Scolaris Content Display Scolaris Content Display

Tratamiento dirigido para el carcinoma metastásico de células renales

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Eichelberg 2015 {published data only}

Eichelberg C, Goebell PJ, Vervenne WL, De Santis M, Fischer von Weikersthal L, Lerchenmüller C, et al. Updated overall survival, multivariate, and Q-TWiST analyses of a randomized, sequential, open-label study (SWITCH) to evaluate the efficacy and safety of sorafenib (So)-sunitinib (Su) versus Su-So in the treatment of metastatic renal cell cancer. Annals of Oncology 2014;25(suppl 4):iv290. CENTRAL [DOI: 10.1093/annonc/mdu337.28]
*
*. Eichelberg C, Vervenne WL, De Santis M, Fischer von Weikersthal L, Goebell PJ, Lerchenmüller C, et al. SWITCH: a randomised, sequential, open-label study to evaluate the efficacy and safety of sorafenib-sunitinib versus sunitinib-sorafenib in the treatment of metastatic renal cell cancer. European Urology 2015;68(5):837-47. CENTRAL [PMID: 25952317]
Goebell PJ, Vervenne W, De Santis M, Fischer Von Weikersthal L, Lerchenmuller CA, Zimmermann U, et al. Subgroup analyses of a randomized sequential open-label study (SWITCH) to evaluate efficacy and safety of sorafenib (SO)/sunitinib (SU) versus SU/SO in the treatment of metastatic renal cell cancer (mRCC). Journal of Clinical Oncology 2014;32(15 suppl):4567. CENTRAL [DOI: 10.1200/jco.2014.32.15_suppl.4567]
Michel MS, Vervenne W, de Santis M, Fischer von Weikersthal L, Goebell PJ, Lerchenmueller J, et al. SWITCH: A randomized sequential open-label study to evaluate efficacy and safety of sorafenib (SO)/sunitinib (SU) versus SU/SO in the treatment of metastatic renal cell cancer (mRCC). Journal of Clinical Oncology 2014;32(4 suppl):393. CENTRAL [DOI: 10.1200/jco.2014.32.4_suppl.393]
NCT00732914. Sequential study to treat renal cell carcinoma [A phase III randomized sequential open-label study to evaluate the efficacy and safety of sorafenib followed by sunitinib versus sunitinib followed by sorafenib in the treatment of first-line advanced / metastatic renal cell carcinoma]. clinicaltrials.gov/ct2/show/NCT00732914 (first received 12 August 2008). CENTRAL [NCT00732914]

Escudier 2010 {published data only}

Bajetta E, Ravaud A, Bracarda S, Négrier S, Szczylik C, Bellmunt J, et al. Efficacy and safety of first-line bevacizumab (BEV) plus interferon-α2a (IFN) in patients (pts) ≥65 years with metastatic renal cell carcinoma (mRCC). Journal of Clinical Oncology 2008;26(15 suppl):5095. CENTRAL [DOI: 10.1200/jco.2008.26.15_suppl.5095]
Bellmunt J, Melichar B, Bracarda S, Negrier SN, Ravaud A, Laeufle R, et al. Bevacizumab (BEV) and interferon (IFN) therapy does not increase risk of cardiac events in metastatic renal cell carcinoma(mRCC). European Journal of Cancer Supplements 2009;7(2):429. CENTRAL [DOI: 10.1016/S1359-6349(09)71454-5]
Bracarda S, Bellmunt J, Negrier SN, Melichar B, Ravaud A, Jethwa S, et al. What is the impact of subsequent antineoplastic therapy on overall survival (OS) following first-line bevacizumab (BEV)/interferonalpha2a (IFN) in metastatic renal cell carcinoma (mRCC)? – Experience from AVOREN. European Journal of Cancer Supplements 2009;7(2):431. CENTRAL [DOI: 10.1016/S1359-6349(09)71459-4]
Bracarda S, Koralewski P, Pluzanska A, Ravaud A, Szczylik C, Chevreau C, et al. Bevacizumab/interferon-alpha2a provides a progression-free survival benefit in all prespecified patient subgroups as first-line treatment of metastatic renal cell carcinoma (AVOREN). European Journal of Cancer Supplements 2007;5(4):281-2. CENTRAL [DOI: 10.1016/S1359-6349(07)71076-5]
Escudier B, Koralewski P, Pluzanska A, Ravaud A, Bracarda S, Szczylik C, et al. A randomized, controlled, double-blind phase III study (AVOREN) of bevacizumab/interferon-a2a vs placebo/interferon-α2a as first-line therapy in metastatic renal cell carcinoma. Journal of Clinical Oncology 2007;25(18 suppl):3. CENTRAL [DOI: 10.1200/jco.2007.25.18_suppl.3]
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 randomized double-blind phase III trial. Lancet 2007;370(9605):2103-11. CENTRAL [PMID: 18156031]
*
*. Escudier BB, 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. CENTRAL [PMID: 20368553]
Escudier BJ, Bellmunt J, Negrier S, Melichar B, Bracarda S, Ravaud A, et al. Final results of the phase III, randomized, double-blind AVOREN trial of first-line bevacizumab (BEV) + interferon-α2a (IFN) in metastatic renal cell carcinoma (mRCC). Journal of Clinical Oncology 2009;27(15 suppl):5020. CENTRAL [DOI: 10.1200/jco.2009.27.15_suppl.5020]
Escudier BJ, Negrier S, Chevreau C, et al. Analysis of prognostic risk categories in the phase III AVOREN trial of first-line bevacizumab plus interferon-α2a in patients with metastatic renal cell carcinoma: French prognostic scoring system. In: Genitourinary Cancers Symposium, ASCO, abst 417. 2010. CENTRAL
Escudier BJ, Ravaud A, Negrier S, Szczylik C, Bellmunt J, Bracarda S, et al. Update on AVOREN trial in metastatic renal cell carcinoma (mRCC): Efficacy and safety in subgroups of patients (pts) and pharmacokinetic (PK) analysis. Journal of Clinical Oncology 2008;26(15 suppl):5025. CENTRAL [DOI: 10.1200/jco.2008.26.15_suppl.5025]
Karakiewicz P, Sun M, Sneller V, Escudier B. Use of a nomogram to quantify overall survival (OS) benefit in patients with metastatic renal cell carcinoma receiving bevacizumab (BEV) with interferon (IFN) versus IFN alone. Journal of Clinical Oncology 2010;28(15 suppl):4592. CENTRAL [DOI: 10.1200/jco.2010.28.15_suppl.4592 ]
Karakiewicz PI, Sun M, Sneller V, et al. Use of a nomogram to quantify progression-free survival benefit in metastatic renal cell carcinoma patients receiving bevacizumab plus interferon or interferon alone. In: Genitourinary Cancers Symposium, ASCO, abstr 392. 2010. CENTRAL
Melichar B, Koralewski P, Pluzanska A, Ravaud A, Bracarda S, Szczylik C, et al. First-line bevacizumab improves progression-free survival with lower doses of interferon-α2a in the treatment of patients with metastatic renal cell carcinoma (AVOREN). European Journal of Cancer Supplements 2007;5(4):304. CENTRAL [DOI: 10.1016/S1359-6349(07)71149-7]
Melichar B, Koralewski P, Ravaud A, Pluzanska A, Bracarda S, Szczylik C, et al. First-line bevacizumab combined with reduced dose interferon-α2a is active in patients with metastatic renal cell carcinoma. Annals of Oncology 2008;19(8):1470-6. CENTRAL [PMID: 18408224]
NCT00738530. A study of avastin (bevacizumab) added to interferon alfa-2a (roferon) therapy in patients with metastatic renal cell cancer with nephrectomy [A randomised, double-blind study to evaluate the efficacy and safety of avastin plus roferon compared with placebo plus roferon on overall survival and tumor assessment in nephrectomised patients with metastatic clear cell renal cell carcinoma]. clinicaltrials.gov/ct2/show/NCT00738530 (first received 20 August 2008). CENTRAL [NCT00738530]

Escudier 2017 {published data only}

Cella D, Grunwald V, Escudier B, Hammers HJ, George S, Nathan P, et al. Patient-reported outcomes of patients with advanced renal cell carcinoma treated with nivolumab plus ipilimumab versus sunitinib (CheckMate 214): a randomised, phase 3 trial. Lancet Oncology 2019;20(2):297-310. CENTRAL [PMID: 30658932]
Escudier B, Tannir N, McDermott DF, Frontera OA, Melichar B, Plimack ER, et al. CheckMate 214: Efficacy and safety of nivolumab + ipilimumab (N+I) v sunitinib (S) for treatment-naive advanced or metastatic renal cell carcinoma (mRCC), including IMDC risk and PD-L1 expression subgroups. Annals of Oncology 2017;28(suppl 5):621-2. CENTRAL [DOI: 10.1093/annonc/mdx440.029]
Motzer RJ, Rini BI, McDermott DF, Aren Frontera O, Hammers HJ, Carducci MA, et al. Nivolumab plus ipilimumab versus sunitinib in first-line treatment for advanced renal cell carcinoma: extended follow-up of efficacy and safety results from a randomised, controlled, phase 3 trial. Lancet Oncology 2019;S1470-2045(19):30413-9. CENTRAL [PMID: 31427204]
*
*. Motzer RJ, Tannir N, McDermott DF, Frontera OA, Melichar B, Choueiri TK, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. New England Journal of Medicine 2018;378(14):1277-90. CENTRAL [PMID: 29562145]
NCT02231749. Nivolumab combined with ipilimumab versus sunitinib in previously untreated advanced or metastatic renal cell carcinoma (CheckMate 214) [A phase 3, randomized, open-label study of nivolumab combined with ipilimumab versus sunitinib monotherapy in subjects with previously untreated, advanced or metastatic renal cell carcinoma]. clinicaltrials.gov/ct2/show/NCT02231749 (first received 4 September 2014). CENTRAL [NCT02231749]
Tannir NM, Hammers HJ, Amin A, Grimm M-O, Rini BI, Mekan S, et al. Characterization of the benefit-risk profile of nivolumab + ipilimumab (N+I) v sunitinib (S) for treatment-naïve advanced renal cell carcinoma (aRCC; CheckMate 214). Journal of Clinical Oncology 2018;28(6 suppl):686. CENTRAL [DOI: 10.1200/JCO.2018.36.6_suppl.686]
Vyas C, Motzer RJ, Tannir NM, McDermott DF, Frontera OA, Melichar B, et al. Nivolumab + ipilimumab (N+I) vs sunitinib (S) for treatment-naive advanced or metastatic renal cell carcinoma (aRCC): results from checkmate 214, including overall survival by subgroups. Journal of Oncology Pharmacy Practice 2018;24(5_suppl):17-8. CENTRAL

