schünemann HJ, Oxman AD, Higgins JPT, Vist GE, Glasziou P, Guyatt GH. Chapter 11: Presenting results and 'Summary of findings' tables. In: Higgins JPT, Green S, editor(s), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Siegel 2014

Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA: a Cancer Journal for Clinicians 2014;64(1):9‐29 (erratum in: CA: a Cancer Journal for Clinicians 2014;64(5):364). [DOI: 10.3322/caac.21208]

Sobin 2009

Sobin LH, Gospodariwicz MK, Wittekind C. TNM Classification of Malignant Tumours. UICC International Union Against Cancer. 7th Edition. Hoboken (NJ): Wiley‐Blackwell, 2009:255‐257.

Stadler 2014

Stadler WM. Maturing of renal cancer therapeutics. Journal of Clinical Oncology 2014;32(8):722‐4.

Takyar 2016

Takyar S, Diaz J, Sehgal M, Sapunar F, Pandha H. First‐line therapy for treatment‐naive patients with advanced/metastatic renal cell carcinoma: a systematic review of published randomized controlled trials. Anticancer Drugs 2016;27(5):383‐97. [DOI: 10.1097/CAD.0000000000000335]

Thompson 2007

Thompson RH, Dong H, Lohse CM, Leibovich BC, Blute ML, Cheville JC, et al. PD‐1 is expressed by tumor‐infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clinical Cancer Research 2007;13(6):1757‐61. [PUBMED: 17363529]

Tierney 2007

Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR. Practical methods for incorporating summary time‐to‐event data into meta‐analysis. Trials 2007;8:16. [PUBMED: 17555582]

Unverzagt 2015

Unverzagt S, Moldenhauer I, Coppin C, Greco F, Seliger B. Immunotherapy for metastatic renal cell carcinoma. Cochrane Database of Systematic Reviews 2015, Issue 4. [DOI: 10.1002/14651858.CD011673]

Weber 2015

Weber JS, Yang JC, Atkins MB, Disis ML. Toxicities of immunotherapy for the practitioner. Journal of Clinical Oncology 2015;33(18):2092‐9. [DOI: 10.1200/JCO.2014.60.0379]

Williamson 2002

Williamson PR, Smith CT, Hutton JL, Marson AG. Aggregate data meta‐analysis with time‐to‐event outcomes. Statistics in Medicine 2002;21(22):3337‐51. [PUBMED: 15685717]

Wittekind 2012

Wittekind BJ, Compton CC, Sobin LH, editor(s). A commentary on uniform use. UICC International Union Against Cancer. 4th Edition. New York (NY): Wiley‐Liss, 2001:106.

Xu 2015

Xu KY, Wu S. Update on the treatment of metastatic clear cell and non‐clear cell renal cell carcinoma. Biomarker Research 2015;3:5. [DOI: 10.1186/s40364‐015‐0030‐7]

Yang 2016

Yang JC. The immunotherapy roadmap. Clinical Cancer Research 2016;22(2):275‐6. [DOI: 10.1158/1078‐0432.CCR‐15‐2144]

Zibelman 2015

Zibelman M, Barth P, Handorf E, Smaldone MC, Kutikov A, Uzzo RG, et al. A review of international clinical trials in renal cell carcinoma: a status report from the ClinicalTrials.gov website. Clinical Genitourinary Cancer 2015;13(2):142‐9. [PUBMED: 25450029]

Zisman 2002

Zisman A, Pantuck AJ, Wieder J, Chao DH, Dorey F, Said JW, et al. Risk group assessment and clinical outcome algorithm to predict the natural history of patients with surgically resected renal cell carcinoma. Journal of Clinical Oncology 2002;20(23):4559‐66. [PUBMED: 12454113]

References to other published versions of this review

Coppin 2000

Coppin C, Porzsolt F, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. Cochrane Database of Systematic Reviews 2000, Issue 3. [DOI: 10.1002/14651858.CD001425]

Coppin 2005

Coppin C, Porzsolt F, Awa A, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. Cochrane Database of Systematic Reviews 2005, Issue 1. [DOI: 10.1002/14651858.CD001425.pub2]

Coppin 2007

Coppin C, Porzsolt F, Autenrieth M, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. Cochrane Database of Systematic Reviews 2007, Issue 1. [DOI: 10.1002/14651858.CD001425.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Amato 2010

Methods

Study design: 2‐arm, parallel‐group, double‐blind, placebo‐controlled RCT.

Study dates: recruitment from October 2006 to March 2008, follow‐up to March 2009, range of follow‐up 12 to 29 months.

Setting: multicentre, international, phase III.

Countries: France, Germany, Israel, Poland, Romania, Russia, Spain, UK, Ukraine, US.

Participants

Inclusion criteria: people with histologically confirmed advanced or metastatic clear‐cell RCC with prior nephrectomy who required first‐line treatment, aged ≥ 18 years, measurable disease, KPS ≥ 80%, MSKCC 0‐2, life expectancy > 12 weeks.

Exclusion criteria: cerebral metastases, prior exposure to MVA‐5T4, known allergy to vaccinia vaccinations or egg proteins, pregnancy.

Sample size:732.

Age (years, median with range): group 1: 58 (18 to 86); group 0: 58 (24 to 85).

Sex (M/F, %): group 1: 69.6/30.4; group 0: 65.1/34.9.

Prognostic factors:

  1. performance status (KPS 80/90/100, %): 30/44/26;

  2. prior nephrectomy (%): 99.9;

  3. prior systemic therapies (%): 0;

  4. risk prognosis favourable/intermediate/poor (Motzer 2002) (%): 58/42/1.

Interventions

Group 1 (n = 365): MVA‐5T4 (modified vaccinia Ankara encoding the tumour antigen 5T4).

Group 0 (n = 367): placebo.

Both as IM injection into the deltoid muscle at weeks 1, 3, 6, 9, 13, 17, 21, 25, 33, 41, 49, 57, 65.

Cointerventions (standard‐of‐care according to local practice):

  1. SC low‐dose IL‐2 (n = 170): initial dose of 250,000 U/kg/dose with an upper limit of 22 MU/dose days 1 to 5 of week 1 + 125,000 U/kg/dose with an upper limit of 11 MU/dose days 1 to 5 of weeks 2 to 6 every 6 weeks;

  2. IFN‐α (n = 377): SC injection 3 times/week on days 1, 3 and 5 of each week at a dose level that reflected local practice between 9 million IU and 18 million IU every 1 week;

  3. sunitinib (n = 185): 50 mg oral dose taken days 1 to 28 every 6 weeks.

Outcomes

OS (primary outcome)

How measured: active follow‐up.

Time points measured: censored to March 2009.

Time points reported: Kaplan‐Meier survival curves over up to 30 months, HR (with 95% CI), median.

Subgroups: risk prognosis and standard of care (no comparison group 1 vs group 0 reported).

AEs, grade ≥ 3 (primary safety outcome)

How measured: NCI‐CTC (treatment‐emergent SAE all, grade 3, 4, 5).

Time points measured and reported: not reported.

Subgroups: not reported.

QoL not evaluated.

PFS (secondary outcome)

How measured: not reported.

Time points measured: week 26.

Time points reported: not reported.

Subgroups: not reported.

Tumour remission (secondary outcome)

How measured: complete response, partial response, stable disease.

Time points measured and reported: week 26.

Subgroups: not reported.

Funding sources

Sponsored by Oxford BioMedica.

Declarations of interest

RH, WHS, SN: named inventors on several Oxford BioMedica patents. REH: minor consultancy role for Oxford BioMedica.

Notes

Trial registration: NCT00397345, at the recommendation of the data safety monitoring board, the sponsor terminated the administration of MVA‐5T4/placebo to participants in July 2008 due to little or no prospect of demonstrating a significant survival benefit.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Stratification for standard of care, severity of disease and geographic location.

Allocation concealment (selection bias)

Unclear risk

No information provided.

Blinding of participants and personnel (performance bias)
Subjective outcomes

Low risk

No performance bias assumed due to blinding of participants and study personnel.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

No performance bias assumed due to blinding of participants and study personnel.

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

No detection bias on OS assumed due to blinding of participants, study personnel and outcome assessors.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

No detection bias on OS assumed due to blinding of participants, study personnel and outcome assessors.

Incomplete outcome data (attrition bias)
(OS and PFS)

Low risk

No attrition bias on OS and PFS assumed, analysis with active follow‐up on survival data after termination of drug administration with no further description, no differences in censoring detected.

Incomplete outcome data (attrition bias)
(safety)

Low risk

No attrition bias on safety outcomes assumed, assessment on the basis of all participants as randomized.

Incomplete outcome data (attrition bias)
(tumour remission)

Low risk

No attrition bias on tumour remission assumed, assessment on the basis of all participants as randomized.

Incomplete outcome data (attrition bias)
(QoL)

Unclear risk

Not evaluated.

Selective reporting (reporting bias)

High risk

PFS was mentioned as secondary outcome, but no information provided.

Other bias

High risk

Safety monitoring board recommended stopping in July 2008 because there was little prospect of demonstrating a significant benefit in OS, second‐line therapies in 32% of placebo and 29% of MVA‐5T4 patients.

Escudier 2007

Methods

Study design: 2‐arm, parallel‐group, double‐blind, placebo‐controlled RCT.

Study dates: randomization from June 2004 to October 2005, clinical cutoff: September 2006, median follow‐up of 13.3 months (0 to 25.6) (clinical cutoff), median follow‐up for OS: group 1: 21, group 0: 23 months to September 2008.

Setting: multicentre, international, phase III.

Countries: Europe (Belgium, Czech Republic, Finland, France, Germany, Hungary, Israel, Italy, Netherlands, Norway, Poland, Russia, Spain, Switzerland, UK), Asia (Russia, Singapore, Taiwan), Australia.

Participants

Inclusion criteria: people with measurable or non‐measurable, predominantly clear‐cell mRCC with no prior systemic therapy, prior total or partial nephrectomy, KPS ≥ 70%; aged ≥ 18 years; normal hepatic, haematopoietic and renal function, and only minimal proteinuria.

Exclusion criteria: prior systemic treatment for mRCC, recent major surgical procedures, evidence of brain metastases, ongoing full‐dose oral or parenteral anticoagulant or antiplatelet aggregation treatment, uncontrolled hypertension on medication, clinically significant cardiovascular disease, chronic corticosteroid treatment.

Sample size:649.

Age (years, median with range): group 1: 61 (30 to 82); group 0: 60 (18 to 81).

Sex (M/F, %): group 1: 68/32; group 0: 73/27.

Prognostic factors:

  1. performance status (KPS 70/80/90/100, %): 6/17/36/41;

  2. prior nephrectomy (%): 100 (inclusion criterion);

  3. prior systemic therapies (%): 0 (inclusion criterion);

  4. risk prognosis (MSKCC) risk score poor/intermediate/high (%): 8/56/28.

Interventions

Group 1 (n = 322): IFN‐α + placebo

IFN α‐2a (Hoffmann‐La Roche Ltd, Basel, Switzerland) 9 MIU 3 times/week SC for maximum 52 weeks or until disease progression, unacceptable toxicity or withdrawal of consent, an initial dose < 9 MIU permitted if the recommended dose was reached within the first 2 weeks of treatment, dose reduction; to 6 MIU or 3 MIU to manage AE attributable to IFN‐α.

Placebo: every 2 weeks until disease progression, unacceptable toxicity or withdrawal of consent.

Group 0 (n = 327): IFN‐α + bevacizumab

IFN α‐2a (Hoffmann‐La Roche Ltd, Basel, Switzerland) 9 MIU 3 times/week SC for maximum 52 weeks or until disease progression, unacceptable toxicity or withdrawal of consent, an initial dose < 9 MIU permitted if the recommended dose was reached within the first 2 weeks of treatment, dose reduction; to 6 MIU or 3 MIU to manage AE attributable to IFN‐α.

Bevacizumab (Hoffmann‐La Roche Ltd, Basel, Switzerland) 10 mg/kg IV every 2 weeks until disease progression, unacceptable toxicity or withdrawal of consent, no dose reduction permitted.

Cointerventions (standard‐of‐care according to local practice): not reported.

Outcomes

OS (primary outcome)

How measured: investigator‐assessed, time between the date of randomization and death due to any cause, censoring on the day of last follow‐up or the last day of study administration if no follow‐up was done.

Time points measured: during treatment and follow‐up before unblinding with cross‐over and second‐line therapies.

Time points reported: Kaplan‐Meier curves over up to 24 months, median OS, number of deaths at data cutoff before cross‐over and second‐line therapies.

Subgroups: MSKCC score.

AEs, grade ≥ 3 (secondary outcome)

How measured: investigator‐assessed, ongoing documentation of AEs (CTCAE v.3.0), physical examination, electrocardiography, urinalysis, measurement of blood pressure.

Time points measured: at each visit, weekly monitoring in participants who developed ≥ grade 3 hypertension, 24‐hour urine collection if protein was observed with a dipstick analysis.

Time points reported: frequency of participants with AEs, SAEs (safety population), AEs with grade ≥ 3, most commonly reported AEs with grade ≥ 3 up to 28 days after the last dose, deaths due to AEs.

Subgroups: not reported.

QoL not evaluated

PFS (secondary outcome)

How measured: time between randomization and first documented disease progression or death due to any cause, investigator‐assessment and independent review committee.

Time points measured: every 8 weeks up to week 32 and every 12 weeks thereafter until disease progression.

Time points reported: Kaplan‐Meier survival curves for up to 24 months, median PFS.

Subgroups: age, sex, MSKCC score, baseline VEGF, number of metastatic sites.

Tumour remission (secondary outcome)

How measured: assessment by the investigator with RECIST, non‐measurable lesions were used to define complete response and disease progression only.

Time points measured: every 8 weeks up to week 32, every 12 weeks thereafter until disease progression, responses had to be confirmed by a second assessment ≥ 4 weeks after the first response was recorded.

Time points reported: best tumour response for participants with measurable disease.

Subgroups: not reported.

Funding sources

Hoffmann‐La Roche Ltd.

Declarations of interest

BE: consulted for and received honoraria from Roche, Bayer, Wyeth, Pfizer, Inate and Antigenics. SB: consulted for Roche, Pfizer, Wyeth and Bayer. AR: acted as an adviser for Bayer, Pfizer, GSK, Novartis and Wyeth. NM: employee of and owns stock of Roche. BM: received honoraria and research funding from Roche.

Notes

BO17705E, NCT00738530 (registered October 2008), preplanned interim analysis with significant benefit in OS, unblinding, data and safety monitoring board recommended cross‐over of participants from the placebo to the bevacizumab group, differences in availability of new second‐line therapies in countries might confound OS results.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

No selection bias assumed, the randomization list bases on block design procedure, stratified by country and MSKCC risk group.

Allocation concealment (selection bias)

Low risk

No selection bias assumed, central allocation by an interactive voice recognition system, list kept in a secure location, not available to any person directly involved in the study.

Blinding of participants and personnel (performance bias)
Subjective outcomes

Low risk

No performance bias on subjective outcomes assumed, blinding of participants and study personnel with same route and timing as bevacizumab until preplanned interim analysis and data cutoff.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

No performance bias on objective outcomes assumed, blinding of participants and study personnel with same route and timing as bevacizumab until preplanned interim analysis and data cutoff.

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

No performance bias on subjective outcomes assumed, blinding of participants and study personnel with same route and timing as bevacizumab until preplanned interim analysis and data cutoff.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

No detection bias on OS assumed due to blinding of participants, study personnel and outcome assessors.

Incomplete outcome data (attrition bias)
(OS and PFS)

Low risk

No attrition bias on OS and PFS assumed due similar censoring between treatment groups, 251 deaths and 505 progression events from 649 participants had occurred at the time of data cutoff.

Incomplete outcome data (attrition bias)
(safety)

Low risk

No attrition bias on safety outcomes assumed, all participants who included at least 1 dose of bevacizumab were compared to participants who received no bevacizumab.

Incomplete outcome data (attrition bias)
(tumour remission)

Low risk

No attrition bias on tumour remission assumed, all participants with measurable disease at baseline were included.

Incomplete outcome data (attrition bias)
(QoL)

Unclear risk

Not evaluated.

Selective reporting (reporting bias)

Low risk

No reporting bias on tumour remission assumed, no reporting bias assumed, all preplanned outcomes reported.

Other bias

High risk

Cross‐over of 13 (4%) participants from group 0 to group 1, 49 (15%) participants in group 1 and 64 (20%) participants in group 0 received second‐line therapy with tyrosine kinase inhibitors.