Hudes 2007 {published data only}

Alemao E, Rajagopalan S, Yang S, et al. Quality adjusted survival in randomized, controlled trials: application of inverse probability weighting in renal cell cancer. In: Genitourinary Cancers Symposium, ASCO, abstr 399. 2010. CENTRAL
Armstrong AJ, George DJ, Halabi S. Serum lactate dehydrogenase as a biomarker for survival with mTOR inhibition in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology 2010;28(15 suppl):4361. CENTRAL [DOI: 10.1200/jco.2010.28.15_suppl.4631]
Dutcher JP, Szczylik C, Tannir N, Benedetto P, Ruff P, Hsu A, et al. Correlation of survival with tumor histology, age, and prognostic risk group for previously untreated patients with advanced renal cell carcinoma receiving temsirolimus or interferon-alpha. Journal of Clinical Oncology 2007;25(18 suppl):5033. CENTRAL [DOI: 10.1200/jco.2007.25.18_suppl.5033]
Dutcher JP, de Souza P, Figlin R, et al. Effect of temsirolimus versus interferon- on survival of patients with advanced renal cell carcinoma of different histologies. In: Genitourinary Cancers Symposium, ASCO, abstr 384. 2008. CENTRAL
Dutcher JP, de Souza P, McDermott D, Figlin RA, Berkenblit A, Thiele A, et al. Effect of temsirolimus versus interferon- on outcome of patients with advanced renal cell carcinoma of different histologies. Medical Oncology 2009;26(2):202–9. CENTRAL [PMID: 19229667]
Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, et al. A phase 3, randomized, 3-arm study of temsirolimus or interferon alpha or the combination in the treatment of first-line, poor-risk patients with advanced renal cell carcinoma. Journal of Clinical Oncology 2006;24(18 suppl):LBA4. CENTRAL [DOI: 10.1200/jco.2006.24.18_suppl.lba4]
*
*. 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 Medicine 2007;356(22):2271–81. CENTRAL [PMID: 17538086]
Logan T, McDermott D, Dutcher J, et al. Exploratory analysis of the influence of nephrectomy status on temsirolimus efficacy in patients with advanced renal cell carcinoma and poor-risk features. In: Genitourinary Cancers Symposium, ASCO, abstr 281. 2009. CENTRAL
NCT00065468. Study Evaluating Interferon And CCI-779 In Advanced Renal Cell Carcinoma (ARCC). clinicaltrials.gov/ct2/show/results/NCT00065468 (first received July 25 2003). CENTRAL [NCT00065468]
Pablo M, Hudes G, Dutcher J, White C, Krygowski M, Cincotta M, et al. Radiographic findings of drug-induced pneumonitis and clinical correlation inpatients with advanced renal cell carcinoma treated with temsirolimus. European Journal of Cancer Supplements 2009;7(2):426-7. CENTRAL [DOI: 10.1016/S1359-6349(09)71446-6]
Parasuraman S, Hudes G, Levy D, Strahs A, Moore L, De Marinise R, et al. Comparison of quality-adjusted survival in patients with advanced renal cell carcinoma receiving first-line treatment with temsirolimus (TEMSR) or interferon- (IFN) or the combination of IFN+TEMSR. Journal of Clinical Oncology 2007;25(18 suppl):5049. CENTRAL [DOI: 10.1200/jco.2007.25.18_suppl.5049]
Yang S, Hudes G, de Souza P, Alemao E, Strahs A, Purvis J. Evaluation of quality of life in patients with advanced renal cell carcinoma treated with temsirolimus vs interferon-alfa: results from a phase III randomized trial. European Journal of Cancer Supplements 2009;7(2):433. CENTRAL [DOI: 10.1016/S1359-6349(09)71467-3]
Zbrozek AS, Hudes G, Levy D, Strahs A, Berkenblit A, DeMarinis R, et al. Q-TWiST analysis of patients receiving temsirolimus or interferon alpha for treatment of advanced renal cell carcinoma. Pharmacoeconomics 2010;28(7):577–84. CENTRAL [PMID: 20550223]

McDermott 2018 {published data only}

Atkins MB, McDermott DF, Powles T, Motzer RJ, Rini BI, Fong L, et al. IMmotion150: A phase II trial in untreated metastatic renal cell carcinoma (mRCC) patients (pts) of atezolizumab (atezo) and bevacizumab (bev) vs and following atezo or sunitinib (sun). Journal of Clinical Oncology 2017;35(15 suppl):4505. CENTRAL [DOI: 10.1200/JCO.2017.35.15_suppl.4505]
McDermott DF, Atkins MB, Motzer RJ, Rini BI, Escudier BJ, Fong L, et al. A phase II study of atezolizumab (atezo) with or without bevacizumab (bev) versus sunitinib (sun) in untreated metastatic renal cell carcinoma (mRCC) patients (pts). Journal of Clinical Oncology 2017;35(6 suppl):431. CENTRAL [DOI: 10.1200/JCO.2017.35.6_suppl.431]
*
*. McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nature Medicine 2018;24(6):749-57. CENTRAL [PMID: 29867230]
NCT01984242. A study of atezolizumab (an engineered anti-programmed death-ligand 1 [PD-L1] antibody) as monotherapy or in combination with bevacizumab (avastin®) compared to sunitinib (Sutent®) in participants with untreated advanced renal cell carcinoma (IMmotion150) [A phase II, randomized study of atezolizumab (anti-PD-L1 antibody) administered as monotherapy or in combination with bevacizumab versus sunitinib in patients with untreated advanced renal cell carcinoma]. clinicaltrials.gov/ct2/show/NCT01984242 (first received 14 November 2013). CENTRAL [NCT01984242]
Pal SK, McDermott DF, Atkins MB, Escudier B, Rini BI, Motzer RJ, et al. Patient-reported outcomes in a phase 2 study comparing atezolizumab alone or with bevacizumab vs sunitinib in previously untreated metastatic renal cell carcinoma. BJU Int 2020;126:73-82. CENTRAL [DOI: 10.1111/bju.15058]
PowlesT, McDermott DF, Rini B, Motzer RJ, Atkins MB, Fong L, et al. IMmotion150: Novel radiological endpoints and updated data from a randomized phase II trial investigating atezolizumab (atezo) with or without bevacizumab (bev) vs sunitinib (sun) in untreated metastatic renal cell carcinoma (mRCC). Annals of Oncology 2017;28(suppl 5):624. CENTRAL [DOI: 10.1093/annonc/mdx440.033]

Motzer 2010 {published data only}

Castellano D, del Muro XG, Pérez-Gracia JL, González-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 [PMID: 19549706]
Cella D, Cappelleri JC, Bushmakin A, Charbonneau C, Li JZ, Kim ST, et al. Quality of life predicts progression-free survival in patients with renal cell carcinoma treated with sunitinib versus interferon alfa. Journal of Oncology Practice 2009;5(2):66-70. CENTRAL [PMID: 20856722]
Cella D, Li JZ, Cappelari 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 [PMID: 18669464]
Cella D, Li JZ, Cappelleri JC, Bushmakin A, Charbonneau C, Kim ST, Chen I, et al. Quality of life (QOL) predicts for progression-free survival (PFS) in patients with metastatic renal cell carcinoma (mRCC) treated with sunitinib compared to interferon-alpha (IFN-α). Journal of Clinical Oncology 2007;25(18 suppl):6594. CENTRAL [DOI: 10.1200/jco.2007.25.18_suppl.6594]
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-a in a phase III trial: final results and geographical analysis. British Journal of Cancer 2010;102(4):658-64. CENTRAL [PMID: 20104222]
Figin RA, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Negrier S, et al. Overall survival with sunitinib versus interferon (IFN)-alfa as first-line treatment of metastatic renal cell carcinoma (mRCC). Journal of Clinical Oncology 2008;26(15 suppl):5024. CENTRAL [DOI: 10.1200/jco.2008.26.15_suppl.5024]
Motzer RJ, Bukowski RM, Figlin RA, Hutson TE, Michaelson MD, Kim ST, et al. Prognostic nomogram for sunitinib in patients with metastatic renal cell carcinoma. Cancer 2008;113(7):1552-8. CENTRAL [PMID: 18720362]
Motzer RJ, Figlin RA, Hutson TE, Tomczak P, Bukowski RM, Rixe O, et al. Sunitinib versus interferon-alfa (IFN-α) as first-line treatment of metastatic renal cell carcinoma (mRCC): Updated results and analysis of prognostic factors. Journal of Clinical Oncology 2007;25(18 suppl):5024. CENTRAL [DOI: 10.1200/jco.2007.25.18_suppl.5024]
Motzer RJ, Figlin RA, Hutson TE, Tomczak P, Bukowski RM, Rixe O, et al. Sunitinib versus interferon-alfa as first-line treatment of metastatic renal cell carcinoma: updated results and analysis of prognostic factors. In: www.asco.org/virtual meeting. 2007. CENTRAL
*
*. 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 [PMID: 19487381]
Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Phase III randomized trial of sunitinib malate (SU11248) versus interferon-alfa (IFN-α) as first-line systemic therapy for patients with metastatic renal cell carcinoma (mRCC). Journal of Clinical Oncology 2006;24(18 suppl):LBA3. CENTRAL [DOI: 10.1200/jco.2006.24.18_suppl.lba3]
Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, 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 [PMID: 17215529]
Motzer RJ, Michaelson MD, Hutson TE, Tomczak P, Bukowski RM, Rixe O, et al. Sunitinib versus interferon (IFN)-alfa as first-line treatment of metastatic renal cell carcinoma (mRCC): updated efficacy and safety results and further analysis of prognostic factors. European Journal of Cancer Supplements 2007;5(4):301. CENTRAL [DOI: 10.1016/S1359-6349(07)71140-0]
NCT00083889. SU011248 versus interferon-alfa as first-line systemic therapy for patients with metastatic renal cell carcinoma [A phase 3, randomized study of SU011248 versus interferon-alfa as first-line systemic therapy for patients with metastatic renal cell carcinoma]. clinicaltrials.gov/ct2/show/NCT00083889 (first received 4 June 2004). CENTRAL [NCT00083889]
Patil S, Figlin RA, Hutson TE, Michaelson D, Negrier S, Kim ST, et al. TWiST analysis to estimate overall benefit for metastatic renal cell carcinoma (mRCC) patients (pts) treated in a phase III trial of sunitinib versus interferon-alfa (IFN-α). Journal of Clinical Oncology 2010;28(15 suppl):4594. CENTRAL [DOI: 10.1200/jco.2010.28.15_suppl.4594]
Wilkerson J, Stein WD, Kim ST, Huang X, Motzer RJ, Fojo AT, et al. Validation of kinetic analysis of renal cancer regression and growth following treatment with sunitinib and interferon-alfa: analysis of the pivotal randomized trial. Journal of Clinical Oncology 2010;28(15 suppl):4597. CENTRAL [DOI: 10.1200/jco.2010.28.15_suppl.4597]

Motzer 2013a {published data only}

*
*. Motzer RJ, Hutson TE, Cella D, Reeves J, Hawkins R, Guo J, et al. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. New England Journal of Medicine 2013;369(8):722-1. CENTRAL [PMID: 23964934]
Motzer RJ, Hutson TE, McCann L, Deen K, Choueiri TK. Overall survival in renal-cell carcinoma with pazopanib versus sunitinib. New England Journal of Medicine 2014;370(18):1769-70. CENTRAL [PMID: 24785224]
NCT00720941. Pazopanib versus sunitinib in the treatment of locally advanced and/or metastatic renal cell carcinoma (COMPARZ) [Study VEG108844, A study of pazopanib versus sunitinib in the treatment of subjects with locally advanced and/or metastatic renal cell carcinoma]. clinicaltrials.gov/ct2/show/NCT00720941 (first received 23 July 2008). CENTRAL [NCT00720941]

Motzer 2013b {published data only}

Hutson T, Nosov D, Tomczak P, Bondarenko I, Lipatov ON, Sternberg CN, et al. Tivozanib vs sorafenib targeted therapy for advanced renal cell carcinoma: Final results of a phase III trial (901) and efficacy results of a 2nd line tivozanib extension study (902). Journal of Clinical Oncology 2015;33(15 suppl):4557. CENTRAL [DOI: 10.1200/jco.2015.33.15_suppl.4557]
*
*. Motzer RJ, Nosov D, Eisen T, Bondarenko I, Lesovoy V, Lipatov O, et al. Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: results from a phase III trial. Journal of Clinical Oncology 2013;31(30):3791-9. CENTRAL [PMID: 24019545]
NCT01030783. A study to compare tivozanib (AV-951) to sorafenib in subjects with advanced renal cell carcinoma (TIVO-1) [A phase 3, randomized, controlled, multi-center, open-label study to compare tivozanib (AV-951) to sorafenib in subjects with advanced renal cell carcinoma (TIVO-1)]. clinicaltrials.gov/ct2/show/NCT01030783 (first received 11 December 2009). CENTRAL [NCT01030783]