Hudes 2007

Methods

Study design: 3‐arm, parallel‐group, open‐label RCT.

Study dates: recruitment from July 2003 to April 2005, follow‐up until second interim analysis after 446 deaths (June 2006), range of follow‐up: 14 to 36 months.

Setting: multicentre, international, phase III.

Countries: Global Advanced Renal Cell Carcinoma (ARCC) Trial including countries of all continents, specifically: Argentina, Australia, Canada, Czech Republic, Germany, Greece, Hungary, Italy, Latvia, Lithuania, Netherlands, Poland, Russia, Serbia, Montenegro, Slovakia, South Africa, Spain, Sweden, Taiwan, Turkey, Ukraine, UK, US.

Participants

Inclusion criteria: histologically confirmed advanced RCC (stage IV or recurrent disease), KPS ≥ 60, no previous systemic therapy, measurable disease, adequate bone marrow, renal and hepatic functions (neutrophil count > 1500 cells/mm3, platelet count > 100,000 cells/mm3, haemoglobin count > 8 g/dL. People with a history of brain metastases if their condition was neurologically stable and they did not require corticosteroids after surgical resection or radiotherapy.

Exclusion criteria: serum creatinine level ≤ 1.5 times ULN; aspartate aminotransferase level ≤ 3 times ULN (≤ 5 times if liver metastases present); total bilirubin level ≤ 1.5 times ULN; fasting level of total cholesterol ≤ 350 mg/dL, triglyceride level ≤ 400 mg/dL.

Sample size:626.

Age (years, median with range): group 1: 59 (32 to 82); group 1a: 60 (23 to 86); group 0: 58 (32 to 81).

Sex (M/F, %): group 1: 69/31; group 1a: 71/28; group 0: 66/33.

Prognostic factors:

  1. performance status (KPS ≤ 70%/>70%): 17/83;

  2. prior nephrectomy (n, %): 67;

  3. prior systemic therapies (n, %): not reported;

  4. risk prognosis: MSKCC risk classification: intermediate risk (1 or 2 of 5 factors)/poor risk (3 or 4 or 5 of 5 factors (n, %)): 28/72.

Interventions

Group 1 (n = 210): IFN‐α + temsirolimus

Temsirolimus (Wyeth Research, 15 mg IV weekly, 30‐minute infusion) + IFN‐α (Roferon‐A, Roche, starting dose 3 MU 3 times/week for week 1 and 6 MU SC 3 times/week thereafter).

Group 1a (n = 207): IFN‐α

IFN‐α starting dose of 3 MU SC 3 times/week for the first week, dose was raised to 9 MU 3 times/week for the second week and to 18 MU 3 times/week for week 3, if tolerated. Participants who were unable to tolerate 9 MU or 18 MU received the highest tolerable dose (3 MU, 4.5 MU or 6 MU).

Group 0 (n = 209): temsirolimus

Temsirolimus 25 mg IV weekly 30‐minute infusion.

Cointerventions for participants treated with temsirolimus (standard‐of‐care according to local practice): premedication with diphenhydramine 25 mg to 50 mg IV or a similar histamine H1 blocker given approximately 30 minutes before each weekly temsirolimus infusion as prophylaxis against an allergic reaction.

Outcomes

OS (primary outcome)

How measured: investigator assessed, time between date of randomization and date of death.

Time points measured: not reported.

Time points reported: Kaplan‐Meier survival curves over up to 30 months, median with 95% CI, HR with 95% CI.

Subgroups: prior nephrectomy, KPS (≤ 70, > 70).

AEs, grade ≥3

How measured: AEs (NCI‐CTC 3.0) occurring in ≥ 20% of participants in any group (all grades and grade 3 or 4), number of AEs, grade 3 or 4 per treatment group, any visit at which the participant reported a symptomatic NCI‐CTC (v.3) grade 3 or 4.

Time points measured: weekly or biweekly.

Time points reported: treatment period.

Subgroups: not evaluated.

QOL

How measured: EQ‐5D and EQ‐VAS questionnaire (self‐report).

Time points measured: at screening, week 12, week 32.

Time points reported: week 12, withdrawal or last recorded visit (only IFN‐α vs temsirolimus) (Yang 2010).

Subgroups: prior nephrectomy.

PFS (secondary outcome)

How measured: determined by the site investigators' assessment and a blinded assessment of imaging studies (performed by Bio‐Imaging Technologies, not shown), time between date of randomization and date of disease progression or death, whichever occurred first.

Time points measured: not reported.

Time points reported: Kaplan‐Meier survival curves over up to 30 months, median with 95% CI, HR with P value.

Subgroups: not reported.

Tumour remission (secondary outcome)

How measured: CT scans of the chest, abdomen and pelvis; radionuclide bone scan MRI or CT scan of the brain; classification into participants with stable disease or objective response (RECIST); % participants who had a confirmed objective response (complete or partial) as their best response to treatment.

Time points measured: before treatment, repeated at 8‐week intervals.

Time points reported: 24 weeks.

Subgroups: not reported.

Funding sources

Wyeth Research, Cambridge, MA, US.

Declarations of interest

GH: financial support from Pfizer and Wyeth; MC, RF, IGHS‐W, RJM: financial support from Wyeth; JD: financial support from Novartis, Chiron, Bayer, Onyx, Pfizer and Wyeth; AK: financial support from Bayer and Wyeth; DMcD: financial support from Bayer, Onyx, Genentech and Novartis. TO'T, SL and LM: full‐time employee of Wyeth Research.

Notes

Trial registration: NCT00065468, study was stopped as a result of the second predefined interim analysis.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Stratified block randomization.

Allocation concealment (selection bias)

Unclear risk

No information reported.

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Different delivery of the interventions, no placebo‐controlled trial, participants and physicians not blinded.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

No performance bias on OS assumed.

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

Kaplan‐Meier estimates of blinded assessment for PFS not shown, outcome assessors not blinded.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

No detection bias on OS assumed.

Incomplete outcome data (attrition bias)
(OS and PFS)

Low risk

No attrition bias on PFS or OS assumed due to small censoring rates.

Incomplete outcome data (attrition bias)
(safety)

Low risk

No attrition bias on safety outcomes assumed, Inclusion of all participants as treated.

Incomplete outcome data (attrition bias)
(tumour remission)

High risk

Differences in postbaseline tumour assessment (group 1: 74% vs group 1a: 80% vs group 0: 92%).

Incomplete outcome data (attrition bias)
(QoL)

High risk

High risk of attrition on QoL due to high differences in completion rates between treatment groups.

Selective reporting (reporting bias)

Low risk

No reporting bias assumed, nearly all outcomes reported (besides quality‐adjusted time without symptoms or toxicity).

Other bias

Low risk

Early stop for benefit, no other risk of bias assumed.

Motzer 2007

Methods

Study design: 2‐arm, parallel‐group, open‐label RCT.

Study dates: enrolment between August 2004 and October 2005, median duration of treatment: 6 months, range 1 month to 15 months.

Setting: inpatients, multicentre (101 centres), international, phase III.

Countries: Australia, Brazil, Canada, Europe, US.

Participants

Inclusion criteria: people with histologically confirmed clear‐cell mRCC; aged ≥ 18 years; no previous treatment with systemic therapy for RCC; measurable disease; ECOG Performance Status 0 to 1; adequate haematological, coagulation, hepatic, renal and cardiac function.

Exclusion criteria: brain metastases, uncontrolled hypertension or clinically significant cardiovascular events or disease during the preceding 12 months.

Sample size:750.

Age (years, median with range): group 1: 59 (34 to 85); group 0: 62 (27 to 87).

Sex (M/F, %): group 1: 72/28; group 0: 71/29.

Prognostic factors:

  1. performance status (ECOG Performance Status 0/1, %): 62/38;

  2. prior nephrectomy (%): 90;

  3. prior systemic therapies (%): not allowed;

  4. risk prognosis (MSKCC favourable/intermediate/poor, %): 35/56/6.4, missing data for 17 participants.

Interventions

Group 1 (n = 375): IFN‐α

IFN‐α‐2a (Roche) SC injection 3 times/week on non‐consecutive days at 3 MU per dose during first week, 6 MU per dose during second week and 9 MU per dose thereafter) until the occurrence of disease progression, unacceptable AEs or withdrawal of consent.

Group 0 (n = 375): sunitinib

Sunitinib orally 50 mg once daily in 6‐week cycles consisting of 4 weeks of treatment followed by 2 weeks without treatment.

Cointerventions (standard‐of‐care according to local practice): not specified.

Outcomes

OS (secondary endpoint)

How measured: not specified.

Time points measured: during follow‐up; off‐study every 2 months.

Time points reported: Kaplan‐Meier survival curves over up to 36 months (data from Motzer 2009), median with 95% CI, unstratified HR and stratified HR with 95% CI.

Subgroups: MSKCC criteria; previous nephrectomy; ECOG Performance Status; lactate dehydrogenase level; time since diagnosis; haemoglobin level; corrected serum calcium level; number of metastatic sites; bone, lung and liver metastases.

AEs, grade ≥ 3 (not specified)

How measured: CTCAE v.3.0.

Time points measured and reported: not reported, no summarized frequencies reported.

Subgroups: not reported.

QoL (secondary outcome)

How measured: FKSI‐15, FACT‐G, EQ‐5D, EQ‐VAS ‐ completion, if > 80% of items in FACT‐G and > 50% of items in FKSI completed.

Time points measured and reported: before randomization, on days 1 and 28 of each cycle, overall mean score after 17 weeks (Cella 2008).

Subgroups: not reported.

PFS (primary endpoint)

How measured: time from randomization to the first documentation of objective disease progression or to death from any cause, whichever occurred first with a blinded central review of radiological images.

Time points measured: during follow‐up (central review to September 2007).

Time points reported: Kaplan‐Meier survival curves over up to 14 months (Motzer 2007), median PFS with 95% CI; HR with 95% CI (central review and investigator‐assessed).

Subgroups: MSKCC criteria, previous nephrectomy, age, sex, ECOG Performance Status, lactate dehydrogenase level, time since diagnosis, haemoglobin level, corrected serum calcium level.

Tumour remission (secondary outcome)

How measured: RECIST with blinded central review of radiological images, complete and partial response, stable disease.

Time points measured: day 28 of cycles 1 through 4, and every two cycles thereafter until the end of treatment.

Time points reported: independent central review to September 2007 (interim analysis), investigator‐assessed update results (not included).

Subgroups: not reported.

Funding sources

IFN‐α‐2a (Roferon‐A, Roche) and sunitinib were provided by Pfizer.

Declarations of interest

RJM: research grants from Pfizer and Genentech, consulting fees from Wyeth and lecture fees from Bayer Pharmaceuticals; TEH: consulting and lecture fees from Pfizer, Bayer Pharmaceuticals and Onyx Pharmaceuticals; DM: consulting fees from Pfizer and Wyeth Pharmaceuticals and lecture fees from Pfizer; RMB: research grants from Pfizer, Bayer Pharmaceuticals, Genentech, Genzyme and Bristol‐Myers Squibb and consulting and lecture fees from Pfizer, Bayer Pharmaceuticals, Onyx Pharmaceuticals and Genentech; OR: consulting and lecture fees from Pfizer; SO: consulting and lecture fees from Pfizer; SN: consulting fees from Pfizer and Bayer Pharmaceuticals; RAF: research grants from Pfizer, consulting fees from Pfizer and Onyx Pharmaceuticals, and lecture fees from Pfizer and Bayer Pharmaceuticals, STK, IC, PWB, CMB: full‐time employees of Pfizer and equity owners in the company.

Notes

Registration: NCT00098657 and NCT00083889, protocol amendment (February 2006), cross‐over of 7% (n = 25) participants from IFN‐α to sunitinib.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Low risk of selection bias due to stratified randomization according to baseline levels of lactate dehydrogenase (> 1.5 vs ≤ 1.5 ULN), ECOG Performance Status (0 vs 1), and previous nephrectomy (yes vs no) with random permuted blocks of 4.

Allocation concealment (selection bias)

Unclear risk

Not reported.

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Blinding not possible due to different routes of interventions, participants and physicians were not blinded.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

No performance bias on OS assumed.

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

Blinded central review of radiological images used to assess primary endpoint and ORR, no blinded assessment of AEs and QoL reported.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

No detection bias on OS assumed.

Incomplete outcome data (attrition bias)
(OS and PFS)

Low risk

All participants were included, similar censoring.

Incomplete outcome data (attrition bias)
(safety)

Low risk

All treated participants included.

Incomplete outcome data (attrition bias)
(tumour remission)

High risk

Images of 88 (12%) participants had not been assessed by a central review at the time of interim analysis and were assessed by investigators (results not included).

Incomplete outcome data (attrition bias)
(QoL)

Low risk

Completion rates for FKSI, FACT‐G and EQ‐5D questionnaires: 95% with at least 1 postbaseline assessment and slightly lower rates in the group with IFN‐α.

Selective reporting (reporting bias)

Low risk

All preplanned outcomes from the protocol reported.

Other bias

High risk

After the interim analysis, participants in the IFN‐α group with progressive disease were allowed to cross over to the sunitinib group and 25 participants crossed over which may influence OS.

Motzer 2015a

Methods

Study design: 2‐arm, parallel‐group, open‐label RCT.

Study dates: randomization October 2012 to March 2014, data cutoff: June 2015, minimal follow‐up of 14 months.

Setting: multicentre (146 centres), international, phase III.

Countries: North America (US, Canada), Western Europe (Belgium, Denmark, Finland, France, Germany, Greece, Ireland, Italy, Norway, Poland, Romania, Spain, Sweden, UK), South America (Argentina, Brazil) and Asia (Israel, Japan).

Participants

Inclusion criteria: people with histologically confirmed advanced or mRCC with a clear‐cell component, aged ≥ 18 years, measurable disease, prior treatment with 1 or 2 antiangiogenic therapies, ≤ 3 previous systemic therapies with cytokines and cytotoxic drugs and disease progression during or > 1 treatment regimen within 6 months before study enrolment, KPS ≥ 70.

Exclusion criteria: central nervous system metastasis, previous treatment with an mTOR inhibitor, condition requiring treatment with glucocorticoids (equivalent to > 10 mg of prednisone > 10 mg daily).

Sample size:821.

Age (years, median with range): group 1: 62 (23‐88); group 0: 62 (18‐86).

Sex (M/F, %): group 1: 77/23; group 0: 74/26.

Prognostic factors:

  1. performance status (KPS < 70/70/80/90/100, %): 1/6/28/34/32;

  2. prior nephrectomy (%): 88;

  3. prior systemic therapies for advanced RCC (1/2, %): 72/28;

  4. prior systemic therapies for mRCC (sunitinib/pazopanib/axitinib, %): 59/30/10;

  5. risk prognosis (MSKCC favourable/intermediate/poor, %): 36/49/15.

Interventions

Group 1 (n = 410): nivolumab

Nivolumab 3 mg/kg IV every 2 weeks, no dose modifications permitted).

Group 0 (n = 411): everolimus

Everolimus 10 mg orally, every day, dose modifications permitted.

Cointerventions (standard‐of‐care according to local practice): not specified.

Outcomes

OS (primary endpoint)

How measured: time from randomization to the date of death.

Time points measured: every 8 weeks for the first year, and then every 12 weeks until disease progression or discontinuation of treatment, after discontinuation of treatment, participants followed every 3 months for assessment of survival and subsequent anticancer therapy.

Time points reported: Kaplan‐Meier survival curves over up to 30 months, median with 95% CI, unstratified HR with 98.5% CI.

Subgroups: MSKCC prognostic score, previous antiangiogenic regimens, region, age, gender.

AEs, grade ≥ 3 (secondary endpoint)

How measured: NCI‐CTC AE V4.0.

Time points measured: at each clinic visit.

Time points reported: overall frequency of all and most common treatment‐related AEs (fatigue, pruritus, stomatitis, anaemia) and AEs grade 3/4 including treatment‐related deaths.

Subgroups: not reported.

QoL (secondary endpoint)

How measured: health‐related QoL assessments with FACT FKSI‐DRS and a resulting summary score and EQ‐5D.

Time points measured: baseline, after randomization but before cycle 1 of therapy, on day 1 of each cycle, at the first 2 follow‐up visits (each assessment before physician contact, treatment doses and any procedures), about 30 and 100 days after last dose, EQ‐5D: additional at each of the 10 survival follow‐ups visits (every 3 months).

Time points reported: FKSI‐DRS and EQ‐5D (utility index and VAS) with completion rates at baseline, mean change from baseline to weeks 4 to 104, clinically important improvements, time to improvement (Cella 2016).

Subgroups: not reported.

PFS (secondary endpoint)

How measured: time from randomization to first documented RECIST‐defined tumour progression or death from any cause.

Time points measured: CT and MRI at baseline, every 8 weeks for the first year and then every 12 weeks until disease progression or discontinuation of treatment.

Time points reported: Kaplan‐Meier survival curves over up to 30 months, median.

Subgroups: not reported.

Tumour remission (secondary endpoint)

How measured: evaluated by the investigator (RECIST 1.1), number of randomized participants with a complete or partial response.

Time points measured: CT and MRI at baseline, every 8 weeks for the first year and then every 12 weeks until disease progression or discontinuation of treatment.

Time points reported: overall response.

Subgroups: not reported.

Funding sources

Bristol Myers Squibb.

Declarations of interest

RJM: honoraria from Bayer, Pfizer, Novartis and GlaxoSmithKline. SG: fees for consulting and serving on advisory boards from Bristol‐Myers Squibb, Novartis, Bayer, Sanofi‐Aventis, Astellas, Xcenda and Onclive; grant support from Bristol‐Myers Squibb, Novartis, Bayer, Pfizer, Acceleron, Merck and Agensys. HJH: grant support from Pfizer, Newlink Genetics, GlaxoSmithKline and SFJ Pharmaceuticals. SST: fees for serving on advisory boards from Prometheus; consulting fees from Amgen; grant support through his institution from Prometheus, Argos Therapeutics, Immatics Biotechnologies, Novartis and Exelixis. GP: fees for serving on advisory boards from Janssen and Novartis; lecture fees from Astellas and Pfizer; grant support from Bayer. ERP: fees for serving on advisory boards from Merck, Dendreon, GlaxoSmithKline, Pfizer, Astellas, Novartis and Genentech; grant support from AstraZeneca, Eli Lilly, Merck, Dendreon, GlaxoSmithKline, Acceleron and Pfizer. TKC: fees for consulting and for serving on advisory boards from GlaxoSmithKline, Novartis, Pfizer, Merck, AstraZeneca, Bayer and Prometheus; grant support through his institution from Bristol‐Myers Squibb, GlaxoSmithKline, Novartis, Exelixis, Pfizer, Merck, Roche, AstraZeneca, TRACON Pharmaceuticals and Peloton. HG: fees for serving on advisory boards from Novartis, Bayer, Sanofi‐Aventis, Astellas and Pfizer. FD: grant support from Novartis, Pfizer and GlaxoSmithKline. PB: honoraria from GlaxoSmithKline, Pfizer and Orion. JW: fees for serving on advisory boards, paid to his institution, from Bristol‐Myers Squibb, Novartis, GlaxoSmithKline, Roche and Amgen. YT: fees for serving on advisory boards from ONO Pharmaceuticals and Pfizer; honoraria and grant support from ONO Pharmaceuticals, Novartis and Pfizer. TCG: fees for consulting and serving on advisory boards from Boehringer Ingelheim, Merck Serono, Novartis, Pfizer, GlaxoSmithKline, Merck Sharp & Dohme, Bayer HealthCare, Roche, Bristol‐Myers Squibb, Eli Lilly and Janssen‐Cilag; honoraria from Boehringer Ingelheim, Merck Serono, Novartis, Pfizer, GlaxoSmithKline, Bayer, Roche, Eli Lilly, Janssen‐Cilag, Sanofi‐Aventis; travel support from Boehringer Ingelheim, Merck Serono, Pfizer, Roche and Eli Lilly; owning stock in Bayer. FAS: fees for serving on advisory boards from Pfizer, GlaxoSmithKline and Novartis; lecture fees from GlaxoSmithKline. CK: fees for serving on advisory boards from Pfizer, Novartis, Sanofi‐Aventis, Bayer and Seattle Genetics; lecture fees from Pfizer and Novartis. AR: lecture fees from Merck Sharp & Dohme. JSS, LAX, IMW: employees of and hold stock in Bristol‐Myers Squibb. PS: reports receiving consulting fees from Jounce Therapeutics, Amgen, Bristol‐Myers Squibb, GlaxoSmithKline and AstraZeneca/MedImmune; also founder of and holds stock in Jounce Therapeutics.

Notes

Registration: NCT01668784 (CheckMate025).

Study was stopped early due to the results of a planned interim analysis by the independent data monitoring committee showing significant benefit for OS.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Low risk of selection bias assumed due to block randomization, stratified by region, MSKCC prognostic risk group and the number of previous antiangiogenic therapy regimens (1 or 2) for advanced RCC.

Allocation concealment (selection bias)

Low risk

Central randomization.

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Open‐label study.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

No performance bias on OS assumed.

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

Open‐label study.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

No detection bias on OS assumed.

Incomplete outcome data (attrition bias)
(OS and PFS)

Low risk

Low risk of attrition bias assumed, high completeness of follow‐up with similar censoring in between treatment groups.

Incomplete outcome data (attrition bias)
(safety)

Low risk

Low risk of attrition bias assumed, safety analysis bases on all treated participants.

Incomplete outcome data (attrition bias)
(tumour remission)

High risk

High risk of bias on attrition bias for tumour remission assumed due to different numbers of non‐evaluated participants (6% with nivolumab vs 12% with everolimus).

Incomplete outcome data (attrition bias)
(QoL)

Low risk

High completion rates (≥ 80% in the first year) with no differences between treatment groups.

Selective reporting (reporting bias)

Low risk

All preplanned outcomes from the protocol reported.

Other bias

High risk

Stopped early for benefit in OS, subsequent systematic therapies (group 1: 55%; group 0: 63%) with cross‐over (25% from group 1 to group 0; 1.7% from group 0 to group 1).

Negrier 2011

Methods

Study design: 3‐arm, parallel‐group, open‐label RCT.

Study dates: randomization from May 2008 to May 2009, median follow‐up 23.2 months, study completion date February 2012.

Setting: multicentre (24 centres), national, phase II.

Country: France.

Participants

Inclusion criteria: histologically confirmed mRCC of all histological subtypes except papillary carcinomas; aged ≥ 18 years; ECOG Performance Status 0 to 2; measurable metastases; liver, renal and haematological functions in the range of 1.5 to 2 times above or below normal values; normal lipid and glycaemic concentrations; normal cardiac function within 6 weeks before randomization.

Exclusion criteria: brain metastases, hypertension, systemic treatment for the disease, history of arterial or venous thrombosis in the past 6 months.

Sample size: 171.

Age (years, median with range): group 1: 62 (40 to 79); group 0a: 62 (33 to 83); group 0b: 61 (33 to 83).

Sex (M/F, %): group 1: 66/34; group 0a: 74/26; group 0b: 76/24.

Prognostic factors (all randomized participants):

  1. performance status (ECOG 0 or 1/2, %): 88/12;

  2. prior nephrectomy (%): 87;

  3. prior systemic therapies: excluded;

  4. risk prognosis (good/intermediate/poor, %): 34/53/13.

Interventions

Group 1 (n = 41): IFN‐α + bevacizumab

IFN‐α 9 mIU SC 3 times/week + bevacizumab 10 mg/kg IV every 2 weeks.

Group 0 (n = 42): sunitinib

Sunitinib 50 mg/day for 4 weeks, followed by 2 weeks off.

Group 0a (n = 88): temsirolimus + bevacizumab (excluded, not standard treatment).

Temsirolimus 25 mg IV weekly + bevacizumab 10 mg/kg IV every 2 weeks.

Treatments continued until disease progression, unacceptable toxicity or protocol violation.

Cointerventions (standard‐of‐care according to local practice): not specified.

Outcomes

OS (secondary endpoint)

How measured: time from randomization to death from any cause.

Time points measured: during follow‐up.

Time points reported: 12‐months OS (study is ongoing for long‐term OS).

Subgroups: not reported.

AEs, grade ≥ 3 (secondary endpoint)

How measured:participants on study medication were assessed (NCI‐CTCAE v.3.0), data safety monitoring committee.

Time points measured: at day 15 and then at least every 6 weeks over 48 weeks.

Time points reported: main types of AEs (all grades and grade ≥ 3), frequency of AEs and SAEs ≥ 3.

Subgroups: not reported.

QoL not evaluated

PFS (primary endpoint)

How measured: time from randomization to disease progression or death from any cause (central reviewed data), 4 follow‐up CT scans according to RECIST 1.0.

Time points measured: baseline and then every 12 weeks over 48 weeks with 4 follow‐up CT scans.

Time points reported: Kaplan‐Meier survival curves over up to 30 weeks, median PFS with 95% CI.

Subgroups: not reported.

Tumour remission (secondary endpoint)

How measured: thoracic, abdominal and pelvic CT scan, brain MRI or CT and bone scan.

Time points measured: baseline and then every 12 weeks.

Time points reported: best response.

Subgroups: not reported.

Funding sources

French Ministry of Health and Wyeth Pharmaceuticals.

Declarations of interest

SN: honoraria from Novartis, Wyeth, Pfizer, GlaxoSmithKline and Roche; research funding from Wyeth, Roche and Novartis. DP: honoraria from Bayer, Eli Lilly and Roche. JOB: honoraria from Amgen; consultant with Novartis. LG, BL: honoraria from Novartis. BE: honoraria from Bayer, Roche, Pfizer, Genentech, Novartis, GlaxoSmithKline and Aveo; consultant with Bayer, Pfizer and Roche. All other authors declared no conflicts of interest.

Notes

Registration: NCT00619268 (TORAVA).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Low risk of selection bias assumed, computer‐generated list, permutated blocks, stratification by participating centre and performance status.

Allocation concealment (selection bias)

Low risk

Low risk of selection bias assumed due to central allocation.

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Participants and investigators were unmasked.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

No performance bias on OS assumed.

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

Masked central review of CT scans done in 89% of all randomized participants.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

No detection bias on OS assumed.

Incomplete outcome data (attrition bias)
(OS and PFS)

Low risk

No attrition bias assumed due to high completeness of, and similar censoring in, different treatment groups during follow‐up.

Incomplete outcome data (attrition bias)
(safety)

Low risk

No attrition bias assumed, all participants who received at least 1 dose of the study drug were included.

Incomplete outcome data (attrition bias)
(tumour remission)

Low risk

No attrition bias assumed, response was reported for all participants.

Incomplete outcome data (attrition bias)
(QoL)

Unclear risk

Not evaluated.

Selective reporting (reporting bias)

High risk

Information on long‐term OS and QoL not published despite planning in protocol.

Other bias

High risk

Blocked randomization in centres in an unblinded trial, differences in second‐line treatment after study treatment failure because of toxicity or progression with lower rates of second‐line therapies with sunitinib (48%) compared to 68% to 69% in other groups.

Rini 2010

Methods

Study design: 2‐arm, parallel‐group RCT.

Study dates: randomization from October 2003 to July 2005, data cutoff March 2009, median follow‐up among surviving participants 46.2 months.

Setting: multicentre, international.

Countries: Canada, US.

Participants

Inclusion criteria: people with mRCC; clear‐cell histological component confirmed by local pathology review; no prior systemic therapy for RCC; KPS ≥ 70%; aged ≥ 18 years; adequate bone marrow, hepatic and renal function; serum creatinine ≤ 1.5 times ULN.

Exclusion criteria: central nervous system metastases; NYHA class II to IV heart failure; bleeding within 6 months; blood pressure that could not be controlled < 160/90 mmHg with medication; history of venous thrombosis within 1 year or arterial thrombosis within 6 months or who required ongoing therapeutic anticoagulation; uncontrolled thyroid function; pregnancy; requirement for systemic corticosteroids greater than physiological replacement doses or delayed healing wounds, ulcers or bone fractures.

Sample size:732.

Age (years, median with IQR): group 1: 61 (56 to 70); group 0: 62 (55 to 70).

Sex (M/F, %): group 1: 73/27; group 0: 66/34.

Prognostic factors:

  1. performance status (ECOG Performance Status 0/1/2, %): 62/37/1;

  2. prior nephrectomy (%): 85;

  3. prior systemic therapies (%): 0 (as per inclusion criteria);

  4. risk prognosis (MSKCC) risk score poor/intermediate/high (%): 10/64/26.

Interventions

Group 1 (n = 363): IFN‐α

IFN‐α‐2a (Intron; Schering‐Plough, Kenilworth, NJ), provided by the NCI Cancer Therapy Evaluation Program, 9 MU SC 3 times/week (non‐consecutive days).

Dose reduction to 6 MU and 3 MU if IFN‐related toxicity present.

Group 0 (n = 369): IFN‐α + bevacizumab

IFN‐α‐2a (Intron; Schering‐Plough, Kenilworth, NJ), provided by the NCI Cancer Therapy Evaluation Program, 9 MU SC 3 times/week (non‐consecutive days).

Dose reduction to 6 MU and 3 MU if IFN‐related toxicity present.

Bevacizumab (provided by the NCI Cancer Therapy Evaluation Program) 10 mg/kg IV every 2 weeks.

Cointerventions (standard‐of‐care according to local practice): not specified.

Outcomes

OS (primary outcome)

How measured: time from registration to death from any cause.

Time points measured: during treatment and follow‐up.

Time points reported: Kaplan‐Meier curves over up to 60 months, median OS.

Subgroups: nephrectomy, MSKCC, liver metastases, age, gender.

AEs, grade ≥ 3 (secondary outcome)

How measured: ongoing documentation of AEs (CTCAE v.3.0).

Time points measured: baseline, every 12 weeks.

Time points reported: frequency of participants with AEs grade ≥ 3, deaths due to AEs, treatment‐related AEs to March 2009.

Subgroups: not reported.

QoL not evaluated

PFS (secondary outcome)

How measured: time between randomization and date of progression or death, investigator assessment of x‐rays.

Time points measured: baseline, every 12 weeks.

Time points reported: Kaplan‐Meier survival curves for up to 60 months, median PFS.

Subgroups: number of adverse risk factors.

Tumour remission (secondary outcome)

How measured: investigator assessment of x‐rays, RECIST criteria.

Time points measured: baseline, every 12 weeks.

Time points reported: overall response rate.

Subgroups: not reported.

Funding sources

Supported in part by National Cancer Institute to the Cancer and Leukemia Group B (CALGB) (Grant No. CA31946, CA33601) and by National Cancer Institute (Grants No. CA60138, CA41287, CA47642, CA45808, CA77440, CA14985, CA77202).

Declarations of interest

BIR: consultant or advisory role: Genentech. WMS: consultant or advisory role: Genentech; research funding: Genentech. JP: consultant or advisory role: Genentech; honoraria: Genentech. JD: consultant or advisory role: Genentech and Novartis; honoraria: Pfizer, Novartis; research funding: Novartis, Genentech, Pfizer. DAV: research funding: Genentech.

Notes

Registration: NCT00072046 (CALGB 90206).

Results on PFS and overall response rate published in Rini 2008, no cross‐over was permitted for participants randomly assigned to IFN‐α monotherapy, a substantial percentage of participants in both arms received systemic anticancer therapy subsequent to progression (62% of participants on IFN‐ monotherapy and 54% of participants on bevacizumab + IFN‐α) (mostly with sunitinib or sorafenib).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Stratified random block design, stratified by nephrectomy status and number of adverse prognostic factors.

Allocation concealment (selection bias)

Unclear risk

No information provided.

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

No blinding.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

No performance bias on OS assumed.

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

Non‐blinded trial and no independent review of x‐rays could potentially have contributed to the improved PFS and overall response rate.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

No detection bias on OS assumed.

Incomplete outcome data (attrition bias)
(OS and PFS)

Low risk

No attrition bias assumed due to similar censoring, 657/732 (90%) participants experienced progression or death.

Incomplete outcome data (attrition bias)
(safety)

Low risk

Based on all participants who were eligible for evaluation for toxicity (362/363 from the intervention and 347/369 from the control group, reasons not reported).

Incomplete outcome data (attrition bias)
(tumour remission)

Unclear risk

No data reported.

Incomplete outcome data (attrition bias)
(QoL)

Unclear risk

Not evaluated.

Selective reporting (reporting bias)

Low risk

No differences in outcomes to the protocol.

Other bias

High risk

Treatment with second‐line systemic anticancer therapy subsequent to progression (62% of participants on IFN‐α monotherapy and 54% of participants on bevacizumab + IFN‐α) might bias OS.

Rini 2015

Methods

Study design: 2‐arm, parallel‐group, open‐label RCT.

Study dates: December 2010 to July 2015 (study start to study completion date according to the trial registration).

Setting: multicentre, international, phase III.

Countries: Europe (France, Germany, Hungary, Italy, Poland, Russia, UK, Netherlands, Romania, Norway), US.