Motzer 2014 {published data only}

Knox JJ, Barrios CH, Kim TM, Cosgriff T, Srimuninnimit V, Pittman K, et al. Final overall survival analysis for the phase II RECORD-3 study of first-line everolimus followed by sunitinib versus first-line sunitinib followed by everolimus in metastatic RCC. Annals of Oncology 2017;28(6):1339-45. CENTRAL [PMID: 28327953]
Knox JJ, Barrios CH, Kim TM, Cosgriff T, Srimuninnimit V, Pittman KB, et al. Final overall survival analysis for the RECORD-3 study of first-line everolimus followed by sunitinib versus first-line sunitinib followed by everolimus in metastatic RCC (mRCC). Journal of Clinical Oncology 2015;33(15 suppl):4554. CENTRAL [DOI: 10.1200/jco.2015.33.15_suppl.4554]
*
*. Motzer RJ, Barrios CH, Kim TM, Falcon S, Cosgriff T, Harker WG, et al. Phase II randomized trial comparing sequential first-line everolimus and second-line sunitinib versus first-line sunitinib and second-line everolimus in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology 2014;32(25):2765-72. CENTRAL [PMID: 25049330]
NCT00903175. Efficacy and safety comparison of RAD001 versus sunitinib in the first-line and second-line treatment of patients with metastatic renal cell carcinoma (RECORD-3) [An open-label, multicenter phase II study to compare the efficacy and safety of RAD001 as first-line followed by second-line sunitinib versus sunitinib as First-line followed by second-line RAD001 in the treatment of patients with metastatic renal cell carcinoma.]. clinicaltrials.gov/ct2/show/NCT00903175 (first received 18 May 2009). CENTRAL [NCT00903175]

Motzer 2019 {published data only}

*
*. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, et al. Avelumab plus axitinib versus sunitinib for advanced renal cell carcinoma. New England Journal of Medicine 2019;380(12):1103-5. CENTRAL [PMID: 30779531]
NCT02684006. A study of avelumab with axitinib versus sunitinib in advanced renal cell cancer (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]. clinicaltrials.gov/ct2/show/NCT02684006 (first received 17 February 2016). CENTRAL [NCT02684006]

Ravaud 2015 {published data only}

NCT00719264. Safety and efficacy of bevacizumab plus RAD001 versus interferon alfa-2a and bevacizumab for the first-line treatment in adult patients with kidney cancer [A randomized, open-label, multi-center phase II study to compare bevacizumab plus RAD001 versus interferon alfa-2a plus bevacizumab for the first-line treatment of patients with metastatic clear cell carcinoma of the kidney]. clinicaltrials.gov/ct2/show/study/NCT00719264 (first received 21 July 2008). CENTRAL [NCT00719264]
*
*. Ravaud A, Barrios CH, 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. CENTRAL [PMID: 25851632]

Retz 2019 {published data only}

NCT01613846. Phase III sequential open-label study to evaluate the efficacy and safety of sorafenib followed by pazopanib versus pazopanib followed by sorafenib in the treatment of advanced / metastatic renal cell carcinoma (SWITCH-II) [Phase III randomized sequential open-label study to evaluate the efficacy and safety of sorafenib followed by pazopanib versus pazopanib followed by sorafenib in the treatment of advanced / metastatic renal cell carcinoma]. clinicaltrials.gov/ct2/show/study/NCT01613846 (first received 7 June 2012). CENTRAL [NCT01613846]
*
*. Retz M, Bedke J, Bögemann M, Grimm MO, Zimmermann U, Müller L, et al. SWITCH II: Phase III randomized, sequential, open-label study to evaluate the efficacy and safety of sorafenib-pazopanib versus pazopanib-sorafenib in the treatment of advanced or metastatic renal cell carcinoma (AUO AN 33/11). European Journal of Cancer 2019;107:37-45. CENTRAL [PMID: 30529901]
Retz M, BedkeJ, Herrmann E, Grimm M, Zimmermann U, Müller L, et al. Phase III randomized, sequential, open-label study to evaluate the efficacy and safety of sorafenib-pazopanib versus pazopanib sorafenib in the treatment of metastatic renal cell carcinoma(SWITCH-II). Annals of Oncology 2017;28(suppl 5):295. CENTRAL [DOI: 10.1093/annonc/mdx371]

Rini 2008 {published data only}

Halabi S, Rini BI, Stadler WM, Small EJ. Use of progression-free survival (PFS) to predict overall survival (OS) in patients with metastatic renal cell carcinoma (mRCC). Journal of Clinical Oncology 2010;28(15 suppl):4525. CENTRAL [DOI: 10.1200/jco.2010.28.15_suppl.4525]
Harzstark AL, Halabi S, Stadler WM, et al. Hypertensionis associated with clinical outcome for patients with metastatic renal cell carcinoma treated with interferon and bevacizumab on CALGB 90206. In: Genitourinary Cancers Symposium, ASCO, abstr 351. 2010. CENTRAL
NCT00072046. Interferon alfa-2b with or without bevacizumab in treating patients with advanced renal cell carcinoma (Kidney Cancer) [A randomized phase III trial of interferon alfa-2B or interferon alfa-2B plus bevacizumab in patients with advanced renal carcinoma]. clinicaltrials.gov/ct2/show/study/NCT00072046 (first received 6 November 2003). CENTRAL [NCT00072046]
Rini BI, Halabi S, Rosenberg J, Stadler WM, Vaena DA, Atkins JN, et al. Bevacizumab plus interferon-alfa versus interferon-alfa monotherapy in patients with metastatic renal cell carcinoma: results of overall survival for CALGB 90206. Journal of Clinical Oncology 2009;27(18 suppl):LBA5019. CENTRAL [DOI: 10.1200/jco.2009.27.18_suppl.lba5019 ]
*
*. Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Ou SS, et al. Bevacizumab plus interferon alfa compared with interferon alfa monotherapy in patients with metastatic renal cell carcinoma: CALGB90206. Journal of Clinical Oncology 2008;26(33):5422-8. CENTRAL [PMID: 18936475]
Rini BI, Halabi S, Rosenberg JE, et al. CALGB 90206:a phase III trial of bevacizumab plus interferon-alfa versus interferon-alfa monotherapy in metastatic renal cell carcinoma. In: Genitourinary Cancers Symposium, ASCO, abstr 350. 2008. CENTRAL
Rini BI, Halabi S, Taylor J, Small EJ, Schilsky RL, Cancer and Leukemia Group B . Cancerand Leukemia Group B 90206: a randomized phase III trial of interferon-a or interferon-a plus anti-vascular endothelial growth factor antibody (bevacizumab) in metastatic renal cell carcinoma. Clinical Cancer Research 2004;10(8):2584-6. CENTRAL [PMID: 15102658]

Rini 2014 {published data only}

NCT00631371. Study Comparing Bevacizumab + Temsirolimus vs. Bevacizumab + Interferon-Alfa In Advanced Renal Cell Carcinoma Subjects (INTORACT). linicaltrials.gov/ct2/show/NCT00631371 (first received 20 May 2019). CENTRAL [NCT00631371]
*
*. 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 [PMID: 24297945]

Rini 2016 {published data only}

NCT01265901. IMA901 in patients receiving sunitinib for advanced/metastatic renal cell carcinoma [A randomized, controlled phase III study investigating IMA901 multipeptide cancer vaccine in patients receiving sunitinib as first-line therapy for advanced/metastatic renal cell carcinoma]. clinicaltrials.gov/ct2/show/study/NCT01265901 (first received 23 December 2010). CENTRAL [NCT01265901]
*
*. Rini BI, Stenzl A, Zdrojowy R, Kogan M, Shkolnik M, Oudard S, et al. IMA901, a multipeptide cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy for advanced or metastatic renal cell carcinoma (IMPRINT): a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncology 2016;17(11):1599-611. CENTRAL [PMID: 27720136]

Rini 2019a {published data only}

NCT02853331. Study to evaluate the efficacy and safety of pembrolizumab (MK-3475) in combination with axitinib versus sunitinib monotherapy in participants with renal cell carcinoma (MK-3475-426/KEYNOTE-426) [A phase III randomized, open-label study to evaluate efficacy and safety of pembrolizumab (MK-3475) in combination with axitinib versus sunitinib monotherapy as a first-line treatment for locally advanced or metastatic renal cell carcinoma (mRCC) (KEYNOTE-426)]. clinicaltrials.gov/ct2/show/NCT02853331 (first received 2 August 2016). CENTRAL [NCT02853331]
Powles T, Plimack ER, Stus V, Gafanov RA, Hawkins RE, Nosov D. Pembrolizumab (pembro) plus axitinib (axi) versus sunitinib as first-line therapy for locally advanced or metastatic renal cell carcinoma (mRCC): phase III KEYNOTE-426 study. Journal of Clinical Oncology 2019;37(7 suppl):543-543. CENTRAL
*
*. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. New England Journal of Medicine 2019;380(12):1116-27. CENTRAL [PMID: 30779529]

Rini 2019b {published data only}

Atkins MB, Rini BI, Motzer RJ, Powles T, McDermott DF, Suarez C, et al. Patient-reported outcomes from the phase 3 randomized IMmotion151 trial: atezolizumab + bevacizumab vs sunitinib in treatment-naive metastatic renal cell carcinoma. Clinical Cancer Research 2020;26(11):2506-2514. CENTRAL
Escudier B, Motzer RJ, Rini BI, Powles T, McDermott DF, Suarez C, et al. Patient-reported outcomes (PROs) in IMmotion151: atezolizumab (atezo) + bevacizumab (bev) vs sunitinib (sun) in treatment (tx) naive metastatic renal cell carcinoma (mRCC). Journal of Clinical Oncology 2018;36(15_suppl):4511. CENTRAL [DOI: 10.1200/JCO.2018.36.15_suppl.4511]
Motzer RJ, Powles T, Atkins MB, Escudier B, McDermott DF, Suarez C, et al. IMmotion151: a randomized phase III study of atezolizumab plus bevacizumab vs sunitinib in untreated metastatic renal cell carcinoma (mRCC). Journal of Clinical Oncology 2018;36(6 suppl):578. CENTRAL [DOI: 10.1200/JCO.2018.36.6_suppl.578]
NCT02420821. A study of atezolizumab in combination with bevacizumab versus sunitinib in participants with untreated advanced renal cell carcinoma (RCC) (IMmotion151) [A phase III, open-label, randomized study of atezolizumab (Anti-PD-L1 Antibody) in combination with bevacizumab versus sunitinib in patients with untreated advanced renal cell carcinoma]. clinicaltrials.gov/ct2/show/NCT02420821 (first received 20 April 2015). CENTRAL [NCT02420821]
*
*. Rini BI, Powles T, Atkins MB, Escudier B, McDermott DF, Suarez C, et al. Atezolizumab plus bevacizumab versus sunitinib in patients with previously untreated metastatic renal cell carcinoma (IMmotion151): a multicentre, open-label, phase 3, randomised controlled trial. Lancet 2019;393(10189):2404-15. CENTRAL [PMID: 31079938]