Participants

Inclusion criteria: metastatic or locally advanced (or both) RCC with clear‐cell histology (histological confirmation by local pathologist required), aged > 18 years, HLA‐A*02‐positive type, candidates for a first‐line therapy with sunitinib, favourable or intermediate‐risk (favourable risk: none, intermediate risk: 1 or 2 of the following criteria applied: haemoglobin < LLN, serum corrected calcium > ULN, KPS < 80%, time from initial diagnosis to initiation of therapy < 1 year, absolute neutrophil count > ULN, platelets > ULN), women who were postmenopausal or surgically sterile or practiced medically acceptable method of contraception, men willing to use contraception or had undergone vasectomy.

Exclusion criteria: prior systemic therapy for metastatic disease; history of or current brain metastases; abnormal ≥ CTC grade 3 laboratory values for haematology, liver and renal function; metastatic second malignancy; localized second malignancy expected to influence the person's lifespan; history or evidence of systemic autoimmune disease; known HIV infection; active infections requiring oral or IV antibiotics; any other known infection with a biological agent that can cause a severe disease and posed a severe danger to laboratory personnel working on participants' blood or tissue; received study drug within any clinical study within 4 weeks before sunitinib start; serious intercurrent illness, which according to the investigator, posed an undue risk for the person when participating in the trial; < 12 months since myocardial infarction, severe or unstable angina, coronary or peripheral artery bypass graft or cerebrovascular event.

Sample size:339.

Age (years, mean): group 1: 62.2; group 0: 59.8.

Sex (M/F, %): group 1: 70/30; group 0: 65/35.

Prognostic factors:

  • performance status (KPS 100/90/≤ 80, %): 48/39/13;

  • prior nephrectomy (%): 90;

  • prior systemic therapies: 0 (as per inclusion criteria);

  • risk prognosis (Heng Score favourable/intermediate/poor, %): 27/71/2.

Interventions

Group 1 (n = 204): IMA901 + sunitinib

Single infusion of cyclophos 300 mg/m2 3 days prior to first vaccination, 10 intradermal vaccination IMA901 + GM‐CSF 75 µg.

1 cycle sunitinib prior to randomization, sunitinib 50 mg orally (4 weeks/2 weeks off).

Group 0 (n = 135): sunitinib alone

1 cycle sunitinib prior to randomization, sunitinib 50 mg orally (4 weeks/2 weeks off).

Cointerventions (standard‐of‐care according to local practice): not specified.

Outcomes

OS (primary endpoint)

How measured: investigator‐assessed.

Time points measured: not reported.

Time points reported: Kaplan‐Meier‐curves over 42 months, median OS, log rank P value.

Subgroups: favourable and intermediate risk.

AEs, grade ≥ 3 (secondary endpoint)

How measured: investigator‐assessed, AEs, physical examinations, vital signs, haematology, clinical chemistry, urinalysis and electrocardiographic changes.

Time points measured: not reported.

Time points reported: most frequent (≥ 10% of participants) AEs.

Subgroups: not reported.

QoL: not measured.

PFS (secondary endpoint)

How measured: RECIST 1.1, central review and investigator analysis.

Time points measured: not reported.

Time points reported: Kaplan‐Meier‐curves over 24 months, median PFS, log rank P value.

Subgroups: not reported.

Tumour remission (secondary endpoint)

How measured: RECIST 1.1, images collected centrally and interpreted by independent radiologists and oncologists who assessed the tumour images without being informed about participant's treatment and the local assessment of site investigators.

Time points measured: not reported.

Time points reported: best objective response.

Subgroups: not reported.

Funding sources

Immatics Biotechnologies GmbH, Pfizer.

Declarations of interest

CR, HS, TW: shareholders of Immatics biotechnologies GmbH. JL, DM, RM, AM, JF, AK: employees of Immatics biotechnologies GmbH.

Notes

Registration: NCT01265901.

Published as abstract.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Missing information on generation of randomization, 3:2 block randomization, stratified by factors included risk group, nephrectomy and region (Western EU, US, Central Eastern EU).

Allocation concealment (selection bias)

Low risk

Low risk of selection bias assumed due to central allocation via fax or email (or both).

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Open‐label trial.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

No performance bias on OS assumed.

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

No performance bias, assessment of response by blinded assessors.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

No detection bias on OS assumed.

Incomplete outcome data (attrition bias)
(OS and PFS)

Low risk

No attrition bias assessed due to similar censoring in the treatment groups.

Incomplete outcome data (attrition bias)
(safety)

Low risk

No attrition bias assessed due to reporting in the safety population of all treated participants.

Incomplete outcome data (attrition bias)
(tumour remission)

Low risk

No attrition bias assessed due to reporting in all randomized participants.

Incomplete outcome data (attrition bias)
(QoL)

Unclear risk

Not evaluated.

Selective reporting (reporting bias)

Low risk

All preplanned outcomes reported.

Other bias

Low risk

No other bias identified.

AE: adverse event; CI: confidence interval; CT: computer tomography; CTC: Common Terminology Criteria; ECOG: Eastern Cooperative Oncology Group; EQ‐5D: EuroQol 5‐Dimension; EQ‐VAS: EuroQol Visual Analogue Scale; FACT‐G: Functional Assessment of Cancer Therapy ‐ General; FKSI‐15: 15‐item Kidney Symptom Index; FKSI‐DRS: Kidney Symptom Index ‐ Disease‐Related Symptoms; HR: hazard ratio; IL: interleukin; IM: intramuscular; IFN‐α: interferon‐α; IV: intravenous; KPS: Karnovsky Performance Status; LLN: lower limit of normal; mIU: milli‐international unit; mRCC: metastatic renal cell carcinoma; MRI: magnetic resonance imaging; MSKCC: Memorial Sloan‐Kettering Cancer Center; MU: million units; n: number of participants; NCI‐CTC: National Cancer Institute Common Terminology Criteria; NYHA: New York Heart Association; OS: overall survival; PFS: progression‐free survival; QoL: quality of life; RCC: renal cell carcinoma; RCT: randomized controlled trial; RECIST: Response Evaluation Criteria In Solid tumours; SAE: serious adverse event; SC: subcutaneous; ULN: upper limit of normal; VAS: visual analogue score; VEGF: vascular endothelial growth factor.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Aass 2005

No comparison to current standard therapy as defined in review protocol (IFN‐α ± 13 cRA).

Adler 1987

No comparison to current standard therapy as defined in review protocol (autologous tumour cells + BCG vs norprogesterone).

Amato 2009

Not an RCT.

Amin 2015

Not an RCT.

Atkins 1993

No comparison to current standard therapy as defined per review protocol (IFN‐α + IL‐2 vs IL‐2).

Atzpodien 2001

No comparison to current standard therapy as defined in review protocol (IL‐2 + IFN‐α + 5‐FU vs tamoxifen).

Atzpodien 2004

No comparison to current standard therapy as defined per review protocol (SC IL‐2/SC IFN‐α + IL‐2/IV 5‐FU vs SC IL‐2/SC IFN‐α + IL‐2/IV 5‐FU/OP 13 cRA vs SC IFN‐α/IV vinblastine).

Atzpodien 2005

Not mostly mRCC (stage IV) patients.

Atzpodien 2006

No comparison to current standard therapy as defined per review protocol (IL‐2 + IFN‐α + PO 13cRA/inhaled + inhaled IL‐2 vs IL‐2 + IFN‐α + PO 13cRA/inhaled).

Bellmunt 2008

Secondary publication to Hudes 2007.

Boccardo 1998

No comparison to current standard therapy as defined in review protocol (IFN‐α + IL‐2 vs IFN‐α vs IL‐2).

Borden 1990

No comparison to current standard therapy as defined in review protocol (IFN‐͎β IV different doses).

Bracarda 2010

Secondary publication to Escudier 2007.

Bracarda 2013

No comparison to current standard therapy as defined in review protocol (sorafenib + high‐dose IFN‐α vs sorafenib + low‐dose IFN‐α).

Brinkmann 2004

No comparison to current standard therapy as defined in review protocol (IL‐2 + IFN‐α + 5‐FU vs misletoe lectin).

Bromwich 2002

Not an RCT.

Buzogany 2001

No comparison to current standard therapy as defined in review protocol (IFN‐α vs vinblastine).

Castellano 2009

Secondary publication to Motzer 2007.

Cella 2008

Secondary publication to Motzer 2007.

Cella 2010

Secondary publication to Motzer 2007.

Cella 2016

Secondary publication to Motzer 2015a.

Choueiri 2014

No comparison to current standard therapy as defined in review protocol (pharmacodynamic study with nivolumab).

Clark 2003

Not mostly mRCC (stage IV) patients (adjuvant study).

Creagan 1991

No comparison to current standard therapy as defined in review protocol (IFN‐α vs aspirin).

De Mulder 1995

No comparison to current standard therapy as defined in review protocol (IFN‐α ± IFN‐γ).

Dexeus 1989

No comparison to current standard therapy as defined in review protocol (combined chemotherapy ± IFN‐α).

Dillman 2003

Study of mixed solid tumours with no separate analysis of mRCC patients.

Donskov 2006

No comparison to current standard therapy as defined in review protocol (reduced dose IL‐2 ± histamine).

Du Bois 1997

Study of mixed solid tumours with no separate analysis of mRCC patients.

Dudek 2008

No comparison to current standard therapy as defined in review protocol (LMI vaccination + cyclophosphamide + IL‐2 vs LMI + cyclophosphamide vs LMI).

Dutcher 2003

No comparison to current standard therapy as defined in review protocol (IFN‐γ + IFN‐α vs IFN‐γ).

Dutcher 2009

Secondary publication to Hudes 2007.

Edsmyr 1985

No comparison to current standard therapy as defined in review protocol (IFN‐α vs pulmonal irradiation + vincristine + bleomycin).

Elkord 2013

No comparison to current standard therapy as defined in review protocol (IFN‐α ± naptumomab estafenox).

Escudier 2009

No comparison to current standard therapy as defined in review protocol (IFN‐α vs first‐line sorafenib).

Escudier 2010

Secondary publication to Escudier 2007.

Escudier 2011

Preliminary publication to Negrier 2011.

Fenton 1996

Not mostly mRCC (stage IV) patients (adjuvant study).

Figlin 1999

No comparison to current standard therapy as defined in review protocol (TIL vaccination + IL‐2 vs IL‐2).

Figlin 2009

Secondary publication to Hudes 2007.

Flanigan 2001

No comparison to current standard therapy as defined in review protocol (IFN‐α ± initial nephrectomy).

Foon 1988

No comparison to current standard therapy as defined in review protocol (IFN‐α + IFN‐γ vs IFN‐γ).

Fosså 1992

No comparison to current standard therapy as defined in review protocol (IFN‐α ± vinblastine).

Fosså 2004

No comparison to current standard therapy as defined in study protocol (IFN‐α ± 13 cRA).

Fujita 1992

No comparison to current standard therapy as defined in review protocol (IFN‐α different doses).

Galligioni 1996

Not mostly mRCC (stage IV) patients (adjuvant study).

Gleave 1998

No comparison to current standard therapy as defined in review protocol (IFN‐γ vs placebo).

Gore 2010

No comparison to current standard therapy as defined in review protocol (IFN‐α vs IFN‐α + IL‐2 + 5‐FU).

Harlin 2004

Not an RCT.

Henriksson 1998

No comparison to current standard therapy as defined in review protocol (IFN‐α + tamoxifen vs tamoxifen).

Jayson 1998

No comparison to current standard therapy as defined in review protocol (IFN‐α + IL‐2 vs IFN‐α).

Jocham 2004

Not mostly mRCC (stage IV) patients (adjuvant study).

Jonasch 2010

No comparison to current standard therapy as defined in review protocol (first‐line sorafenib + IFN‐α vs sorafenib).

Keefe 2015

No immunotherapeutic intervention (CRLX101 + bevacizumab).

Kempf 1986

No comparison to current standard therapy as defined in review protocol (high IFN‐α vs low‐dose IFN‐α).

Kim 2015

Secondary publication to Rini 2015.

Kinouchi 2004

No comparison to current standard therapy as defined in review protocol (IFN‐α ± cimetidine).

Kirkwood 1985

No comparison to current standard therapy as defined in review protocol (high‐dose vs low‐dose IFN‐α).

Koretz 1991

No comparison to current standard therapy as defined in review protocol (LAK vaccination + IL‐2 vs IL‐2).

Kriegmair 1995

No comparison to current standard therapy as defined in review protocol (IFN‐α + vinblastine vs medroxyprogesterone acetate).

Kwitkowski 2010

Secondary publication to Hudes 2007.

Law 1995

No comparison to current standard therapy as defined in review protocol (LAK vaccination + IL‐2 vs IL‐2).

Lissoni 1993

No comparison to current standard therapy as defined in review protocol (IL‐2 + IFN‐α vs IL‐2).

Lissoni 2000

No comparison to current standard therapy as defined in review protocol (reduced‐dose IL‐2 ± melatonin).

Lissoni 2003

No comparison to current standard therapy as defined in review protocol (IL‐2 + GM‐CSF vs IL‐2).

Liu 2012

No comparison to current standard therapy as defined in review protocol (CIK vaccination vs IL‐2 + IFN‐α).

Lummen 1996

No comparison to current standard therapy as defined in review protocol (IL‐2 + IFN‐α vs IFN‐γ).

Majhail 2006

Not mostly mRCC (stage IV) patients (adjuvant study).

Margolin 1997

Study of mixed solid tumours with no separate analysis of mRCC patients.

McCabe 1991

No comparison to current standard therapy as defined in review protocol (reduced‐dose IL‐2 vs LAK).

McDermott 2005

No comparison to current standard therapy as defined in review protocol (high‐dose IL‐2 vs reduced‐dose IL‐2 + IFN‐α).

Melichar 2008

Secondary publication to Escudier 2007.

Messing 2003

Not mostly mRCC (stage IV) patients (adjuvant study).

Mickisch 2001

No comparison to current standard therapy as defined in review protocol (IFN‐α ± initial nephrectomy).

Motzer 2000

No comparison to current standard therapy as defined in review protocol (IFN‐α ± 13 cRA).

Motzer 2001

No comparison to current standard therapy as defined in review protocol (IL‐2 + IFN‐α vs IL‐2).

Motzer 2009

Secondary publication to Motzer 2007.

Motzer 2015b

Preliminary study to Motzer 2015a.

Motzer 2015c

No comparison to current standard therapy as defined in review protocol (dose‐response study of nivolumab).

MRCRCC 1999

No comparison to current standard therapy as defined in review protocol (IFN‐α vs medroxyprogesterone acetate).

Muss 1987

No comparison to current standard therapy as defined in review protocol (IV IFN‐α vs SC IFN‐α).

Naglieri 1998

No comparison to current standard therapy as defined in review protocol (IFN‐α + IL‐2 vs IL‐2 + epidoxorubicin).

NCT00352859

No comparison to current standard therapy as defined in review protocol (sorafenib + IFN‐α vs sorafenib + gemcitabine ‐ early termination of study because of slow accrual with no data analysis).

NCT00678288

Stopped early due to slow accrual, no data analysis performed (sorafenib + IFN‐α vs sorafenib).

Negrier 1998

No comparison to current standard therapy as defined in review protocol (IL‐2 + IFN‐α vs IL‐2).

Negrier 2000

No comparison to current standard therapy as defined in review protocol (IFN‐α + IL‐2 ± 5‐FU).

Negrier 2007

No comparison to current standard therapy as defined in review protocol (IL‐2 + IFN‐α vs IL‐2 vs IFN‐α vs medroxyprogesterone acetate).

Negrier 2008

No comparison to current standard therapy as defined in review protocol (IV IL‐2 + IFN‐α vs SC IL‐2 + IFN‐α ).

Negrier 2010

No immunotherapeutic intervention (sorafenib vs placebo).

Neidhart 1991

No comparison to current standard therapy as defined in review protocol (IFN‐α ± vinblastine).

Osband 1990

No comparison to current standard therapy as defined in review protocol (cimetidine + ALT vaccination vs cimetidine).

Otto 1988

No comparison to current standard therapy as defined in review protocol (IFN‐α ± vinblastine).

Oudard 2011

Secondary publication to Motzer 2007.

Passalacqua 2010

No comparison to current standard therapy as defined in review protocol (maintenance therapy after disease progression).

Passalacqua 2014

Not mostly mRCC (stage IV) patients (adjuvant study).

Patel 2008

No comparison to current standard therapy as defined in review protocol (study A. no RCT; study B: IL‐2 + SRL172 vaccination vs IL‐2).

Patil 2012

Secondary publication to Motzer 2007.

Pedersen 1980

No comparison to current standard therapy as defined in review protocol (nephrectomy ± plasma).

Pickering 2009

Secondary publication to Motzer 2007.

Pizzocaro 2001

Not mostly mRCC (stage IV) patients (adjuvant study).

Porzsolt 1988

No comparison to current standard therapy as defined in review protocol (IFN‐α ± medroxyprogesterone acetate).

Powles 2015

No immunotherapeutic intervention.

Procopio 2011

No comparison to current standard therapy as defined in review protocol (first‐line sorafenib + IL‐2 vs sorafenib).

Pyrhönen 1999

No comparison to current standard therapy as defined in review protocol (IFN‐α + vinblastine vs vinblastine).

Quesada 1985

No comparison to current standard therapy as defined in review protocol (high‐dose vs low‐dose IFN‐α).

Radosavljevic 2000

No comparison to current standard therapy as defined in review protocol (IFN‐α + vinblastine ± medroxyprogesterone acetate).

Ravaud 2015

No comparison to current standard therapy (IFN + bevacizumab vs everolimus + bevacizumab).

Reddy 2006

Preliminary publication to Motzer 2007.

Rini 2004

Protocol to Rini 2010.

Rini 2008

Preliminary publication without OS to Rini 2010.

Rini 2012

No immunotherapeutic intervention (sorafenib ± AMG 386).

Rini 2014

No comparison to current standard therapy (IFN + bevacizumab vs temsirolimus + bevacizumab).

Rosenberg 1993

No comparison to current standard therapy as defined in review protocol (LAK vaccination + IL‐2 vs IL‐2).

Rossi 2010

No comparison to current standard therapy as defined in review protocol (dose‐response study of siltuximab).

Sagaster 1995

No comparison to current standard therapy as defined in review protocol (IFN‐α ± coumarin + cimetidine).

Scardino 1997

No comparison to current standard therapy as defined in review protocol (postoperative + preoperative IL‐2 vs postoperative IL‐2).

Schwaab 2000

No comparison to current standard therapy as defined in review protocol (AV vaccination + IFN‐α + IFN‐γ (together with AV) vs AV + IFN‐α + IFN‐γ (after initiation of AV)).

Sharma 2015

Preliminary study to Motzer 2015a.

Simons 1997

No comparison to current standard therapy as defined in review protocol (autologous vaccine ± GM‐CSF).

Smith 2003

Study of mixed solid tumours with no separate analysis of mRCC patients.

Soret 1996

Not mostly mRCC (stage IV) patients (adjuvant study).

Steineck 1990

No comparison to current standard therapy as defined in review protocol (IFN‐α vs medroxyprogesterone acetate).

Sternberg 2013

No immunotherapeutic intervention (pazopanib vs placebo).

Summers 2010

Secondary publication to Escudier 2007.

Tannir 2006

No comparison to current standard therapy as defined in review protocol (intermediate‐dose vs low‐dose IFN‐α).

Tsavaris 2000

No comparison to current standard therapy as defined in review protocol (IFN‐α + vinblastine vs IFN‐α).

Voss 2015

No immunotherapeutic intervention (CRLX101 + bevacizumab vs SOC).

Walter 2012

No comparison to current standard therapy as defined in review protocol (cyclophosphamide + IMA 901 vaccination vs IMA 901 vaccination).

Wang 2015

Not an RCT.

Weiss 1992

No comparison to current standard therapy as defined in review protocol (continuous IL‐2 vs bolus IL‐2 + LAK vaccination).

Witte 1995

No comparison to current standard therapy as defined in review protocol (IL‐2 + IFN‐β vs IL‐2).

Wood 2008

Not mostly mRCC (stage IV) patients (adjuvant study).

Yang 1995

No comparison to current standard therapy as defined in review protocol (IL‐2 + PEG‐IL‐2 vs IL‐2).

Yang 2003

No comparison to current standard therapy as defined in review protocol (IL‐2 different doses).

Yang 2007

Not an RCT.

Yang 2010

Secondary publication to Hudes 2007.

Zhan 2012

Not mostly mRCC (stage IV) patients (adjuvant study).

Zhao 2015

No comparison to current standard therapy as defined in review protocol (DC‐CIK vaccination vs IL‐2 + IFN‐α).

5‐FU: 5‐fluorouracil; ALT: alanine transaminase; CIK: cytokine‐induced killer; DC‐CIK: dendritic cell cytokine‐induced killer; GM‐CSF: granulocyte‐macrophage colony‐stimulating factor; IL: interleukin; IV: intravenous; LAK: lymphokine‐activated killer; mRCC: metastatic renal cell carcinoma; PEG‐IL‐2: pegylated interferon‐2; PO: per os (orally); RCT: randomized controlled trial; SC: subcutaneous; SOC: standard of care.

Characteristics of ongoing studies [ordered by study ID]

EudraCT2016‐002170‐13

Trial name or title

NIVOSWITCH: a Randomized Phase II Study with NIVOlumab or Continuation of Therapy as an Early SWITCH Approach in Patients with Advanced or Metastatic Renal Cell Carcinoma (RCC) and Disease Control after 3 Months of Treatment with a Tyrosine Kinase Inhibitor.

Methods

Study design: 2‐arm, parallel‐group, open‐label RCT.

Setting: multicentre, international, phase II.

Countries: Europe.

Participants

Main inclusion criteria: aged ≥ 18 years, either gender, histological confirmation of RCC with a clear‐cell component, ECOG Performance Status 0 to 2, metastatic or locally advanced RCC with clear‐cell component, not amenable to surgery with curative intention, first‐line treatment with a TKI for 10 to 12 weeks (limited to sunitinib or pazopanib), people with measurable disease (RECIST 1.1), adequate blood count, liver‐enzymes, and renal function.

Main exclusion criteria: prior systemic therapy other than 10 to 12 weeks SOC TKI treatment, complete remission or progression during SOC TKI first‐line treatment, termination of first‐line treatment with TKI due to intolerance, prior therapy with antitumour vaccines, anti‐PD‐L1, anti‐PD1, anti‐CTLA‐4, or other immunomodulatory antitumour agents, known chronic infection and intercurrent illness.

Sample size planned: 244.

Interventions

Group 1: nivolumab after TKI (sunitinib or pazopanib) and disease control.

Group 0: pazopanib after TKI (sunitinib or pazopanib) and disease control.

Outcomes

Primary outcome: 24 months OS.

Secondary outcomes: best overall response, PFS, QoL, safety, other.

Starting date

September 2016.

Contact information

AIO‐Studien‐gGmbH, Dr Aysun Karatas, info@aio‐studien‐ggmbh.de.

Notes

Sponsor protocol no: AIO‐NZK‐0116.

Figlin 2014

Trial name or title

ADAPT: an International Phase 3 Randomised Trial of Autologous Dendritic Cell Immunotherapy (AGS‐003) Plus Standard Treatment of Advanced Renal Cell Carcinoma (NCT01582672).

Methods

Study design: 2‐arm, parallel‐group, open‐label RCT.

Setting: multicentre, international, phase III.

Countries: Canada, Czech Republic, Hungary, Israel, Italy, Spain, UK, US.

Participants

Main inclusion criteria: aged ≥ 18 years; either gender; histological confirmation of advanced RCC with predominantly clear‐cell histology; advanced disease; metastatic disease (measurable or non‐measurable per RECIST 1.1); people who were candidates for standard first‐line therapy initiating with sunitinib; time from diagnosis to treatment < 1 year; KPS ≥ 70%; life expectancy ≥ 6 months; resolution of all acute toxic effects of prior radiotherapy or surgical procedures to grade ≤ 1 (NCI‐CTC 4.0); adequate haematological, renal, hepatic and coagulation function; negative serum pregnancy test for women with reproductive potential and agreement of both men and women of reproductive potential to use a reliable form of contraception during the study and for 12 weeks after the last dose of study drug.

Main exclusion criteria: prior systemic therapy of any type for RCC, including immunotherapy, chemotherapy, hormonal or investigational therapy; prior history of malignancy within the preceding 3 years, except for adequately treated in situ carcinomas or non‐melanoma skin cancer; adequately treated early‐stage breast cancer, superficial bladder cancer and non‐metastatic prostate cancer with a normal PSA; history of, or known, brain metastases; spinal cord compression, carcinomatous meningitis or evidence of brain or leptomeningeal disease; people with ≥ 4 of the following risk factors: haemoglobin < LLN, corrected calcium > 10 mg/dL, KPS < 80%, neutrophils > ULN, platelets > ULN, planned or elective surgical treatment postnephrectomy for the direct management of RCC, within 28 days before visit 1, NCI CTCAE grade 3 haemorrhage < 28 days before day 0, clinically significant comorbidities.

Sample size planned:450.

Interventions

Group 1: AGS‐003 + standard treatment (sunitinib).

Group 0: standard treatment (sunitinib).

Outcomes

Primary outcome: OS, duration from randomization to death.

Secondary outcomes: PFS, tumour response, AEs.

Starting date

November 2012.

Contact information

Robert Figlin, MD, principal Investigator.

Notes

Final data collection date for primary outcome measure: April 2017.

Hammers 2015

Trial name or title

(CheckMate 214): A Phase 3, Randomised, Open‐Label Study of Nivolumab Combined with Ipilimumab versus Sunitinib Monotherapy in Subjects with Previously Untreated, Advanced or Metastatic Renal Cell Carcinoma (NCT02231749).

Methods

Study design: 2‐arm, parallel‐group, open‐label RCT.

Setting: multicentre, international, phase III.

Countries: Argentina, Australia, Austria, Belgium, Brazil, Canada, Chile, Colombia, Czech Republic, Denmark, Finland, France, Germany, Hungary, Ireland, Israel, Italy, Japan, Korea, Republic of, Mexico, Netherlands, Poland, Spain, Sweden, Taiwan, Turkey, UK, US.

Participants

Main inclusion criteria: aged ≥ 18 years, either gender, histological confirmation of RCC with a clear‐cell component, advanced or metastatic (AJCC Stage IV) RCC, no prior systemic therapy for RCC with predefined exceptions (regular adjuvant or neoadjuvant therapy), KPS ≥ 70%, measurable disease (RECIST 1.1), archival or recent tumour tissue.

Exclusion criteria: cerebral metastases; prior systemic treatment with VEGF or VEGF receptor targeted therapy; prior treatment with an anti‐PD‐1, anti‐PD‐L1, anti‐PD‐L2, anti‐CD137 or anti‐CTLA‐4 antibody or any other antibody or drug specifically targeting T‐cell costimulation or checkpoint pathways; any active or recent history of a known or suspected autoimmune disease or recent history of a syndrome that required systemic corticosteroids or immunosuppressive medications except for syndromes which would not be expected to recur in the absence of an external trigger; vitiligo or type 1 diabetes mellitus or residual hypothyroidism due to autoimmune thyroiditis only requiring hormone replacement are permitted to enrol, any condition requiring systemic treatment with corticosteroids or other immunosuppressive medications within 14 days prior to first dose of study drug; inhaled steroids and adrenal replacement steroid doses > 10 mg daily; prednisone equivalents are permitted in the absence of active autoimmune disease.

Sample size planned:1070.

Interventions

Group 1: nivolumab 3 mg/kg + ipilimumab 1 mg/kg.

Group 0: sunitinib 50 mg.

Outcomes

Primary outcome: OS, PFS (coprimary).

Secondary outcomes: ORR, safety.

Starting date

October 2014.

Contact information

Sponsor: Bristol‐Myers Squibb.

Notes

Estimated primary completion date: May 2019.

NCT00930033

Trial name or title

CARMENA: Randomised Phase III Trial Evaluating the Importance of Nephrectomy in Patients Presenting with Metastatic Renal Cell Carcinoma Treated with Sunitinib.

Methods

Study design: 2‐arm, parallel‐group, open‐label RCT.

Setting: phase III, multicentre, national.

Countries: France.

Participants

Main inclusion criteria: aged ≥ 18 years, either gender, ECOG Performance Status 0 or 1, biopsy (primary tumour or metastases) confirming the diagnosis of clear‐cell carcinoma, documented metastatic disease, absence of prior systemic treatment for kidney cancer including antiangiogenic, tumour amenable to nephrectomy in the opinion of the patient's urologist, patients for whom the indication of sunitinib is considered according to the recommendation rules given by national health authorities of participating countries, prescription of sunitinib in the circumstances of the study is considered as a standard treatment, people with predefined adequate organ function.

Main exclusion criteria: prior systemic treatment for kidney cancer, bilateral kidney cancer, pregnant or breastfeeding women, specified comorbidities, symptomatic brain metastases.

Sample size planned: 576.

Interventions

Group 1: nephrectomy + sunitinib.

Group 0: sunitinib.

Outcomes

Primary outcome: OS.

Secondary outcome: ORR, PFS.

Starting date

September 2009.

Contact information

Principal Investigator: Arnaud Mejean, MD PhD, [email protected].

Notes

Estimated primary completion date: September 2019.

NCT01984242

Trial name or title

IMmotion 150: a Phase II, Randomised Study of Atezolizumab Administered as Monotherapy or In Combination with Bevacizumab versus Sunitinib In Patients with Untreated Advanced Renal Cell Carcinoma.

Methods

Study design: 3‐arm, parallel‐group, open‐label RCT.

Setting: phase II, multicentre, international.

Countries: Czech Republic, France, Germany, Italy, Poland, Romania, Spain, UK, US.

Participants

Main inclusion criteria: aged ≥18 years, either gender, unresectable advanced or metastatic renal cell carcinoma with component of clear‐cell histology or component of sarcomatoid histology that has not been previously treated with any systemic agents (or both), including treatment in the adjuvant setting, measurable disease, as defined by RECIST v1.1, KPS ≥ 70, adequate haematological and end‐organ function as defined by protocol.

Main exclusion criteria: cerebral metastases; radiotherapy for RCC within 28 days prior to cycle 1; uncontrolled pleural effusion, pericardial effusion or ascites; pregnancy and lactating women; life expectancy < 12 weeks.

Sample size planned: 305.

Interventions

Group 1: atezolizumab + bevacizumab.

Group 1.1: atezolizumab (following PD ‐ atezolizumab + bevacizumab).

Group 0: sunitinib (following PD ‐ atezolizumab + bevacizumab).

Outcomes

Primary outcome: PFS central reading.

Secondary outcomes: PFS investigator assessed, ORR, OS.

Starting date

January 2014.

Contact information

Sponsor: Hoffmann La Roche.

Notes

Estimated primary completion date: September 2018.

NCT02014636

Trial name or title

A Phase I/II Study to Assess the Safety and Efficacy of Pazopanib and MK 3475 in Subjects with Advanced Renal Cell Carcinoma.

Methods

Study design: part 1. non‐randomized dose escalation, part 2: 3‐arm, parallel‐group, open‐label RCT.

Setting: multicentre, international.

Countries: UK, US.

Participants

Inclusion criteria: aged ≥ 18 years, either gender, people with histologically confirmed advanced or mRCC, measurable disease, no prior systemic therapy, ECOG Performance Status 0 or 1, adequate organ function.

Exclusion criteria: cerebral metastases, active autoimmune disease, pregnancy, history of a malignancy (other than the disease under treatment in the study) within 5 years.

Sample size planned:228.

Interventions

Group 1: MK 3475 (pembrolizumab) + pazopanib.

Group 1.1: MK 3475 (pembrolizumab).

Group 0: pazopanib.

Outcomes

Primary outcome: part 2: PFS.

Secondary outcomes: part 2: ORR, OS, safety.

Starting date

December 2013.

Contact information

Sponsor: Novartis Pharmaceuticals.

Notes

Estimated primary completion date: May 2021.

NCT02089685

Trial name or title

KEYNOTE 029 ‐ a Phase I/II Clinical Trial to Study the Safety and Tolerability of MK‐3475 + Pegylated Interferon Alfa‐2b (PEG‐IFN) and MK‐3475 + Ipilimumab (IPI) in Subjects with Advanced Melanoma (MEL) and Renal Cell Carcinoma (RCC).

Methods

Study design: 3‐arm, parallel‐group, open‐label RCT.

Setting: multicentre, international.

Countries: Australia, New Zealand, US.

Participants

Inclusion criteria: aged ≥ 18 years, either gender, people with histologically confirmed advanced or metastatic melanoma or RCC, RCC participants must have received ≥ 1 prior line of therapy for metastatic disease, ECOG Performance Status of 0 or 1, adequate organ function.