Sternberg 2010 {published data only}

Cella D, Pickard AS, Duh MS, Guerin A, Mishagina N, Antràs L, et al. Health-related quality of life in patients with advanced renal cell carcinoma receiving pazopanib or placebo in a randomised phase III trial. European Journal of Cancer 2012;48(3):311-23. CENTRAL [PMID: 21689927]
Hutson TE, Bukowski RM. A phase II study of GW786034 using a randomized discontinuation design in patients with locally recurrent or metastatic clear-cell renal cell carcinoma. Clinical Genitourinary Cancer 2006;4(4):296-8. CENTRAL [PMID: 16729915]
Hutson TE, Davis ID, Machiels JP, de Souza PL, Hong BF, Rottey S, et al. Pazopanib (GW786034) is active in metastatic renal cell carcinoma (RCC): Interim results of a phase II randomized discontinuation trial (RDT). Journal of Clinical Oncology 2007;25(18 suppl):5031. CENTRAL [DOI: 10.1200/jco.2007.25.18_suppl.5031]
NCT00334282. Safety and efficacy of GW786034 (pazopanib) In metastatic renal cell carcinoma [A randomised, double-blind, placebo controlled, multi-center phase III study to evaluate the efficacy and safety of pazopanib (GW786034) compared to placebo in patients with locally advanced and/or metastatic renal cell carcinoma]. clinicaltrials.gov/ct2/show/study/NCT00334282 (first received 7 June 2006). CENTRAL [NCT00334282]
*
*. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. Journal of Clinical Oncology 2010;28(6):1061-8. CENTRAL [PMID: 20100962]
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 [PMID: 23321547]
Sternberg CN, Szczylik C, Lee E, Salman PV, Mardiak J, Davis ID, et al. A randomized, double-blind phase III study of pazopanib in treatment-naive and cytokine-pretreated patients with advanced renal cell carcinoma (RCC). Journal of Clinical Oncology 2009;27(15S):5021. CENTRAL [DOI: 10.1200/jco.2009.27.15s.5021]

Referencias de los estudios excluidos de esta revisión

Armstrong 2016 {published data only}

Armstrong AJ, Halabi S, Eisen T, Broderick S, Stadler WM, Jones RJ, et al. Everolimus versus sunitinib for patients with metastatic non-clear cell renal cell carcinoma (ASPEN): a multicentre, open-label, randomised phase 2 trial. Lancet Oncology 2016;17(3):378-88. CENTRAL [PMID: 26794930]

Atkins 2004 {published data only}

Atkins MB, Hidalgo M, Stadler W, Logan TF, Dutcher JP, Hudes GR, et al. Randomized phase II study of multiple dose levels of CCI-779, a novel mammalian target of rapamycin inhibitor, in patients with advanced refractory renal cell carcinoma. Journal of Clinical Oncology 2004;22:909-18. CENTRAL [PMID: 14990647]

Bracarda 2010 {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 Urology 2013;63(2):254-61. CENTRAL [PMID: 22964169]

Broom 2015 {published data only}

Broom RJ, Hinder V, Sharples K, Proctor J, Duffey S, Pollard S, et al. Everolimus and zoledronic acid in patients with renal cell carcinoma with bone metastases: A randomized first-line phase II trial. Clinical Genitourinary Cancer 2015;13(1):50-8. CENTRAL [PMID: 25163397]

Bukowski 2007 {published data only}

Bukowski RM, Kabbinavar F, Figlin RA, Flaherty K, Srinivas S, Vaishampayan UN, et al. Randomized phase II study of erlotinib combined with bevacizumab compared with bevacizumab alone in metastatic renal cell cancer. Journal of Clinical Oncology 2007;25(29):4536–41. CENTRAL [PMID: 17876014]

Choueiri 2015 {published data only}

Choueiri TK, Escudier B, Powles T, Mainwaring PN, Rini BI, Donskov F, et al. Cabozantinib versus everolimus in advanced renal-cell carcinoma. New England Journal of Medicine 2015;373(19):1814–23. CENTRAL [PMID: 26406150]

Choueiri 2017 {published data only}

Choueiri TK, Halabi S, Sanford BL, Hahn O, Michaelson MD, Walsh MK, et al. Cabozantinib versus sunitinib as initial targeted therapy for patients with metastatic renal cell carcinoma of poor or intermediate risk: The Alliance A031203 CABOSUN Trial. Journal of Clinical Oncology 2017;35(6):591-7. CENTRAL [PMID: 28199818]

Cirkel 2016 {published data only}

Cirkel GA, Hamberg P, Sleijfer S, Loosveld OJL, Dercksen MW, Los M, et al. Alternating treatment with pazopanib and everolimus vs continuous pazopanib to delay disease progressionin patients with metastatic clear cell renal cell cancer: The ROPETAR randomized clinical trial. JAMA Oncology 2017;3(4):501-8. CENTRAL [PMID: 27918762]

Dorff 2015 {published data only}

Dorff TB, Longmate JA, Pal SK, Stadler WM, Fishman MN, Vaishampayan UN, et al. Bevacizumab alone or in combination with TRC105 for patients with refractory metastatic renal cell cancer. Cancer 2017;123(23):4566-73. CENTRAL [PMID: 28832978]

Ebbinghaus 2007 {published data only}

Ebbinghaus S, Hussain M, Tannir N, Gordon M, Desai AA, Knight RA, et al. Phase 2 study of ABT-510 in patients with previously untreated advanced renal cell carcinoma. Clinical Cancer Research 2007;13(22 Pt 1):6689–95. CENTRAL [PMID: 18006769]

Eisen 2015 {published data only}

Eisen T, Loembé A-B, Shparyk Y, MacLeod N, Jones RJ, Mazurkiewicz M, Temple G, et al. A randomised, phase II study of nintedanib or sunitinib in previously untreated patients with advanced renal cell cancer: 3-year results. British Journal of Cancer 2015;113(8):1140-7. CENTRAL [PMID: 26448178]

Escudier 2007 {published data only}

Escudier B, Choueiri TK, Oudard S, Szczylik C, Négrier S, Ravaud A, et al. Prognostic factors of metastatic renal cell carcinoma after failure of immunotherapy: new paradigm from a large phase III trial with shark cartilage extract AE 941. Journal of Urology 2007;178:1901-5. CENTRAL [PMID: 17868728]

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;27:1280–9. CENTRAL [PMID: 19171708]

Escudier 2010a {published data only}

Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. New England Journal of Medicine 2007;356(2):125-34. CENTRAL [PMID: 17215530]

Flaherty 2015 {published data only}

Flaherty KT, Manola JB, Pins M, McDermott DF, Atkins MB, Dutcher JJ, et al. BEST: A randomized phase II study of vascular endothelial growth factor, RAF Kinase, and mammalian target of rapamycin combination targeted therapy with bevacizumab, sorafenib, and temsirolimus in advanced renal cell carcinoma--A trial of the ECOG-ACRIN cancer research group (E2804). Journal of Clinical Oncology 2015;33(21):2384-91. CENTRAL [PMID: 26077237]

Gordon 2004 {published data only}

Gordon MS, Manola J, Fairclough D, Cella D, Richardson R, Sosman J, et al. Low dose interferon-α2b + thalidomide in patients with previously untreated renal cell cancer. Improvement in progression-free survival but not quality of life or overall survival. A phase III study of the Eastern Cooperative Oncology Group(E2898). Journal of Clinical Oncology 2004;22(14 suppl):4516. CENTRAL [DOI: 10.1200/jco.2004.22.90140.4516]

Hainsworth 2015 {published data only}

Hainsworth JD, Reeves JA, Mace JR, Crane EJ, Hamid O, Stille JR, et al. A randomized, open-Label phase 2 study of the CXCR4 Inhibitor LY2510924 in combination with sunitinib versus sunitinib alone in patients with metastatic renal cell carcinoma (RCC). Targeted Oncology 2016;11(5):643-53. CENTRAL [PMID: 27154357]

Hawkins 2016 {published data only}

Hawkins R, Gore M, Shparyk Y, Bondar V, Gladkov O, Ganev T, et al. A randomized phase II/III study of naptumomab estafenatox þ IFNa versus IFNa in renal cell carcinoma: final analysis with baseline biomarker subgroup and trend analysis. Clinical Cancer Research 2016;22(13):3172-81. CENTRAL [PMID: 26851187]

Hutson 2013 {published data only}

Hutson TE, Lesovoy V, Al-Shukri S, Stus VP, Lipatov ON, Bair AH, et al. Axitinib versus sorafenib as first-line therapy in patients with metastatic renal-cell carcinoma: a randomised open-label phase 3 trial. Lancet Oncology 2013;14(13):1287-94. CENTRAL [PMID: 24206640]

Hutson 2014 {published data only}

Hutson TE, Escudier B, Esteban E, Bjarnason GA, Lim HY, Pittman KB, et al. Randomized phase III trial of temsirolimus versus sorafenib as second-line therapy after sunitinib in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology 2014;32(8):760-7. CENTRAL [PMID: 24297950]

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. Cancer 2010;116(1):57-65. CENTRAL [PMID: 19862815]

Jonasch 2017 {published data only}

Jonasch E, Hasanov E, Corn PG, Moss T, Shaw KR, Stovall S, et al. A randomized phase 2 study of MK-2206 versus everolimus in refractory renal cell carcinoma. Annals of Oncology 2017;28(4):804-8. CENTRAL [PMID: 28049139]

Lee 2006 {published data only}

Lee CP, Patel PM, Selby PJ, Hancock BW, Mak I, Pyle L, et al. Randomized phase II study comparing thalidomide with medroxyprogesterone acetate in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology 2006;24(6):898-903. CENTRAL [PMID: 16484699]

Lee 2015 {published data only}

Lee JL, Kim MK, Park I, Ahn JH, Lee DH, Ryoo HM, et al. Randomized phase II trial of sunitinib four weeks on and two weeks off versus Two weeks on and One week off in metastatic clear-cell type Renal cell carcinoma: RESTORE trial. Annals of Oncology 2015;26(11):2300-5. CENTRAL [PMID: 26347107]

Madhusudan 2004 {published data only}

Madhusudan S, Protheroe A, Vasey P, Patel P, Selby P, Altman D, et al. A randomized phase II study of interferon alpha alone or in combination with thalidomide in metastatic renal cancer. Journal of Clinical Oncology 2004;22(14_suppl):4742. CENTRAL [DOI: 10.1200/jco.2004.22.90140.4742]

Motzer 2008 {published data only}

Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomized, placebo-controlled phase III trial. Lancet 2008;372(9637):449-56. CENTRAL [PMID: 18653228]

Motzer 2012 {published data only}

Motzer RJ, Hutson TE, Olsen MR, Hudes GR, Burke JM, Edenfield WJ, et al. Randomized phase II trial of sunitinib on an intermittent versus continuous dosing schedule as first-line therapy for advanced renal cell carcinoma. Journal of Clinical Oncology 2012;30(12):1371-7. CENTRAL [PMID: 22430274 ]

Motzer 2014b {published data only}

Motzer RJ, Porta C, Vogelzang NJ, Sternberg CN, Szczylik C, Zolnierek J, et al. Dovitinib versus sorafenib for third-line targeted treatment of patients with metastatic renal cell carcinoma:an open-label, randomised phase 3 trial. Lancet Oncology 2014;15(3):286-96. CENTRAL [PMID: 24556040]

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. CENTRAL [PMID: 26406148]

Motzer 2015b {published data only}

Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel T, Harrison MR, et al. Nivolumab for metastatic renal cell carcinoma (mRCC): Results of a randomized, dose-ranging phase II trial. Journal of Clinical Oncology 2014;32(15_suppl):5009. CENTRAL [DOI: 10.1200/jco.2014.32.15_suppl.5009]

Motzer 2015c {published data only}

Motzer RJ, Hutson TE, Glen H, Michaelson MD, Molina A, Eisen T, et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial. Lancet Oncology 2015;16(15):1473-82. CENTRAL [PMID: 26482279]

Mulders 2012 {published data only}

Mulders P, Hawkins R, Nathan P, de Jong I, Osanto S, Porfiri E, et al. Cediranib monotherapy in patients with advanced renal cell carcinoma: results of a randomised phase II study. European Journal of Cancer 2012;48(4):527-37. CENTRAL [PMID: 22285180]

Négrier 2011 {published data only}

Négrier S, Gravis G, Pérol 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 [PMID: 21664867]

Nosov 2012 {published data only}

Nosov DA, Esteves B, Lipatov ON, Lyulko AA, Anischenko AA, Chacko RT, et al. Antitumor activity and safety of tivozanib (AV-951) in a phase II randomized discontinuation trial in patients with renal cell carcinoma. Journal of Clinical Oncology 2012;30(14):1678-85. CENTRAL [PMID: 22493422]

Pal 2015 {published data only}

Pal S, Azad A, Bhatia S, Drabkin H, Costello B, Sarantopoulos J, et al. A Phase I/II trial of BNC105P with everolimus in metastatic renal cell carcinoma. Clinical Cancer Research 2015;21(15):3420-7. CENTRAL [PMID: 25788492]

Pili 2015 {published data only}

Pili R, Manola J, Carducci MA, Dutcher JP, Appleman LJ, Groteluschen DL, et al. Randomized phase II study of two different doses of AVE0005 (VEGF Trap, aflibercept) in patients (pts) with metastatic renal cell carcinoma (RCC): An ECOG-ACRIN study [E4805]. Journal of Clinical Oncology 2015;33(15_suppl):4549. CENTRAL [DOI: 10.1200/jco.2015.33.15_suppl.4549]

Powles 2014 {published data only}

Powles T, Oudard S, Escudier BJ, Brown JE, Hawkins RE, Castellano DE, et al. A randomized phase II study of GDC-0980 versus everolimus in metastatic renal cell carcinoma (mRCC) patients (pts) after VEGF-targeted therapy (VEGF-TT). Journal of Clinical Oncology 2014;32(15_suppl):4525. CENTRAL [DOI: 10.1200/jco.2014.32.15_suppl.4525]

Powles 2016a {published data only}

Powles T, Wheater M, Din O, Geldart T, Boleti E, Stockdale A, et al. A randomised phase 2 study of AZD2014 versus everolimus in patients with VEGF-refractory metastatic clear cell renal cancer. European Urology 2016;69(3):450-6. CENTRAL [PMID: 26364551]

Powles 2016b {published data only}

Powles T, Brown J, Larkin J, Jones R, Ralph C, Hawkins R, et al. A randomized, double-blind phase II study evaluating cediranib versus cediranib and saracatinib in patients with relapsed metastatic clear-cell renal cancer(COSAK). Annals of Oncology 2016;27(5):880–6. CENTRAL [PMID: 26802156]

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 [PMID: 21448165]

Ratain 2006 {published data only}

Ratain MJ, Eisen T, Stadler WM, Flaherty KT, Kaye SB, Rosner GL, et al. Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology 2006;24(16):2505-12. CENTRAL [PMID: 16636341]

Ravaud 2008 {published data only}

Ravaud A, Hawkins R, Gardner JP, von der Maase H, Zantl N, Harper P, et al. Lapatinib versus hormone therapy in patients with advanced renal cell carcinoma: a randomized phase III clinical trial. Journal of Clinical Oncology 2008;26(14):2285-91. CENTRAL [PMID: 18467719]

Rini 2011 {published data only}

Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 2011;378(9807):1931-9. CENTRAL [PMID: 22056247]

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 [PMID: 22692704]

Rini 2013 {published data only}

Rini BI, Melichar B, Ueda T, Grünwald V, Fishman MN, Arranz JA, et al. Axitinib with or without dose titration for first-line metastatic renal-cell carcinoma: a randomised double-blind phase 2 trial. Lancet Oncology 2013;14(12):1233-42. CENTRAL [PMID: 24140184]

Srinivas 2005 {published data only}

Srinivas S, Guarding AE. A lower dose of thalidomide is better than a high dose in metastatic renal cell carcinoma. BJU International 2005;96(4):536-9. CENTRAL [PMID: 16104906]

Stadler 2005 {published data only}

Stadler WM, Rosner G, Small E, Hollis D, Rini B, Zaentz DS, et al. Successful implementation of the randomized discontinuation trial design: an application to the study of the putative antiangiogenic agent carboxyaminoimidazole in renal cell carcinoma -CALGB 69901. Journal of Clinical Oncology 2005;23(16):3726-32. CENTRAL [PMID: 15923569]

Tannir 2016 {published data only}

Tannir NM, Jonasch E, Albiges L, Altinmakas E, Ng CS, Matin SF, et al. Everolimus versus sunitinib prospective evaluation in metastatic non-clear cell renal cell carcinoma (ESPN): A randomized multicenter phase 2 trial. European Urology 2016;69(5):866-74. CENTRAL [PMID: 26626617]

Tannir 2018 {published data only}

Tannir NM, Ross JA, Devine CE, Chandramohan A, Wang X, Lim ZD, et al. A randomized phase II trial of pazopanib (PAZ) versus temsirolimus (TEM) in patients (pts) with advanced clear-cell renal cell carcinoma (aCCRCC) of intermediate and poor-risk (the TemPa trial). Journal of Clinical Oncology 2018;36(6_suppl):583. CENTRAL [DOI: 10.1200/JCO.2018.36.6_suppl.583]

Tomita 2014 {published data only}

Tomita Y, Naito S, Sassa N, Takahashi A, Kondo T, Koie T, et al. Sunitinib versus sorafenib as first-line therapy for patients with metastatic renal cell carcinoma with favourable or intermediate MSKCC risk factors: A multicenter randomized trial, CROSS-J-RCC. Journal of Clinical Oncology 2014;32(4_suppl):502. CENTRAL [DOI: 10.1200/jco.2014.32.4_suppl.502]

Twardowski 2015 {published data only}

Twardowski P, Plets M, Plimack ER, Agarwal N, Tangen CM, Vogelzang NJ, et al. SWOG 1107: Parallel (randomized) phase II evaluation of tivantinib (ARQ-197) and tivantinib in combination with erlotinib in patients (Pts) with papillary renal cell carcinoma (pRCC). Journal of Clinical Oncology 2015;33(15_suppl):4523. CENTRAL [DOI: 10.1200/jco.2015.33.15_suppl.4523]

Yang 2003 {published data only}

Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, Topalian SL, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. New England Journal of Medicine 2003;349(5):427-34. CENTRAL [PMID: 12890841]

Choueiri 2018 {published and unpublished data}NCT03141177

*
*. Choueiri TK, Apolo AB, Powles T, Escudier B, Aren OR, Shah A, et al. A phase 3, randomized, open-label study of nivolumab combined with cabozantinib vs sunitinib in patients with previously untreated advanced or metastatic renal cell carcinoma (RCC; CheckMate 9ER). Journal of Clinical Oncology 2018;36(15_suppl):TPS4598. CENTRAL [DOI: 10.1200/JCO.2018.36.15_suppl.TPS4598]
NCT03141177. A study of nivolumab combined with cabozantinib compared to sunitinib in previously untreated advanced or metastatic renal cell carcinoma (CheckMate 9ER). clinicaltrials.gov/ct2/show/NCT03141177 (first received May 2019). CENTRAL

Choueiri 2019 {published and unpublished data}NCT03937219

*
*. Choueiri TK, Albiges L, Powles T, Scheffold C, Wang F, Motzer RJ, et al. A phase III study (COSMIC-313) of cabozantinib in combination with nivolumab and ipilimumab in patients with previously untreated advanced renal cell carcinoma of intermediate or poor-risk. Journal of Clinical Oncology 2020;38(6_suppl):TPS767. CENTRAL [DOI: 10.1200/JCO.2020.38.6_suppl.TPS767]
NCT03937219. Study of Cabozantinib in Combination With Nivolumab and Ipilimumab in Patients With Previously Untreated Advanced or Metastatic Renal Cell Carcinoma (COSMIC-313). https://clinicaltrials.gov/ct2/show/NCT03937219 (first received June 2020). CENTRAL

Grünwald 2018 {published and unpublished data}NCT02959554

Grunwald V, Bergmann L, Steiner T, Grullich C, Muller L, Staib P, et al. 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 (NIVOSWITCH). Oncology Research and Treatment 2018;41:75. CENTRAL
*
*. NCT02959554. Study in Which Therapy is Either Switched to Nivolumab After 3 Months of Treatment or Therapy is Continued With a Tyrosine Kinase Inhibitor in Patients With Metastatic Renal Cell Carcinoma (RCC) and Disease Control (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 (NIVOSWITCH)]. https://clinicaltrials.gov/ct2/show/NCT02959554. CENTRAL

Motzer 2018 {published and unpublished data}NCT02811861

*
*. Motzer RJ, Grünwald V, Hutson TE, Porta C, Powles T, Eto M, et al. A phase 3 trial to compare efficacy and safety of lenvatinib in combination with everolimus or pembrolizumab versus sunitinib alone in first-line treatment of patients with metastatic renal cell carcinoma. Journal of Clinical Oncology 2018;36(6 suppl):TPS706. CENTRAL [DOI: 10.1200/JCO.2018.36.6_suppl.TPS706]
NCT02811861. Lenvatinib/everolimus or Lenvatinib/pembrolizumab versus sunitinib alone as treatment of advanced renal cell carcinoma (CLEAR) [A multicenter, open-label, randomized, phase 3 trial to compare the efficacy and safety of lenvatinib in combination with everolimus or pembrolizumab versus sunitinib alone in first-line treatment of subjects with advanced renal cell carcinoma]. clinicaltrials.gov/ct2/show/study/NCT02811861 (first received 23 June 2016). CENTRAL

Adjei 2005

Adjei AA, Hidalgo M. Intracellular signal transduction pathway proteins as targets for cancer therapy. Journal of Clinical Oncology 2005;23(23):5386-403. [PMID: 15983388]

Bergsland 2006

Bergsland EK. When does the presence of the target predict response to the targeted agent? Journal of Clinical Oncology 2006;24(2):213-6. [PMID: 16365177]

Coppin 2004

Coppin C, Porzsolt F, Autenrieth M, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. Cochrane Database of Systematic Reviews 2004, Issue 3. Art. No: CD001425. [DOI: 10.1002/14651858.CD001425.pub2]

CTCAE

National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0. ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_5x7.pdf (accessed 18 May 2019).

Deeks 2017

Deeks JJ, Higgins JPT, Altman DG (editors) on behalf of the Cochrane Statistical Methods Group. Chapter 9: Analysing data and undertaking meta-analyses. In: Higgins JPT, Churchill R, Chandler J, Cumpston MS (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.2.0 (updated June 2017), The Cochrane Collaboration, 2017. Available from www.training.cochrane.org/handbook.

Deininger 2005

Deininger M, Buchdunger E, Druker BJ. The development of imatinib as a therapeutic agent for chronic myeloid leukeamia. Blood 2005;105(7):2640-53. [PMID: 15618470]

EAU Guidelines 2020

Ljungberg B, Albiges L, Bensalah K, Bex A, Giles RH, Hora M, et al. EAU Guidelines on Renal Cell Carcinoma. Edn. presented at the EAU Annual Congress Amsterdam. EAU Guidelines Office, 2020. 978-94-92671-07-3.