Exclusion criteria: cerebral metastases, diagnosis of immunodeficiency or receiving systemic steroid therapy, additional malignancy, active infection requiring systemic therapy, pregnancy, breastfeeding women.

Sample size planned:343.

Interventions

Group 1: pembrolizumab + PegIFN‐2b.

Group 1.1: pembrolizumab + ipilimumab.

Group 0: pembrolizumab.

Outcomes

Primary outcome: dose‐limiting toxicities, AE, PFS.

Secondary outcomes: ORR, OS.

Starting date

March 2014.

Contact information

Sponsor: Merck Sharp & Dohme Corp.

Notes

Estimated primary completion date: April 2017.

NCT02210117

Trial name or title

A Pilot Randomised Tissue‐Based Study Evaluating Anti‐PD1 Antibody or Anti‐PD1 + Bevacizumab or Anti‐PD1 + Anti‐CTLA‐4 in Patients with Metastatic Renal Cell Carcinoma who are Eligible for Cytoreductive Nephrectomy, Metastasectomy or Post‐Treatment Biopsy.

Methods

Study design: 3‐arm, parallel‐group, double‐blind RCT.

Setting: multicentre.

Countries: US.

Participants

Main inclusion criteria: informed consent; histologically or cytologically confirmed clear‐cell mRCC who are eligible for cytoreductive nephrectomy; metastasectomy or post‐treatment biopsy; diagnosis must be confirmed by pathologist review of screening biopsy; measurable disease defined as a lesion that can be accurately measured in at least 1 dimension and measures ≥ 15 mm with conventional techniques or ≥ 10 mm with more sensitive techniques such as MRI or spiral CT scan; prior treatment for RCC including prior surgery, radiotherapy, immunotherapy with IL‐2 or interferon (but not anti‐PD1 or anti‐CTLA‐4); target therapy with RTK inhibitors/mTOR inhibitors, such as sunitinib, sorafenib, pazopanib, axitinib, everolimus and temsirolimus (but not bevacizumab) or chemotherapy allowed; ECOG Performance Status 0 or 1.

Main exclusion criteria: any other malignancy from which the person has been disease‐free for < 2 years, except for non‐melanoma skin cancer, in situ carcinoma of any site, organ allografts, major surgical procedure, open biopsy or significant traumatic injury with poorly healed wound within 6 weeks prior to first dose of study drug; or anticipation of need for major surgical procedure during the course of the study (other than in protocol); autoimmune disease; known history of testing positive for HIV or known AIDS; positive test for hepatitis B virus or positive test for hepatitis C virus.

Sample size planned: 60.

Interventions

Group 1: nivolumab.

Group 1.1: nivolumab + bevacizumab.

Group 1.2: nivolumab + ipilimumab.

Outcomes

Primary outcome: safety.

Secondary outcomes: immunological changes in tumour tissue, ORR.

Starting date

November 2014.

Contact information

Padmanee Sharma, MD, PhD.

Notes

Estimated primary data: November 2018.

NCT02420821

Trial name or title

A Phase III, Open‐Label, Randomised Study of Atezolizumab (Anti‐PD‐L1 Antibody) in Combination with Bevacizumab versus Sunitinib in Patients with Untreated Advanced Renal Cell Carcinoma.

Methods

Study design: 2‐arm, parallel‐group, open‐label RCT.

Setting: multicentre, international, phase III.

Countries: Australia, Bosnia and Herzegovina, Brazil, Canada, Czech Republic, Denmark, France, Germany, Italy, Japan, Korea, Republic of, Mexico, Poland, Russian Federation, Serbia, Singapore, Spain, Taiwan, Thailand, Turkey, Ukraine, UK, US.

Participants

Inclusion criteria: aged ≥ 18 years, definitive diagnosis of unresectable locally advanced or mRCC with clear‐cell histology or a component of sarcomatoid carcinoma (or both), with no prior treatment in the metastatic setting, evaluable MSKCC risk score, measurable disease (RECIST 1.1), KPS ≥ 70%, adequate haematological and end‐organ function.

Exclusion criteria: radiotherapy for RCC within 14 days prior to treatment; central nervous system disease; uncontrolled pleural effusion, pericardial effusion or ascites; uncontrolled hypercalcaemia; any other malignancies within 5 years except for low‐risk prostate cancer or those with negligible risk of metastasis or death; life expectancy < 12 weeks.

Sample size planned:830.

Interventions

Group 1: atezolizumab (MPDL3280A ‐ PD‐L1 AB) + bevacizumab.

Group 0: sunitinib.

Outcomes

Primary outcome: PFS, OS.

Secondary outcomes: ORR, safety, pharmacokinetics.

Starting date

May 2015.

Contact information

[email protected].

Notes

Primary results expected: June 2020.

NCT02432846

Trial name or title

An Open‐Label, Randomised, Controlled, Multicenter, Phase II Study Evaluating Safety and Efficacy of Intratumourally Administered Intuvax Pre‐nephrectomy Followed by Sunitinib Post‐Nephrectomy, Compared to Sunitinib Post‐Nephrectomy in Metastatic Renal Cell Carcinoma Patients (MERECA).

Methods

Study design: 2‐arm, parallel‐group, open‐label RCT, 2:1 randomization.

Setting: multicentre, phase II.

Countries: Sweden.

Participants

Main inclusion criteria: aged ≥ 18 years, either gender, informed consent, recent (< 6 months) diagnosed RCC with at least 1 CT‐verified metastasis, planned resection of primary tumour, primary tumour diameter ≥ 4 cm, candidate for standard first‐line therapy with sunitinib, adequate haematological parameters and liver function.

Main exclusion criteria: life expectancy < 4 months; active autoimmune disease requiring treatment with systemic immunosuppressive agents, e.g. inflammatory bowel disease, multiple sclerosis, sarcoidosis, psoriasis, autoimmune haemolytic anaemia, rheumatoid arthritis, systemic lupus erythematosus, vasculitis, Sjögren's syndrome, scleroderma and autoimmune hepatitis; treatment with systemic corticosteroids within 7 days before screening, known cardiomyopathy or clinical significant finding in electrocardiography at screening (or both); KPS < 70%.

Sample size planned:90.

Interventions

Group 1: intuvax + nephrectomy + sunitinib.

Group 0: nephrectomy + sunitinib.

Outcomes

Primary outcome: median OS from randomization for high‐risk patients, 18‐month OS in the intermediate‐risk mRCC patients.

Secondary outcomes: safety, PFS.

Starting date

April 2015.

Contact information

[email protected].

Notes

Estimated primary completion date: February 2018.

NCT02684006

Trial name or title

JAVELIN RENAL 101 ‐ A Phase 3, Multinational, Randomised, Open‐Label, Parallel‐Arm Study of Avelumab (MSB0010718C) in Combination with Axitinib (Inlyta(Registered)) versus Sunitinib (Sutent(Registered)) Monotherapy in the First‐Line Treatment of Patients with Advanced Renal Cell Carcinoma.

Methods

Study design: parallel‐arm, open‐label RCT.

Setting: multicentre, international phase III.

Countries: Japan, US.

Participants

Main inclusion criteria: aged ≥ 18 years; either gender; histologically or cytologically confirmed advanced or mRCC with clear‐cell component; availability of a recent formalin‐fixed, paraffin‐embedded tumour tissue block; ≥ 1 measurable lesion (RECIST 1.1) not previously irradiated; ECOG Performance Status 0 or 1; adequate bone marrow function, renal and liver functions.

Main exclusion criteria: prior systemic therapy directed at advanced or mRCC; prior adjuvant or neoadjuvant therapy for RCC if disease progression or relapse has occurred during or within 12 months after the last dose of treatment; prior immunotherapy with IL‐2, IFN‐α or anti‐PD‐1, anti‐PD‐L1, anti‐PD‐L2, anti‐CD137 or anti‐CTLA‐4 antibody (including ipilimumab) or any other antibody or drug specifically targeting T cell costimulation or immune checkpoint pathways; prior therapy with axitinib or sunitinib (or both) and any prior therapies with other VEGF pathway inhibitors; known severe hypersensitivity reactions to monoclonal antibodies (grade ≥ 3), any history of anaphylaxis or uncontrolled asthma; any of the following in the previous 6 months: myocardial infarction, severe/unstable angina, coronary/peripheral artery bypass graft, symptomatic congestive heart failure, cerebrovascular accident, transient ischaemic attack, deep vein thrombosis or symptomatic pulmonary embolism; vaccination within 4 weeks of the first dose of avelumab and while on trial is prohibited except for administration of inactivated vaccines.

Sample size planned: 583.

Interventions

Group 1: avelumab in combination with axitinib.

Group 0: sunitinib.

Outcomes

Primary outcome: PFS.

Secondary outcomes: OS, ORR, QoL, safety.

Starting date

March 2016.

Contact information

Pfizer CT.gov call centre.

Notes

Estimated primary completion date: June 2018.

NCT02781506

Trial name or title

Nivolumab and Stereotactic Ablative Radiation Therapy versus Nivolumab Alone for Metastatic Renal Cancer.

Methods

Study design: parallel‐arm, open‐label RCT.

Setting: national, phase II.

Countries: US.

Participants

Main inclusion criteria: aged 18 to 100 years, either gender, pathological diagnosis of metastatic RCC with clear‐cell component, measurable disease in at least 2 non‐radiated sites, progression or intolerance to ≥ 1 prior systemic antiangiogenic therapy, ECOG Performance Status 0, 1, 2 or 3, adequate organ and marrow function.

Main exclusion criteria: major surgery within 2 weeks prior to first dose; prior treatment with any anti‐PD‐1, anti‐PD‐L1, anti‐PD‐L2, anti‐CD137 or anti‐CTLA‐4 antibody or any other antibody or drug specifically targeting T‐cell costimulation or checkpoint pathways; active known or suspected autoimmune disease; history of hypersensitivity to monoclonal antibodies.

Sample size planned: 87.

Interventions

Group 1: nivolumab.

Group 0: nivolumab with radiation.

Outcomes

Primary outcome: ORR.

Secondary outcomes: OS, PFS, safety, other.

Starting date

June 2016.

Contact information

Raquibu Hannan, MD, PhD, tel: +1 214‐645‐8525.

Notes

Contact: Jean Wu, RN, MSN, OCN.

NCT02853331

Trial name or title

KEYNOTE‐426 ‐ Study to Evaluate the Efficacy and Safety of Pembrolizumab (MK‐3475) in Combination with Axitinib versus Sunitinib Monotherapy in Participants with Renal Cell Carcinoma.

Methods

Study design: parallel‐arm, open‐label RCT.

Setting: multicentre, international phase III.

Countries: Japan, Hungary, Spain, Korea, Russia, US.

Participants

Main inclusion criteria: aged ≥ 18 years, either gender, histologically or cytologically confirmed advanced or mRCC (stage IV) with clear‐cell component with or without sarcomatoid features, availability of an archival tumour tissue sample or fresh biopsy, measurable disease (RECIST 1.1), no previously systemic therapy, KPS ≥ 70%.

Main exclusion criteria: prior treatment with VEGF receptor or mTOR targeting agents; prior treatment with anti‐PD‐1, anti‐PD‐L1, anti‐PD‐L2 or other immunotherapy; known severe hypersensitivity reactions; active autoimmune disease; active infection; major surgery within 4 weeks prior to randomization; heart failure NYHA III or IV.

Sample size planned: 840.

Interventions

Group 1: pembrolizumab and axitinib.

Group 0: sunitinib.

Outcomes

Primary outcome: PFS, OS.

Secondary outcomes: ORR, safety.

Starting date

September 2016.

Contact information

Merck Sharp & Dohme Corp.

Notes

Estimated primary completion date: December 2019; EudraCT: 2016‐000588‐17.

AE: adverse event; AJCC: American Joint Committee on Cancer; CT: computer tomography; CTLA‐4: cytotoxic T‐lymphocyte antigen 4; ECOG: Eastern Cooperative Oncology Group; IL: interleukin; IFN‐α: interferon‐α; KPS: Karnovsky Performance Status; LLN: lower limit of normal; mRCC: metastatic renal cell carcinoma; MRI: magnetic resonance imaging; MSKCC: Memorial Sloan‐Kettering Cancer Center; mTOR: mammalian target of rapamycin; NCI‐CTC: National Cancer Institute Common Terminology Criteria; NCI CTCAE: National Cancer Institute Common Terminology Criteria for Adverse Events; NYHA: New York Heart Association; ORR: objective response rate; PD: programmed death; PD‐1: programmed death‐1; PFS: progression‐free survival; PSA: prostate‐specific antigen; QoL: quality of life; RCC: renal cell carcinoma; RCT: randomized controlled trial; RECIST: Response Evaluation Criteria In Solid tumours; SOC: standard of care; ULN: upper limit of normal; VEGF: vascular endothelial growth factor.

Data and analyses

Open in table viewer
Comparison 1. Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

2

1166

Risk Ratio (M‐H, Random, 95% CI)

1.30 [1.13, 1.51]

Analysis 1.1

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

2 Overall survival Show forest plot

2

Hazard Ratio (Random, 95% CI)

1.28 [1.11, 1.49]

Analysis 1.2

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

3 Quality of life Show forest plot

2

Mean Difference (Random, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Quality of life.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Quality of life.

3.1 FACT‐G

1

730

Mean Difference (Random, 95% CI)

‐5.58 [‐7.25, ‐3.91]

3.2 FKSI‐15

1

730

Mean Difference (Random, 95% CI)

‐3.27 [‐4.18, ‐2.36]

3.3 FKSI‐DRS

1

730

Mean Difference (Random, 95% CI)

‐1.98 [‐2.51, ‐1.45]

3.4 EQ‐5D

2

1000

Mean Difference (Random, 95% CI)

‐0.06 [‐0.12, ‐0.00]

3.5 EQ‐VAS

2

1000

Mean Difference (Random, 95% CI)

‐4.68 [‐6.53, ‐2.83]

4 Adverse events (grade ≥ 3) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Adverse events (grade ≥ 3).

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Adverse events (grade ≥ 3).

5 Progression‐free survival Show forest plot

2

Hazard Ratio (Random, 95% CI)

2.23 [1.79, 2.77]

Analysis 1.5

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Progression‐free survival.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Progression‐free survival.

6 Tumour remission Show forest plot

2

1007

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.12, 0.75]

Analysis 1.6

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 6 Tumour remission.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 6 Tumour remission.

Open in table viewer
Comparison 2. Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.1

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

2 Overall survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

Analysis 2.2

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

3 Adverse events (grade ≥ 3) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.3

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).

4 Progression‐free survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

Analysis 2.4

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.

5 Tumour remission Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.5

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.

Open in table viewer
Comparison 3. Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

2

1381

Risk Ratio (M‐H, Random, 95% CI)

1.17 [1.00, 1.36]

Analysis 3.1

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

2 Overall survival Show forest plot

2

Hazard Ratio (Random, 95% CI)

1.13 [1.00, 1.28]

Analysis 3.2

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

3 Adverse events (grade ≥ 3) Show forest plot

2

1350

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.71, 0.84]

Analysis 3.3

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).

4 Progression‐free survival Show forest plot

2

Hazard Ratio (Random, 95% CI)

1.53 [1.36, 1.73]

Analysis 3.4

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.

5 Tumour remission Show forest plot

2

1205

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.31, 0.50]

Analysis 3.5

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.

Open in table viewer
Comparison 4. Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.1

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

2 Adverse events (grade ≥ 3) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.2

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Adverse events (grade ≥ 3).

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Adverse events (grade ≥ 3).

3 Progression‐free survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

Analysis 4.3

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Progression‐free survival.

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Progression‐free survival.

4 Tumour remission Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.4

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Tumour remission.

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Tumour remission.

Open in table viewer
Comparison 5. Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

2

1034

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.91, 1.32]

Analysis 5.1

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

2 Overall survival Show forest plot

2

1071

Hazard Ratio (Fixed, 95% CI)

1.14 [0.96, 1.37]

Analysis 5.2

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

3 Adverse events (grade ≥ 3) Show forest plot

2

1065

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.97, 1.39]

Analysis 5.3

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).

4 Progression‐free survival Show forest plot

1

339

Hazard Ratio (Random, 95% CI)

1.05 [0.87, 1.27]

Analysis 5.4

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.

5 Tumour remission Show forest plot

2

1071

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.76, 1.13]

Analysis 5.5

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.

Open in table viewer
Comparison 6. Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

1

821

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.56, 0.87]

Analysis 6.1

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

2 Overall survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.73 [0.60, 0.89]

Analysis 6.2

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 2 Overall survival.

3 Quality of life Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 6.3

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 3 Quality of life.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 3 Quality of life.