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. [PMID: 19097774]

ESMO Clinical Practice Guidelines 2019

Escudier B, Porta C, Schmidinger M, Rioux-Leclerq N, Bex A, Khoo V, et al. Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology 2019;30(5):706-20. [PMID: 30788497]

Fernández‐Pello 2017

Fernández-Pello S, Hofmann F, Tahbaz R, Marconi L, Lam TB, Albiges L, et al. A systematic review and meta-analysis comparing the effectiveness and adverse effects of different systemic treatments for non-clear cell renal cell carcinoma. European Urology 2017;71(3):426-43. [PMID: 27939075]

Gleave 1998

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. New England Journal of Medicine 1998;338(18):1265-71. [PMID: 9562580]

GLOBOCAN 2018

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians 2018;68(6):394-424. [PMID: 30207593]

GRADEpro GDT 2015 [Computer program]

GRADEpro GDT. Version accessed 10 March 2019. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015. Available at gradepro.org.

Guyatt 2008

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

Guyatt 2011

Guyatt G, Oxman AD, Akl EA, 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. [PMID: 21195583]

Hacker 2010

Hacker KE, Rathmell WK. Emerging molecular classification in renal cell carcinoma: implications for drug development. Targeted Oncology 2010;5(2):75-84. [PMID: 20645016]

Hart 2012

Hart B, Lundh A, Bero L. Effect of reporting bias on meta-analyses of drug trials: reanalysis of meta-analyses. BMJ 2012;344:d7202. [PMID: 22214754]

Higgins 2002

Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta-analysis. Statistics in Medicine 2002;21(11):1539-58. [PMID: 12111919]

Higgins 2003

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

Higgins 2009

Higgins JP, Thompson SG, Spiegelhalter DJ. A re-evaluation of random-effects meta-analysis. Journal of the Royal Statistical Society. Series A, (Statistics in Society) 2009;172(1):137-59. [PMID: 19381330]

Higgins 2017a

Higgins JPT, Churchill R, Chandler J, Cumpston MS (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.2.0 (updated June 2017), The Cochrane Collaboration, 2017. Available from www.training.cochrane.org/handbook.

Higgins 2017b

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Churchill R, Chandler J, Cumpston MS (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.2.0 (updated June 2017), The Cochrane Collaboration, 2017. Available from www.training.cochrane.org/handbook.

Howlader 2017

Howlader N, Noone AM, Krapcho M, Miller D, Bishop K, Kosary CL, et al (eds). SEER Cancer Statistics Review, 1975-2014, National Cancer Institute. Bethesda, MD. seer.cancer.gov/csr/1975_2014/, based on November 2016 SEER data submission, posted to the SEER web site, April 2017.

Johnson 2011

Johnson JR, Ning Y-M, Farrell A, Justice R, Keegan P, Pazdur R. Accelerated approval of oncology products: the Food and Drug Adminstration experience. Journal of the National Cancer Institute 2011;103(8):636-44. [PMID: 21422403]

Kaelin 2008

Kaelin WG Jr. Kidney cancer: now available in a new flavor. Cancer Cell 2008;14(6):423-4. [PMID: 19061830]

Kuusk 2017

Kuusk T, Albiges L, Escudier B, Grivas N, Haanen J, Powles T, et al. Antiangiogenic therapy combined with immune checkpoint blockade in renal cancer. Angiogenesis 2017;20(2):205-15. [PMID: 28401381]

Lalani 2018

Lalani AA, McGregor BA, Albiges L, Choueiri TK, Motzer R, Powles T, et al. Systemic treatment of metastatic clear cell renal cell carcinoma in 2018: Current paradigms, use of immunotherapy, and future directions. European Urology 2018;75(1):100-10. [PMID: 30327274]

Linehan 2005

Linehan WM, Grubb RL, Coleman JA, Zbar B, Walther MM. The genetic basis of cancer of kidney cancer: implications for gene-specific clinical management. BJU International 2005;95 Suppl 2:2-7. [PMID: 15720328]

Mickisch 2003

Mickisch GH. Rational selection of a control arm for randomised trials in metastatic renal cell carcinoma. European Urology 2003;43(6):670-9. [PMID: 12767369]

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 advanced renal cell carcinoma. Journal of Clinical Oncology 2002;20(1):289-96. [PMID: 11773181]

Oliver 1989

Oliver RTD, Nethersell ABW, Bottomly JM. Unexplained spontaneous regression and alpha-interferon as treatment for metastatic renal carcinoma. British Journal of Urology 1989;63(2):128-31. [PMID: 2702395]

Patil 2010

Patil S, Ishill N, Deluca J, Motzer RJ. Stage migration and increasing proportion of favorable-prognosis metastatic renal cell carcinoma patients. Cancer 2010;116(2):347-54. [PMID: 19921736]

Review Manager 2020 [Computer program]

Review Manager 5 (RevMan 5). Version 5.4. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2020.

Riley 2011

Riley RD, Higgins JP, Deeks JJ. Interpretation of random effects meta-analyses. BMJ 2011;342:d549. [PMID: 21310794]

Rini 2005

Rini BI, Small EJ. Biology and clinical development of vascular endothelial growth factor-targeted therapy in renal cell carcinoma. Journal of Clinical Oncology 2005;23(5):1028-43. [PMID: 15534359]

Roberts 2003

Roberts TG Jr, Lynch TJ Jr, Chabner BA. The phase III trial in the era of targeted therapy: unravelling the "go or no go" decision. Journal of Clinical Oncology 2003;21(19):3683-95. [PMID: 14512401]

Sawyers 2004

Sawyers C. Targeted cancer therapy. Nature 2004;432(7015):294-7. [PMID: 15549090]

Schroll 2015

Schroll J, Bero L. Regulatory agencies hold the key to improving Cochrane Reviews of drugs. Cochrane Database of Systematic Reviews 2015, Issue 4. [DOI: 10.1002/14651858.ED000098]

Schünemann 2017

Schünemann HJ, Oxman AD, Higgins JPT, Vist GE, Glasziou P, Akl E, et al, on behalf of the Cochrane GRADEing Methods Group and the Cochrane Statistical Methods Group. Chapter 11: Completing ‘Summary of findings’ tables and grading the confidence in or quality of the evidence. In: Higgins JPT, Churchill R, Chandler J, Cumpston MS (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.2.0 (updated June 2017). The Cochrane Collaboration, 2017. Available from www.training.cochrane.org/handbook.

Srigley 2013

Srigley JR, Delahunt B, Eble JN, Egevad L, Epstein JI, Grignon D, et al. The International Society of Urological Pathology (ISUP) Vancouver Classification of Renal Neoplasia. American Journal of Surgical Pathology 2013;37(10):1469-89. [PMID: 24025519]

Stadler 2006

Stadler WM. New targets, therapies, and toxicities: lessons to be learned. Journal of Clinical Oncology 2006;24(1):4-5. [PMID: 16314613]

Sterne 2011

Sterne JA, Sutton AJ, Ioannidis JP, Terrin N, Jones DR, Lau J, et al. Recommendations for examining and interpreting funnel plot asymmetry in meta-analyses of randomised controlled trials. BMJ 2011;343:d4002. [PMID: 21784880]

Strebhardt 2008

Strebhardt K, Ullrich A. Paul Ehrlich's magic bullet concept: 100 years of progress. Nature Reviews Cancer 2008;8(6):473-80. [PMID: 18469827]

Unverzagt 2017

Unverzagt S, Moldenhauer I, Nothacker M, Roßmeißl D, Hadjinicolaou AV, Peinemann F, et al. Immunotherapy for metastatic renal cell carcinoma. Cochrane Database of Systematic Reviews 2017, Issue 5. Art. No: CD011673. [DOI: 10.1002/14651858.CD011673.pub2]

Uzzo 2003

Uzzo RG, Cairns P, Al-Saleem T, Hudes G, Haas N, Greenberg RE, et al. The basic biology and immunobiology of renal cell carcinoma: considerations for the clinician. Urologic Clinics of North America 2003;30(3):423-36. [PMID: 12953746]

Wallis 2018

Wallis CJD, Klaassen Z, Bhindi B, Ye XY, Chandrasekar T, Farrell AM, et al. First-line systemic therapy for metastatic renal cell carcinoma: A systematic review and network meta-analysis. European Urology 2018;74(3):309-321. [PMID: 29656851]

Wood 2008

Wood L, Egger M, Gluud LL, Schulz KF, Jüni P, Altman DG, et al. Empirical evidence of bias in treatment effect estimates in controlled trials with different interventions and outcomes: meta-epidemiological study. BMJ 2008;336(7644):601-5. [PMID: 18316340]

Young 2009

Young AC, Craven RA, Cohen D, Taylor C, Booth C, Harnden P, et al. Analysis of VHL gene alterations and their relationship to clinical parameters in sporadic conventional renal cell carcinoma. Clinical Cancer Research 2009;15(24):7582-92. [PMID: 19996202]

Referencias de otras versiones publicadas de esta revisión

Coppin 2006

Coppin C, Porzolt F, Le L, Autenrieth M, Wilt T. Targeted therapy for advanced renal cell carcinoma. Cochrane Database of Systematic Reviews 2006, Issue 1. Art. No: CD006017. [DOI: 10.1002/14651858.CD006017]

Coppin 2008

Coppin C, Le L, Wilt TJ, Kollmannsberger C. Targeted therapy for advanced renal cell carcinoma. Cochrane Database of Systematic Reviews 2008, Issue 2. Art. No: CD006017. [DOI: 10.1002/14651858.CD006017.pub2]

Coppin 2011

Coppin C, Kollmannsberger C, Le L, Porzsolt F, Wilt TJ. Targeted therapy for advanced renal cell cancer (RCC): a Cochrane systematic review of published randomised trials. BJU International 2011;108(10):1556-63. [PMID: 21952069]

Hofmann 2017

Hofmann F, Marconi L, Stewart F, Lam T, Bex A, Canfield SE, et al. Targeted therapy for metastatic renal cell carcinoma. Cochrane Database of Systematic Reviews 2017, Issue 9. Art. No: CD012796. [DOI: 10.1002/14651858.CD012796]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Eichelberg 2015

Study characteristics

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: February 2009 to December 2011

Blinding: open label study

Strata: MSKCC risk category

IMC: data not found

Crossover: from first to second line after disease progression

Participants

Histology: all histologies

Prior systemic therapy: treatment‐naïve

Measurable disease: required

Non‐metastatic %: combined data not found

M/F: 274/91

Eligible PS: ECOG PS 1 or better

Age median (range): 65 (39 to 84)

Prior nephrectomy: 335

Prognostic strata: system, good/intermediate/poor risk: MSKCC; 153/202/2

Interventions

sorafenib 400 mg po twice daily followed, on progression or toxicity, by sunitinib 50 mg po daily 4 wks on, 2 wks off or vice versa

Outcomes

PFS: primary outcome (time from randomisation to confirmed progression or death during second‐line therapy); secondary outcome (time from randomisation to confirmed progression or death during first‐line therapy)

OS: secondary endpoint

AE: reported in toxicity table

QoL: not analysed

RR: secondary outcome

Other: disease control rate, total time to progression, time to first‐line treatment failure, cardiotoxicity

Funding Sources

German Cancer Society (DKG), Austrian Social Security Institutions, grants from industry study sponsor

Declarations of interest

Reported

Notes

Planned as a non‐inferiority study, amended to a superiority design; power reduced from 90% to 85% due to slower rate of events than expected

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Central randomisation via fax"

Allocation concealment (selection bias)

Low risk

"the person who generated the randomisation list was not involved in the study project management, monitoring, or data management."