3.1 Clinical important MID in FKSI‐DRS

1

704

Risk Ratio (M‐H, Random, 95% CI)

1.51 [1.28, 1.78]

3.2 Clinical important MID in EQ‐5D‐VAS

1

703

Risk Ratio (M‐H, Random, 95% CI)

1.37 [1.16, 1.61]

4 Adverse events (grade ≥ 3) Show forest plot

1

803

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.40, 0.65]

Analysis 6.4

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 4 Adverse events (grade ≥ 3).

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 4 Adverse events (grade ≥ 3).

5 Progression‐free survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.88 [0.75, 1.03]

Analysis 6.5

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 5 Progression‐free survival.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 5 Progression‐free survival.

6 Tumour remission Show forest plot

1

750

Risk Ratio (M‐H, Random, 95% CI)

4.39 [2.84, 6.80]

Analysis 6.6

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 6 Tumour remission.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 6 Tumour remission.

Study flow diagram.
Figures and Tables -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 2

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

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figures and Tables -
Figure 3

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

Forest plot of comparison: 1 Interferon‐α (IFN‐α) alone versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, outcome: 1.1 1‐year mortality.
Figures and Tables -
Figure 4

Forest plot of comparison: 1 Interferon‐α (IFN‐α) alone versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, outcome: 1.1 1‐year mortality.

Forest plot of comparison: 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, outcome: 3.1 1‐year mortality.
Figures and Tables -
Figure 5

Forest plot of comparison: 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, outcome: 3.1 1‐year mortality.

Forest plot of comparison: 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, outcome: 5.1 1‐year mortality.
Figures and Tables -
Figure 6

Forest plot of comparison: 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, outcome: 5.1 1‐year mortality.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.
Figures and Tables -
Analysis 1.1

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.
Figures and Tables -
Analysis 1.2

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Quality of life.
Figures and Tables -
Analysis 1.3

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Quality of life.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Adverse events (grade ≥ 3).
Figures and Tables -
Analysis 1.4

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Adverse events (grade ≥ 3).

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Progression‐free survival.
Figures and Tables -
Analysis 1.5

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Progression‐free survival.

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 6 Tumour remission.
Figures and Tables -
Analysis 1.6

Comparison 1 Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 6 Tumour remission.

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.
Figures and Tables -
Analysis 2.1

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.
Figures and Tables -
Analysis 2.2

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).
Figures and Tables -
Analysis 2.3

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.
Figures and Tables -
Analysis 2.4

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.
Figures and Tables -
Analysis 2.5

Comparison 2 Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.
Figures and Tables -
Analysis 3.1

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.
Figures and Tables -
Analysis 3.2

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).
Figures and Tables -
Analysis 3.3

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.
Figures and Tables -
Analysis 3.4

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.
Figures and Tables -
Analysis 3.5

Comparison 3 Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.
Figures and Tables -
Analysis 4.1

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Adverse events (grade ≥ 3).
Figures and Tables -
Analysis 4.2

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Adverse events (grade ≥ 3).

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Progression‐free survival.
Figures and Tables -
Analysis 4.3

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Progression‐free survival.

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Tumour remission.
Figures and Tables -
Analysis 4.4

Comparison 4 Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Tumour remission.

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.
Figures and Tables -
Analysis 5.1

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.
Figures and Tables -
Analysis 5.2

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).
Figures and Tables -
Analysis 5.3

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 3 Adverse events (grade ≥ 3).

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.
Figures and Tables -
Analysis 5.4

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 4 Progression‐free survival.

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.
Figures and Tables -
Analysis 5.5

Comparison 5 Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma, Outcome 5 Tumour remission.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 1 1‐year mortality.
Figures and Tables -
Analysis 6.1

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 1 1‐year mortality.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 2 Overall survival.
Figures and Tables -
Analysis 6.2

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 2 Overall survival.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 3 Quality of life.
Figures and Tables -
Analysis 6.3

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 3 Quality of life.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 4 Adverse events (grade ≥ 3).
Figures and Tables -
Analysis 6.4

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 4 Adverse events (grade ≥ 3).

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 5 Progression‐free survival.
Figures and Tables -
Analysis 6.5

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 5 Progression‐free survival.

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 6 Tumour remission.
Figures and Tables -
Analysis 6.6

Comparison 6 Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma, Outcome 6 Tumour remission.

Summary of findings for the main comparison. Interferon‐α alone versus standard targeted therapies (sunitinib or temsirolimus) in first‐line therapy of metastatic renal cell carcinoma

IFN‐α alone versus standard targeted therapy for mRCC

Patient population: previously untreated patients with mRCC

Settings: phase III, international, multicentre, open‐label

Intervention: IFN‐α alone

Comparison: standard targeted therapy (sunitinib or temsirolimus)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Risk with standard targeted therapy

Risk difference with IFN‐α alone (95% CI)

1‐year mortality
Follow‐up: 1 to 36 months

Lowa

RR 1.3
(1.13 to 1.51)

1166
(2 studies)

⊕⊕⊕⊝
Moderate1

150 per 1000

45 more per 1000
(from 20 more to 76 more)

Moderatea

280 per 1000

84 more per 1000
(from 36 more to 143 more)

Higha

550 per 1000

165 more per 1000
(from 71 more to 280 more)

QoL
FACT‐G
Follow‐up: median 17 weeks

The mean QoL in the control group was
82.3 pointsb

MD 5.58 lower
(7.25 to 3.91 lower)

730
(1 study)

⊕⊕⊝⊝
Low2,3

QoL
FKSI‐15
Follow‐up: median 17 weeks

The mean QoL in the control group was
45.3 pointsb

MD 3.27 lower

(4.18 to 2.36 lower)

730
(1 study)

⊕⊕⊝⊝
Low2,3

QoL
FKSI‐DRS
Follow‐up: median 17 weeks

The mean QoL in the control group was
29.4 pointsb

MD 1.98 lower

(2.51 to 1.46 lower)

730
(1 study)

⊕⊕⊝⊝
Low2,3

QoL
EQ‐5D
Follow‐up: range 12 to 17 weeks

The mean QoL in the control group was
0.711pointsb

MD 0.06 lower

(0.12 lower to 0 higher)

1002

(2 studies)

⊕⊕⊝⊝
Low2,3

QoL
EQ‐VAS
Follow‐up: range 12 to 17 weeks

The mean QoL in the control groups was
70.4pointsb

MD 4.68 lower

(6.53 to 2.83 lower)

1002

(2 studies)

⊕⊕⊝⊝
Low2,3

Adverse events (grade ≥ 3)
Follow‐up: 14 to 36 months

668 per 1000

114 more per 1000
(from 20 more to 214 more)

RR 1.17
(1.03 to 1.32)

408
(1 study)

⊕⊕⊝⊝
Low2,3

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; EQ‐5D: EuroQol 5‐Dimension; EQ‐VAS: EuroQol Visual Analogue Scale; FACT‐G: Functional Assessment of Cancer Therapy ‐ General; FKSI‐15: FACT‐Kidney Symptom Index; FKSI‐DRS: FACT‐Kidney Symptom Index Disease Related Symptoms; IFN‐α: interferon‐α; MD: mean difference; mRCC: metastatic renal cell carcinoma; QoL: quality of life; RR: risk ratio.

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

1 Downgraded for selection bias and performance bias due to cross‐over.
2 Downgraded for performance and detection bias.
3 Downgraded for imprecision due to wide confidence intervals; clinical action would differ between lower and upper boundary of the confidence interval.
a Moderate risk of 1‐year mortality from the SEER (Surveillance, Epidemiology, and End Results) Cancer Statistics Review (Howlader 2015), low risk from participants with favourable risk in Rini 2015, high risk from Hudes 2007.
b Mean postbaseline value on treatment.

Figures and Tables -
Summary of findings for the main comparison. Interferon‐α alone versus standard targeted therapies (sunitinib or temsirolimus) in first‐line therapy of metastatic renal cell carcinoma
Summary of findings 2. Interferon‐α combined with targeted therapies versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma

IFN‐α alone or combined with targeted therapy compared to standard targeted therapy in first‐line therapy of mRCC

Patient population: previously untreated patients with mRCC

Setting: phase III, international, multicentre, open‐label

Intervention: IFN‐α combined with targeted therapy

Comparison: standard targeted therapy (temsirolimus)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Risk with standard targeted therapy

Risk difference with IFN‐α combined with targeted therapy (95% CI)

1‐year mortality
Follow‐up: 14‐36 months

Lowa

RR 1.13
(0.95 to 1.34)

419
(1 study)

⊕⊕⊕⊝
Moderate1

150 per 1000

20 more per 1000
(from 8 fewer to 51 more)

Moderatea

280 per 1000

36 more per 1000
(from 14 fewer to 95 more)

Higha

550 per 1000

71 more per 1000
(from 28 fewer to 187 more)

Quality of life

No evidence available

Adverse events (grade ≥ 3)
Follow‐up: 14 to 36 months

668 per 1000

200 more per 1000
(from 114 more to 301 more)

RR 1.30
(1.17 to 1.45)

416
(1 study)

⊕⊕⊝⊝
Low1,2

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; IFN‐α: interferon‐α; mRCC: metastatic renal cell carcinoma; RR: risk ratio.

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

1 Downgraded for imprecision due to wide confidence intervals; clinical action would differ between lower and upper boundary of the confidence interval.
2 Downgraded for performance and detection bias.
a Moderate risk of 1‐year mortality from the SEER Cancer Statistics Review (Howlader 2015), low risk from participants with favourable risk in Rini 2015, high risk from Hudes 2007.

Figures and Tables -
Summary of findings 2. Interferon‐α combined with targeted therapies versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma
Summary of findings 3. Interferon‐α alone versus interferon‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma

IFN‐α alone versus IFN‐α + bevacizumab in first‐line therapy of mRCC

Patient population: previously untreated patient with mRCC

Setting: phase III, international, multicentre, Escudier 2007: double‐blind, placebo‐controlled; Rini 2010: open‐label

Intervention: IFN‐α alone

Comparison: IFN‐α alone + bevacizumab

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Risk with standard therapy (IFN‐α + bevacizumab)

Risk difference with IFN‐α alone (95% CI)

1‐year mortality
Follow‐up: 13.3 to 22 months

Low

RR 1.17
(1.00 to 1.36)

1381
(2 studies)

⊕⊕⊝⊝
Low1,2

150 per 1000

25 more per 1000
(from 0 more to 54 more)

Moderate

280 per 1000

48 more per 1000
(from 0 more to 101 more)

High

550 per 1000

93 more per 1000
(from 0 more to 198 more)

Quality of life

No evidence available

Adverse events (grade ≥ 3)

Follow‐up: up to 28 days after last dose to 65 months

705 per 1000

162 fewer per 1000
(from 113 fewer to 205 fewer)

RR 0.77
(0.71 to 0.84)

1350
(2 studies)

⊕⊕⊕⊝
Moderate3

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; IFN‐α: interferon‐α; mRCC: metastatic renal cell carcinoma; RR: risk ratio.

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

1 Downgraded for selection bias and performance bias due to substantial cross‐over.
2 Downgraded for imprecision due to wide confidence intervals; clinical action would differ between lower and upper boundary of the confidence interval.
3 Downgraded for performance and detection bias.

Figures and Tables -
Summary of findings 3. Interferon‐α alone versus interferon‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma
Summary of findings 4. Interferon‐α plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma

IFN‐α + bevacizumab versus targeted therapies in first‐line therapy of mRCC

Patient population: previously untreated patients with mRCC

Setting: phase II, national (France), multicentre, open‐label

Intervention: IFN‐α + bevacizumab

Comparison: standard targeted therapies (sunitinib)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Risk with standard targeted therapies

Risk difference with IFN‐α + bevacizumab (95% CI)

1‐year mortality
median

Follow‐up: 23.2 months

Lowa

RR 0.37
(0.13 to 1.08)

83
(1 study)

⊕⊕⊝⊝
Low1,2

150 per 1000

95 fewer per 1000
(131 fewer to 12 more)

Moderatea

280 per 1000

176 fewer per 1000
(from 244 fewer to 22 more)

Higha

550 per 1000

347 fewer per 1000
(from 479 fewer to 44 more)

Quality of life

No evidence available

Adverse events (grade ≥ 3)

Follow‐up: 48 weeks

595 per 1000

107 more per 1000
(from 89 fewer to 369 more)

RR 1.18
(0.85 to 1.62)

82
(1 study)

⊕⊕⊝⊝
Low2,3

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; mRCC: metastatic renal cell carcinoma; RR: risk ratio.

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

1 Downgraded for reporting and performance bias due to differences in second‐line treatment.
2 Downgraded for imprecision due to wide confidence intervals; clinical action would differ between lower and upper boundary of the confidence interval.
3 Downgraded for performance and detection bias.
a Moderate risk of 1‐year mortality from the SEER Cancer Statistics Review (Howlader 2015), low risk from participants with favourable risk in Rini 2015, high risk from Hudes 2007.

Figures and Tables -
Summary of findings 4. Interferon‐α plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma
Summary of findings 5. Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma

Vaccine treatment versus standard therapies in first‐line therapy of mRCC

Patient population: previously untreated patients with mRCC

Setting: phase III, international, multicentre, double‐blind, placebo‐controlled (Amato 2010), open‐label (Rini 2015)

Intervention: vaccine treatment (MVA‐5T4 or IMA0901)

Comparison: placebo and standard therapies (IL‐2, IFN‐α and sunitinib)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Risk with standard therapies

Risk difference with vaccine treatment (95% CI)

1‐year mortality
Follow‐up: 12 to 48 months

Lowa

RR 1.10
(0.91 to 1.32)

1034
(2 studies)

⊕⊕⊝⊝
Low1,2,3

150 per 1000

15 more per 1000
(from 13 fewer to 48 more)

Moderatea

280 per 1000

28 more per 1000
(from 25 fewer to 90 more)

Higha

550 per 1000

55 more per 1000
(from 49 fewer to 176 more)

Quality of life

No evidence available

Adverse events (grade ≥ 3)

Follow‐up: not reported

241 per 1000

39 more per 1000
(from 7 fewer to 94 more)

RR 1.16
(0.97 to 1.39)

1065
(2 studies)

⊕⊕⊝⊝
Low3,4,5

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; mRCC: metastatic renal cell carcinoma; RR: risk ratio.

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

1 Not downgraded for performance bias, borderline decision due to second‐line therapies in one study.
2 Downgraded for indirectness due to non‐standard therapies (low‐dose interleukin‐2, IFN‐α) in 75% participants of both treatment arms.
3 Downgraded for imprecision due to wide confidence intervals; clinical action would differ between lower and upper boundary of the confidence interval.
4 Downgraded for performance and detection bias.
5 Not downgraded for indirectness, borderline decision due to non‐standard therapies in both treatment arms.
a Moderate risk of 1‐year mortality from the SEER Cancer Statistics Review (Howlader 2015), low risk from participants with favourable risk in Rini 2015, high risk from Hudes 2007.

Figures and Tables -
Summary of findings 5. Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma
Summary of findings 6. Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma

Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with mRCC

Patient population: previously treated patients with mRCC

Setting: phase III, international, multicentre, open‐label

Intervention: targeted immunotherapy (nivolumab) alone

Comparison: standard targeted therapies (everolimus)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Risk with standard targeted therapies

Risk difference with targeted immunotherapy alone (95% CI)

1‐year mortality
Follow‐up: > 14 months

341 per 1000

102 fewer per 1000
(from 44 fewer to 150 fewer)

RR 0.70
(0.56 to 0.87)

821
(1 study)

⊕⊕⊕⊝
Moderate1

Quality of life: Clinically relevant improvement in FKSI‐DRS
Follow‐up: 1 to 104 weeks

367 per 1000

187 more per 1000

(from 103 more to 287 more)

RR 1.51 (1.28 to 1.78)

704
(1 study)

⊕⊕⊕⊝
Moderate2

Quality of life: clinically relevant improvement in EQ‐5D VAS
Follow‐up: 1 to 104 weeks

391 per 1000

145 more per 1000

(from 63 more to 238 more)

RR 1.37 (1.16‐1.61)

703

(1 study)

⊕⊕⊕⊝
Moderate2

Adverse events (grade ≥ 3)

365 per 1000

179 fewer per 1000
(from 128 fewer to 219 fewer)

RR 0.51
(0.40 to 0.65)

803
(1 study)

⊕⊕⊕⊝
Moderate2

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; EQ‐5D VAS: EuroQol 5‐Dimension Visual Analogue Scale; FKSI‐DRS: FACT‐Kidney Symptom Index Disease Related Symptoms; mRCC: metastatic renal cell carcinoma; RR: risk ratio.

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

1 Downgraded for performance bias due to cross‐over.
2 Downgraded for performance and detection bias.

Figures and Tables -
Summary of findings 6. Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma
Table 1. Overall survival

Study ID

Comparison (group 1 vs group 0)

Median OS (95% CI) (months)

1‐year mortality

Comments

Group 1

Group 0

Group 1

Group 0

Hudes 2007

1 (IFN‐α alone vs standard targeted therapies)

7.3 (6.1 to 8.8)

10.9 (8.6 to 12.7)

70%

53%

From curves.

Motzer 2007

21.8 (17.9 to 26.9)

26.4 (23.0 to 32.9)

12.3%

10.1%

Numbers reported.

Hudes 2007

2 (IFN‐α + targeted therapies vs standard targeted therapies)

8.4 (6.6 to 10.3)

10.9 (8.6 to 12.7)

60%

53%

From curves.

Escudier 2007

3 (IFN‐α alone vs IFN‐α + bevacizumab)

21.3

23.3

32%

26%

From curves.

Rini 2010

17.4 (14.4 to 20.0)

18.3 (16.5 to 22.5)

39%

34%

From curves with numbers and censoring marks.

Negrier 2011

4 (IFN‐α + bevacizumab vs standard targeted therapies)

Not reported

Not reported

10%

26%

Reported.

Amato 2010

5 (Vaccine treatment vs standard therapies)

19.2

20.1

35%

33%

From curves with censoring.

Rini 2015

33.1

Not reached

17%

22%

Numbers reported.

Motzer 2015a

6 (Targeted immunotherapy alone vs targeted standard therapy)

25.0 (21.8 to NE)

19.6 (17.6 to 23.1)

24%

34%

From curves with numbers with censoring marks.

CI: confidence interval; IFN‐α: interferon‐α; NE: not estimable; OS: overall survival.

Figures and Tables -
Table 1. Overall survival
Table 2. Overall survival subgroups

Comparison (group 1 vs group 0)

Study

Subgroup

Sample size

Treatment effects (95% CI) or P values

1 (IFN‐α alone vs standard targeted therapies)

Hudes 2007

Prior nephrectomy

278

HR 1.2 (0.9 to 1.6)

Motzer 2007

Prior nephrectomy

674

HR 1.2 (0.95 to 1.5)

Pooled

Prior nephrectomy

952

HR 1.2 (1.0 to 1.43), I2 = 0%

Hudes 2007

No prior nephrectomy

138

HR 1.7 (1.1 to 2.5)

Motzer 2007

No prior nephrectomy

76

HR 1.23 (0.8 to 2.1)

Pooled

No prior nephrectomy

214

HR 1.48 (1.1 to 2.0), I2 = 1%

Hudes 2007

KPS ≤ 70

340

HR 1.39 (1.11 to 1.75)

Hudes 2007

KPS > 70

75

HR 0.93 (0.53 to 1.67)

Motzer 2007

With poor risk

48

HR 1.15 (0.83 to 2.78)

Motzer 2007

Intermediate risk

421

HR 1.27 (1.00 to 1.62)

Motzer 2007

ECOG Performance Status 0

460

HR 1.1 (0.87 to 1.5)

Motzer 2007

ECOG Performance Status 1

290

HR 1.4 (1.05 to 1.7)

3 (IFN‐α alone vs IFN‐α + bevacizumab)

Escudier 2007

Favourable risk

180

HR 1.09 (0.73 to 1.61), P = 0.6798

Rini 2010

Favourable risk

192

HR 1.11 (0.8 to 1.56), P = 0.5189

Pooled

Favourable risk

372

HR 1.10 (0.85 to 1.43), I2 = 0%

Escudier 2007

Intermediate risk

392

HR 1.20 (0.95 to 1.54), P = 0.1230

Rini 2010

Intermediate risk

465

HR 1.15 (0.94 to 1.41), P = 0.1688

Pooled

Intermediate risk

857

HR 1.18 (1.01 to 1.37), I2 = 0%

Escudier 2007

Poor risk

59

HR 1.18 (0.68 to 2.04), P = 0.5594

Rini 2010

Poor risk

75

HR 1.33 (0.82 to 2.17), P = 0.2439

Pooled

Poor risk

124

HR 1.27 (0.88 to 1.82), I2 = 0%

Rini 2010

Prior nephrectomy

620

HR 1.10 (0.93 to 1.32), P = 0.2871

Rini 2010

No prior nephrectomy

112

HR 1.54 (1.02 to 2.27), P = 0.0381

5 (Vaccine treatment vs standard therapies)

Amato 2010

Favourable risk, treated with IL‐2 (SOC)

100

HR 0.54 (0.30 to 0.98), P = 0.046

Amato 2010

Favourable risk, treated with IFN‐α (SOC)

206

P > 0.05

Amato 2010

Good prognosis, treated with sunitinib (SOC)

119

P > 0.05

Amato 2010

Intermediate prognosis, treated with IL‐2 (SOC)

70

P > 0.05

Amato 2010

Intermediate prognosis, treated with IFN‐α (SOC)

169

P > 0.05

Amato 2010

Intermediate prognosis, treated with sunitinib (SOC)

65

P > 0.05

Rini 2015

Favourable risk (n = 92)

92

HR 0.82, P = 0.59

Rini 2015

Intermediate risk (n = 240)

240

HR 1.52, P < 0.05

6 (Targeted immunotherapy alone vs standard targeted therapies)

Motzer 2015a

Favourable risk group (MSKCC risk group)

293

HR 0.89 (0.59 to 1.32)

Motzer 2015a

Intermediate risk group (MSKCC risk group)

404

HR 0.76 (0.58 to 0.99)

Motzer 2015a

Poor risk group (MSKCC risk group)

124

HR 0.47 (0.30 to 0.73)

Motzer 2015a

1 previous antiangiogenic regimen

591

HR 0.71 (0.56 to 0.90)

Motzer 2015a

2 previous antiangiogenic regimens

230

HR 0.89 (0.61 to 1.29)

CI: confidence interval; ECOG: Eastern Cooperative Oncology Group; HR: hazard ratio; IFN‐α: interferon‐α; IL: interleukin; KPS: Karnovsky Performance Score; MSKCC: Memorial Sloan‐Kettering Cancer Center; n: number of participants; SOC: standard of care.

Figures and Tables -
Table 2. Overall survival subgroups
Table 3. Quality of life

Comparison (group 1 vs group 0)

Study (reported in)

Measurement instrument

Group 1

Group 0

Favours

Difference (95% CI) or P values

1 (IFN‐α alone vs standard targeted therapies)

Motzer 2007 (reported in Cella 2008)

FACT‐G total score, mean postbaseline score over 17 weeks

76.8 (n = 357)

82.3 (n = 373)

Group 0

‐5.58 (‐7.25 to ‐3.91)

Motzer 2007 (reported in Cella 2008)

FKSI‐15, mean postbaseline score over 17 weeks

42.1 (n = 357)

45.3 (n = 373)

Group 0

‐3.27 (‐4.18 to ‐2.36)

Motzer 2007 (reported in Cella 2008)

FKSI‐DRS, mean postbaseline score over 17 weeks

27.4 (n = 357)

29.4 (n = 373)

Group 0

‐1.98 (‐2.51 to ‐1.46)

Hudes 2007, (reported in Yang 2010)

EQ‐5D Index, mean score on treatment

0.492 (n = 115)

0.590 (n = 157)

Group 0

‐0.099 (95% CI ‐0.162 to ‐0.036)

Motzer 2007 (reported in Cella 2008)

EQ‐5D Index, mean postbaseline score over 17 weeks

0.725 (n = 357)

0.762 (n = 373)

Group 0

‐0.0364 (‐0.0620 to ‐0.0109)

Pooled EQ‐5D

472

530

Group 0

‐0.06 (‐0.12 to 0), I2 = 69%

Hudes 2007, (reported in Yang 2010)

EQ‐VAS, mean score on treatment

58.83 (n = 115)

63.33 (n = 157)

Group 0

‐4.50 (‐8.184 to ‐0.819)

Motzer 2007, (reported in Cella 2008)

EQ‐VAS, mean postbaseline score over 17 weeks

68.7 (n = 357)

73.4 (n = 373)

Group 0

‐4.74 (‐6.87 to ‐2.60)

Pooled EQ‐VAS

472

530

Group 0

‐4.68 (‐6.53 to ‐2.83), I2 = 0%

6 (Targeted immunotherapy alone vs standard targeted therapies)

Motzer 2015a (reported in Cella 2016)

FKSI‐DRS, mean score at baseline

30.2 ± 4.4 (n = 362)

30.1 ± 4.8 (n = 344)

Difference in mean change 1.6 (1.4 to 1.9), P < 0.0001

FKSI‐DRS, mean change from baseline to week 28

0.4 ± 5 (n = 164)

‐1.2 ± 4 (n = 122)

Group 1

FKSI‐DRS, mean change from baseline to week 52

1.6 ± 4 (n = 97)

‐1.0 ± 6 (n = 63)

Group 1

FKSI‐DRS, mean change from baseline to week 104

3.5 ± 4.1 (n = 20)

0.2 ± 6 (n = 9)

Group 1

Clinically important improvement from baseline by ≥ 2 FKSI‐DRS points

200 (55%)/361

126 (37%)/343

Group 1

RR 1.51 (1.28 to 1.78); P < 0.0001

Time to clinically important improvement ≥ 2 FKSI‐DRS points

Median: 4.7 months (3.7 to 7.5)

Median not reached

Group 1

HR 1.66 (1.33 to 2.08); P < 0.0001

Clinically important improvement from baseline by ≥ 3 FKSI‐DRS points

148 (41%)/361

95 (28%)/343

Group 1

RR 1.48 (1.20 to 1.83); P = 0.0002

Time to clinically important improvement ≥ 3 FKSI‐DRS points

Median not estimable

Median not estimable

Group 1

HR 1.61 (1.24 to 2.09); P < 0.0003

EQ‐5D utility index, mean score at baseline

0.78 ± 0.24 (n = 362)

0.78 ± 0.21 (n = 344)

No significant differences in proportion of participants with clinical important improvements (P = 0.070) or time to improvement (P = 0.86).

EQ‐5D utility index, mean change from baseline to week 28

0.052 ± 0.22 (n = 164)

‐0.03 ± 0.2 (n = 122)

Group 1

EQ‐5D utility index, mean change from baseline to week 52

0.06 ± 0.1 (n = 98)

‐0.01 ± 0.2 (n = 63)

Group 1

EQ‐5D utility index, mean change from baseline to week 104

0.13 ± 0.7 (n = 20)

‐0.02 ± 0.15 (n = 9)

Group 1

EQ‐5D VAS, mean score at baseline

73.3 ± 18.5 (n = 362)

72.5 ± 18.7 (n = 344)

EQ‐5D VAS, mean change from baseline to week 28

5 ± 13 (n = 164)

‐3 ± 11 (n = 122)

Group 1

EQ‐5D VAS, mean change from baseline to week 52

7 ± 15 (n = 98)

‐2 ± 16 (n = 63)

Group 1

EQ‐5D VAS, mean change from baseline to week 104

9 ± 9 (n = 20)

1 ± 18 (n = 9)

Group 1

Clinically important improvement from baseline by ≥ 7 EQ‐5D VAS points

192 (53%)/360

134(39%)/343

RR 1.37 (1.16 to 1.61); P = 0.0001

Time to clinically important improvement

Median: 6.5 months (3.9 to 12.2)

Median: 23.1 months (15.4 to not estimated)

HR 1.37 (1.10 to 1.71)

CI: confidence interval; EQ‐5D Index: EuroQol 5‐Dimension (MID 0.06 to 0.08, Pickard 2007); EQ‐VAS: EuroQol Visual Analog Scale (MID 7, Pickard 2007); FACT‐G: Functional Assessment of Cancer Therapy ‐ General (MID 4 points for better rating and 8 points for worse rating, Ringash 2007); FKSI‐15: FACT‐Kidney Symptom Index (MID 3 points, Cella 1997); FKSI‐DRS: FACT‐Kidney Symptom Index Disease Related Symptoms (MID 2 points, Cella 1997); HR: hazard ratio; MID: minimal important difference; n: number of participants.

Figures and Tables -
Table 3. Quality of life
Comparison 1. Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

2

1166

Risk Ratio (M‐H, Random, 95% CI)

1.30 [1.13, 1.51]

2 Overall survival Show forest plot

2

Hazard Ratio (Random, 95% CI)

1.28 [1.11, 1.49]

3 Quality of life Show forest plot

2

Mean Difference (Random, 95% CI)

Subtotals only

3.1 FACT‐G

1

730

Mean Difference (Random, 95% CI)

‐5.58 [‐7.25, ‐3.91]

3.2 FKSI‐15

1

730

Mean Difference (Random, 95% CI)

‐3.27 [‐4.18, ‐2.36]

3.3 FKSI‐DRS

1

730

Mean Difference (Random, 95% CI)

‐1.98 [‐2.51, ‐1.45]

3.4 EQ‐5D

2

1000

Mean Difference (Random, 95% CI)

‐0.06 [‐0.12, ‐0.00]

3.5 EQ‐VAS

2

1000

Mean Difference (Random, 95% CI)

‐4.68 [‐6.53, ‐2.83]

4 Adverse events (grade ≥ 3) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5 Progression‐free survival Show forest plot

2

Hazard Ratio (Random, 95% CI)

2.23 [1.79, 2.77]

6 Tumour remission Show forest plot

2

1007

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.12, 0.75]

Figures and Tables -
Comparison 1. Interferon‐α (IFN‐α) alone versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma
Comparison 2. Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2 Overall survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

3 Adverse events (grade ≥ 3) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4 Progression‐free survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

5 Tumour remission Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Figures and Tables -
Comparison 2. Interferon‐α (IFN‐α) combined with targeted therapy versus standard targeted therapy in first‐line therapy of metastatic renal cell carcinoma
Comparison 3. Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

2

1381

Risk Ratio (M‐H, Random, 95% CI)

1.17 [1.00, 1.36]

2 Overall survival Show forest plot

2

Hazard Ratio (Random, 95% CI)

1.13 [1.00, 1.28]

3 Adverse events (grade ≥ 3) Show forest plot

2

1350

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.71, 0.84]

4 Progression‐free survival Show forest plot

2

Hazard Ratio (Random, 95% CI)

1.53 [1.36, 1.73]

5 Tumour remission Show forest plot

2

1205

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.31, 0.50]

Figures and Tables -
Comparison 3. Interferon (IFN‐α) alone versus IFN‐α plus bevacizumab in first‐line therapy of metastatic renal cell carcinoma
Comparison 4. Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2 Adverse events (grade ≥ 3) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3 Progression‐free survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

4 Tumour remission Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Figures and Tables -
Comparison 4. Interferon‐α (IFN‐α) plus bevacizumab versus standard targeted therapies in first‐line therapy of metastatic renal cell carcinoma
Comparison 5. Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

2

1034

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.91, 1.32]

2 Overall survival Show forest plot

2

1071

Hazard Ratio (Fixed, 95% CI)

1.14 [0.96, 1.37]

3 Adverse events (grade ≥ 3) Show forest plot

2

1065

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.97, 1.39]

4 Progression‐free survival Show forest plot

1

339

Hazard Ratio (Random, 95% CI)

1.05 [0.87, 1.27]

5 Tumour remission Show forest plot

2

1071

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.76, 1.13]

Figures and Tables -
Comparison 5. Vaccine treatment versus standard therapies in first‐line therapy of metastatic renal cell carcinoma
Comparison 6. Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 1‐year mortality Show forest plot

1

821

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.56, 0.87]

2 Overall survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.73 [0.60, 0.89]

3 Quality of life Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 Clinical important MID in FKSI‐DRS

1

704

Risk Ratio (M‐H, Random, 95% CI)

1.51 [1.28, 1.78]

3.2 Clinical important MID in EQ‐5D‐VAS

1

703

Risk Ratio (M‐H, Random, 95% CI)

1.37 [1.16, 1.61]

4 Adverse events (grade ≥ 3) Show forest plot

1

803

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.40, 0.65]

5 Progression‐free survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.88 [0.75, 1.03]

6 Tumour remission Show forest plot

1

750

Risk Ratio (M‐H, Random, 95% CI)

4.39 [2.84, 6.80]

Figures and Tables -
Comparison 6. Targeted immunotherapy alone versus standard targeted therapies in previously treated patients with metastatic renal cell carcinoma