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

Open label study design, participants and personnel not blinded to treatment; no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Open label study design, participants and personnel not blinded to treatment; both arms treated with same drugs in different sequence

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Assessed by investigator; no effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

PFS assessed by investigator

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

All patients reported, 5/7 did not receive treatment in first line

Incomplete outcome data (attrition bias)
Response rate

Low risk

All patients reported, 5/7 did not receive treatment in first line

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated patients in first and second line reported

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

Unclear risk

Study design changed from non‐inferiority design to superiority design after randomisation of 138 participants

Escudier 2010

Study characteristics

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: June 2006 to October 2005

Blinding: double‐blind study

Strata: country, risk group

IMC: data safety monitoring board used

Crossover: not planned but at preplanned interim OS analysis, difference in PFS clinically and statistically significant and DSMB recommended that patients in the control group who had not experienced progression should cross over to receive bevacizumab

Participants

Histology: clear cell

Prior systemic therapy: treatment‐naïve

Measurable disease: required

Non metastatic %: data not found

M/F: 457/192

Eligible PS: Karnofsky > 60

Age median (range): 61 (18 to 82)

Prior nephrectomy: required

Prognostic strata: system, good/intermediate/poor risk %: MSKCC 30/61/9

Interventions

Interferon‐a2a 9 MU sc tiw (subcutaneous thrice weekly) plus either (1) BEVACIZUMAB 10 mg/kg IV q2w, or (2) placebo [crossed over at final PFS analysis if not progressed]

Outcomes

PFS: reported [protocol modified to permit final PFS analysis before mature OS data]

OS: primary endpoint

AE: reported in toxicity table

QoL: not assessed

RR: secondary outcome

Other:

Funding Sources

Industry sponsored

Declarations of interest

Reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

“randomization was done centrally”

Allocation concealment (selection bias)

Low risk

Clearly described; interactive voice recognition system

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"Double blind placebo controlled trial". Participants and personnel were blinded to treatment

Blinding of participants and personnel (performance bias)
Subjective outcomes

Low risk

"Double blind placebo controlled trial". Participants and personnel were blinded to treatment

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Investigator assessed, no effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

PFS assessed by investigator who were blinded to treatment

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

4% in each arm lost or withdrew consent; intention to treat population analysed

Incomplete outcome data (attrition bias)
Response rate

Low risk

4% in each arm lost or withdrew consent; intention to treat population analysed

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants analysed

Selective reporting (reporting bias)

Low risk

All protocol endpoints reported

Other bias

Unclear risk

Early unblinding and addition of bevacizumab; recommended for unprogressed placebo‐assigned patients by independent monitoring committee based on unplanned final PFS and preplanned interim survival analysis

Escudier 2017

Study characteristics

Methods

Study design: multicentre RCT

Phase: 3

Accrual period: October 2014 to February 2016

Blinding: open label study design

Strata: IMDC score, region

IMC (independent monitoring committee): a data and safety monitoring committee reviewed efficacy and safety

Crossover: not planned

Participants

Histology: clear cell

Prior systemic therapy: treatment‐naïve

Measurable disease: required

Non metastatic %: data not found; at least 78% of participants had 2 or more target or non‐target lesions

M/F: 808/288

Eligible PS: Karnofsky PS 70% or better

Age median: 62 (21 to 85)

Prior nephrectomy: 890

Prognostic strata: system, % good/intermediate/poor/NA risk: IMDC risk score (0 vs. 1 or 2 vs. 3 to 6) and geographic region

Interventions

nivolumab 3 mg/kg + ipilimumab 1 mg/kg every 3 wk for 4 doses followed by nivolumab 3 mg/kg every 2 wk vs sunitinib 50 mg daily orally for 4 wk (6‐wk cycles)

Outcomes

PFS: primary endpoint

OS: primary endpoint

AE: toxicity table reported

QoL: reported

RR: primary endpoint

Other:

Funding Sources

Industry sponsored

Declarations of interest

Published online

Notes

Stopped early

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Begins with the randomization call to the Interactive Voice Response System (IVRS)" (from protocol)

Allocation concealment (selection bias)

Low risk

"Begins with the randomization call to the Interactive Voice Response System (IVRS)" (from protocol)

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"Open‐label, phase 3 trial" participants and personnel were not blinded to treatment; no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"Open‐label, phase 3 trial" participants and personnel were not blinded to treatment

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

No effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

(from protocol) Progression free survival; Tumor assessments for ongoing study treatment decisions will be completed by the investigator using RECIST (Response Evaluation Criteria in Solid Tumors) 1.1 criteria

Health related quality of life, Minor adverse events; Open label trial

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

All IMDC intermediate and poor risk patients analysed for efficacy as predefined outcome

Incomplete outcome data (attrition bias)
Response rate

Low risk

All IMDC intermediate and poor risk patients analysed for efficacy as predefined outcome

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants analysed

Incomplete outcome data (attrition bias)
Quality of life

Unclear risk

More than 80% completion rate in both groups but final analysed participants were 44/425 (10.3%) in Nivolumab + Ipilimumab arm and 26/422 (6.1%) in sunitinib arm.

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

Low risk

Completed planned accrual

Hudes 2007

Study characteristics

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: June 2003 to April 2005

Blinding: imaging

Strata: according to the geographic location of the centre and nephrectomy status

IMC: an independent data and safety monitoring committee reviewed the study at 6‐month intervals

Crossover: not allowed

Participants

Histology: all histologies

Prior therapy: naïve

Measurable disease: required (RECIST)Non metastatic %: <20M/F: 432/194

Eligible PS: Karnofsky > 50; actual KPS( > 70) = 17%

Age median (range): 59 (23 to 86)

Prior nephrectomy: 419

Prognostic strata: system, % good/intermediate/poor risk: MSKCC, ‐/26/74%

Interventions

TEMSIROLIMUS 25mg IV weekly, Interferon‐a2a 3‐18MU sc tiw, or both

Outcomes

PFS: secondary endpoint

OS: primary endpoint

AE: reported in toxicity table

QoL: reported
RR: (RECIST)

Other:

Funding Sources

Industry sponsored

Declarations of interest

Reported in main publication

Notes

Only poor risk (76%) and intermediate risk (26%) patients included in trial

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomly assigned in equal proportions, with the use of permuted blocks of three, to one of three treatment groups." Central randomisation presumed

Allocation concealment (selection bias)

Low risk

"Patients were randomly assigned in equal proportions, with the use of permuted blocks of three, to one of three treatment groups." Central randomisation presumed

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

Participants and personnel were not blinded to treatment, no effects on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Participants and personnel were not blinded to treatment, active treatment in all 3 groups

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Unblinded clinical investigator assessment, no effects on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

Shorter investigator assessed PFS in comparison to independent radiologic assessment; which is explained by different inclusion criteria

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

Intention‐to‐treat population reported

Incomplete outcome data (attrition bias)
Response rate

High risk

82% of ITT population reported; only selected participants included in analysis which underwent tumour assessment after the baseline

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants reported

Incomplete outcome data (attrition bias)
Quality of life

High risk

65% of participants were evaluable for QoL analysis

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

Low risk

Completed planned accrual; stopped early at the second interim analysis

McDermott 2018

Study characteristics

Methods

Study type: multicentre RCT

Phase: 2

Accrual period: January 2014 to March 2015

Blinding: open label design

Strata: MSKCC risk category, prior nephrectomy status, and PD‐L1 status

IMC: "Independent review facility‐assessed efficacy endpoints"

Crossover: allowed

Participants

Histology: clear cell

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnowsky PS 70% or better

Measurable disease: required

Non metastatic %: not specified

M/F: 230/75

Age median (range): 61

Prior nephrectomy: 184

Prognostic strata: system, good/intermediate/poor risk: MSKCC, 77/201/27

Interventions

atezolizumab 1200 mg IV q3w + bevacizumab 15 mg/kg IV q3w, atezolizumab alone or sunitinib 50 mg PO QD 4 wk on/2 wk off

Outcomes

PFS: co‐primary endpoint

OS: secondary endpoint

AE: reported

QoL: exploratory outcome

RR: secondary endpoint

Other: primary endpoint: percentage of participants with disease progression per response evaluation

Funding Sources

Industry sponsored

Declarations of interest

Published online

Notes

Crossover to atezolizumab + bevacizumab arm allowed on progression

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"interactive voice/web response system (IxRS)" used; "Stratified permuted block randomization was used to assign patients in a 1:1:1 ratio to one of three treatment arms"

Allocation concealment (selection bias)

High risk

"allocation was unmasked"

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"The study was open‐label"; participants and personnel were not blinded to treatment; no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"The study was open‐label"; participants and personnel were not blinded to treatment, all participants received an active treatment with different administration forms

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Independent review facility (IRF)‐assessed efficacy

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

Independent review facility (IRF)‐assessed efficacy, no blinding used

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

ITT population reported, 1 patient did not receive treatment

Incomplete outcome data (attrition bias)
Response rate

Low risk

ITT population reported, 1 patient did not receive treatment

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants included in safety analysis

Incomplete outcome data (attrition bias)
Quality of life

Low risk

"96 (95%) of 101 patients in the atezolizumab plus bevacizumab group and 93 (92%) of 101 patients in the sunitinib
group completed the MDASI at baseline"

Selective reporting (reporting bias)

Low risk

All planned outcomes except for quality of life reported

Other bias

Unclear risk

Industry sponsored

Motzer 2010

Study characteristics

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: August 2004 to October 2005

Blinding: imaging

Strata: LDH, PS, nephrectomy status

IMC: used for data and safety

Crossover: study amended when sunitinib approved in January 2006 to allow cross‐over of patients on IFN on documented disease progression as primary endpoint of PFS had been met ‒ agreed with IMC

Participants

Histology: clear cell

Prior systemic therapy: treatment‐naïve

Measurable disease: required

Non metastatic %: metastatic disease required

M/F %: 536/214

Eligible PS: ECOG 0 to 1

Age median (range): 61 (27 to 87)

Prior nephrectomy: 675

Prognostic strata: system, good/intermediate/poor risk %: MSKCC, 37/56/7%

Interventions

(1) SUNITINIB 50 mg oral daily for 4 weeks of 6‐week cycle; (2) Interferon‐alfa2a
9 MU sc tiw (with cross‐over to SUNITINIB at disease progression, after second interim
analysis)

Outcomes

PFS: primary endpoint

OS: secondary endpoint

AE: toxicity table, secondary endpoint

QoL: secondary endpoint (FACT‐G, FKSI)

RR: secondary endpoint

Other: cross‐over post study

Funding Sources

Industry sponsored

Declarations of interest

Reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

“patients were randomly assigned”, presumed central randomisation

Allocation concealment (selection bias)

Unclear risk

Data not found

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

Open label study design, participants and personnel were not blinded to treatment, no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Open label study design, participants and personnel were not blinded to treatment, 2 active treatments used with different administration forms

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Assessed by investigator, no effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

"A blinded central review of radiologic images was used to assess the primary end point and the objective response rate"

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

8% of patients withdrew consent on the control arm (vs 1%, P < 0.001) but primary end‐ point was analysed by allocation

Incomplete outcome data (attrition bias)
Response rate

Low risk

All randomised participants analysed

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants analysed

Incomplete outcome data (attrition bias)
Quality of life

Unclear risk

No information on how many participants completed questionnaire found

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

High risk

Cross‐over permitted after second interim analysis but planned accrual had been completed, OS analysis secondary endpoint

Motzer 2013a

Study characteristics

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: Aug 2008 to Sep 2011

Blinding: open label design

Strata: Karnofsky PS, LDH, nephrectomy status

IMC: safety reviewed

Crossover: not allowed

Participants

Histology: clear cell component

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnofsky 70% or more

Measurable disease: required

Non metastatic %: < 20

M/F: 813/297

Age median (range): 61 (18 to 88)

Prior nephrectomy: 924

Prognostic strata: system, good/intermediate/poor risk: MSKCC, 303/650/119

Interventions

pazopanib 800 mg po vs sunitinib 50 mg po 4 wks on, 2 wks off

Outcomes

PFS: primary outcome as non‐inferiority measure

OS: secondary outcome

AE: reported in toxicity table, secondary outcome

QoL: secondary outcome

RR: secondary outcome

Other: medical resource utilization

Funding Sources

Industry sponsored

Declarations of interest

Published online

Notes

Non‐inferiority design

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

“GSK interactive voice response system called RAMOS (Registration And Medication Ordering System), by the investigator or authorized site staff for stratification and central randomization.” from protocol

Allocation concealment (selection bias)

Low risk

“GSK interactive voice response system called RAMOS (Registration And Medication Ordering System), by the investigator or authorized site staff for stratification and central randomization.” from protocol

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

Open label study design, participants and personnel were not blinded to treatment, OS reported as secondary outcome

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Open label study design, participants and personnel were not blinded to treatment, oral administered, active drugs used in both arms

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Not specifically reported but no effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

"The primary end point was progression‐free survival as assessed by independent review"; tumour response and PFS assessed independently, lack of blinding

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

ITT population assessed, 8 patients not treated

Incomplete outcome data (attrition bias)
Response rate

Low risk

ITT population assessed, 8 patients not treated

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants assessed

Incomplete outcome data (attrition bias)
Quality of life

High risk

High losses in questionnaire completion rates

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

Unclear risk

Protocol amended to increase sample size for planned events, non‐inferiority design

Motzer 2013b

Study characteristics

Methods

Study design: multicentre RCT

Phase: 3

Accrual period: February 2010 to August 2010

Blinding: open label design

Strata: region, number of prior treatments, number of metastatic sites and organs involved

IMC: data monitored not specified

Crossover: at progression

Participants

Histology: clear cell component

Prior systemic therapy: treatment‐naïve; except for immunotherapy, chemotherapy, or hormonal therapy

Eligible PS: ECOG PS 1 or better

Measurable disease: required

Non metastatic %: ≤ 21

M/F: 374/143

Age median (range): 59 (23 to 85)

Prior nephrectomy: required

Prognostic strata: system, good/intermediate/poor risk: MSKCC 157/333/27

Interventions

tivozanib 1.5 mg, 3 wks on/1 wks off vs sorafenib 400 mg bid

Outcomes

PFS: primary outcome

OS: secondary outcome

AE: secondary outcome

QoL: secondary outcome

RR: secondary outcome

Other: tolerability, kidney specific symptoms

Funding Sources

Industry sponsored

Declarations of interest

Reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

“An Interactive Voice Response / Interactive Web Response (IVR/IWR) system will be used for enrolment, randomization and drug management” from protocol

Allocation concealment (selection bias)

Low risk

“An Interactive Voice Response / Interactive Web Response (IVR/IWR) system will be used for enrolment, randomization and drug management” from protocol; central randomization

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"This was an open‐label, randomized phase III trial"; participants and personnel were not blinded to treatment, no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"This was an open‐label, randomized phase III trial"; participants and personnel were not blinded to treatment; oral administered, active drugs used in both arms

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Assessment not specified; no effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

"The primary end point was progression‐free survival (PFS) by independent review", open label trial design

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

ITT population analysed, 1 patient not treated in experimental arm

Incomplete outcome data (attrition bias)
Response rate

Low risk

ITT population analysed, 1 patient not treated in experimental arm

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants assessed for safety

Incomplete outcome data (attrition bias)
Quality of life

Low risk

"HRQoL questionnaires were completed by more than 99% of patients in both arms at baseline. Completion rates decreased over time, in line with study dropout, falling below 50% after cycle 13"

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

High risk

Possible confounding of OS data by cross‐over design

Motzer 2014

Study characteristics

Methods

Study design: multicentre RCT

Phase: 2

Accrual period: October 2009 to June 2011

Blinding: open label design

Strata: MSKCC risk category

IMC: data not found

Crossover: within treatment arms from first to second line

Participants

Histology: any

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnofsky PS 70% or better

Measurable disease: required

Non metastatic %: not specified

M/F: 342/129

Age median (range): 62 (20 to 89)

Prior nephrectomy: 315

Prognostic strata: system, good/intermediate/poor risk: MSKCC, 139/263/67

Interventions

first line: everolimus 10 mg po; second line: sunitinib 50 mg po 4 wks on, 2 wks off vs first line: sunitinib 50 mg po 4 wks on, 2 wks off; second line everolimus 10 mg po

Outcomes

PFS: primary outcome for first line treatment

OS: secondary outcome

AE: reported in toxicity table

QoL: reported

RR: assessed as secondary outcome

Other: tolerability, PFS after second line treatment

Funding Sources

Industry sponsored

Declarations of interest

Reported

Notes

Non‐inferiority design

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomly assigned in a 1:1 manner; Interactive Voice Response System (IVRS) to randomize the patient"

Allocation concealment (selection bias)

Low risk

Patient randomisation list used; described in detail

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"RECORD‐3 [...] was an open‐label, randomized, multicenter, phase II study...", no effects on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"RECORD‐3 [...] was an open‐label, randomized, multicenter, phase II study..."; same drugs with different sequence used in both arms but participants and personnel were not blinded to treatment

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Assessment not specified, no effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

"The primary end point was to assess progression‐free survival (PFS) non‐inferiority of first‐line everolimus compared with sunitinib by investigator assessment"

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

ITT population reported, 2 patients in control arm did not receive treatment

Incomplete outcome data (attrition bias)
Response rate

Low risk

ITT population reported, 2 patients in control arm did not receive treatment

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants reported

Incomplete outcome data (attrition bias)
Quality of life

Low risk

High initial questionnaire completion rate, decreased in both arms over time

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

High risk

Difference in baseline performance status, non‐inferiority design

Motzer 2019

Study characteristics

Methods

Study design: multicentre RCT

Phase: 3

Accrual period: March 2016 to December 2017

Blinding: open label design

Strata: ECOG PS (0 or 1), geographic region

IMC: external data monitoring committee used for efficacy and safety

Crossover: not planned

Participants

Histology: clear cell renal cell carcinoma

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnofsky PS 70% or better

Measurable disease: required

Non metastatic %: not specified

M/F: 660/226

Age median (range): 61 (27 to 88)

Prior nephrectomy: 707

Prognostic strata: system, good/intermediate/poor risk: MSKCC, 196/576/96

Interventions

Avelumab administered at 10 mg/kg IV every 2 weeks in combination with Axitinib, 5 mg PO BID versus Sunitinib given at 50 mg PO QD on schedule 4/2

Outcomes

PFS: primary outcome among patients with PD‐L1–positive tumours, secondary endpoint for overall population

OS: primary outcome among patients with PD‐L1–positive tumours, secondary endpoint for overall population

AE: reported in toxicity table

QoL: not assessed

RR: assessed as secondary outcome

Other: pharmacokinetic measures, tumour‐tissue biomarker

Funding Sources

Industry sponsored

Declarations of interest

Reported online

Notes

Primary endpoints changed after protocol amendment in June 2017 to evaluate PFS or OS for PD‐L1 positive participants

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"the study treatment assignment using the Interactive Response Technology (IRT) system (interactive web‐based response [IWR]/interactive voice response [IVR] system)" from protocol

Allocation concealment (selection bias)

Low risk

"the study treatment assignment using the Interactive Response Technology (IRT) system (interactive web‐based response [IWR]/interactive voice response [IVR] system)" from protocol

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"This was a multicenter, randomized, open‐label, phase‐3 trial..."; open label trial design, no effects on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"This was a multicenter, randomized, open‐label, phase‐3 trial..."; different drugs and administration form, participants and personnel were not blinded to treatment

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

OS assessment of PD‐L1 positive population. The evaluation of PD‐L1 status a priori to assessment of OS is not considered as objective. Trial design is open‐label. However, we extracted OS result from overall population.

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

Primary and main secondary endpoints determined by blinded independent central review

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

ITT and PD‐L1 population reported separately for PFS, 8 and 5 participants did not receive treatment

Incomplete outcome data (attrition bias)
Response rate

Low risk

ITT and PD‐L1 population reported separately, 8 and 5 participants did not receive treatment

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants reported in toxicity table

Selective reporting (reporting bias)

Unclear risk

OS data not mature at time of publication, protocol amended after study start but initially planned outcomes reported

Other bias

Low risk

Planned accrual completed, protocol changes after study start.

Ravaud 2015

Study characteristics

Methods

Study type: multicentre RCT

Phase: 2

Accrual period: data not found

Blinding: open label design

Strata: MSKCC risk category

IMC: used to analyse tumour responses

Crossover: not allowed

Participants

Histology: predominantly clear‐cell mRCC

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnofsky PS 70% or better

Measurable disease: required

Non metastatic %: 0, metastatic disease mandatory

M/F: 269/96

Age median (range): 60 (20 to 84)

Prior nephrectomy: partial or radical nephrectomy mandatory

Prognostic strata: system, good/intermediate/poor risk: MSKCC, 131/208/26

Interventions

Everolimus 10 mg po and bevacizumab 10mg/kg iv every 2 wks vs IFN 9 MIU 3 times per wk plus bevacizumab 10 mg/kg every 2 weeks

Outcomes

PFS: primary endpoint

OS: secondary outcome

AE: secondary outcome, reported in toxicity table

QoL: not assessed

RR: secondary outcome

Other: duration of response

Funding Sources

Industry sponsored

Declarations of interest

Reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"randomized 1:1"

Allocation concealment (selection bias)

Unclear risk

Data not found

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"open‐label, phase II RECORD‐2 trial"; personnel and participants were not blinded to treatment, different drug administration forms used, no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

Unclear risk

"open‐label, phase II RECORD‐2 trial"; personnel and participants were not blinded to treatment, different drug administration forms used

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Tumor response and progression were evaluated by the local radiologist and independent central review

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

Tumor response and progression were evaluated by the local radiologist and independent central review; trial design is open‐label

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

ITT population reported, 3 patients did not receive treatment

Incomplete outcome data (attrition bias)
Response rate

Low risk

ITT population reported, 3 patients did not receive treatment

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All but 1 participant who received treatment were included in safety analysis

Incomplete outcome data (attrition bias)
Quality of life

Unclear risk

No available data

Selective reporting (reporting bias)

Low risk

All outcomes reported

Other bias

Unclear risk

Study not powered to show significant treatment effect

Retz 2019

Study characteristics

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: June 2012 to November 2016

Blinding: open label design

Strata: low versus intermediate MSKCC risk score, clear cell versus non‐clear cell histology

IMC: data not found

Crossover: from first to second‐line after disease progression

Participants

Histology: any

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnofsky PS 70% or better

Measurable disease: required

Non metastatic %: data not found

M/F: 189/188

Age median (range): 68 (26 to 86)

Prior nephrectomy: 328

Prognostic strata: system, good/intermediate/poor risk: MSKCC, 186/179/9

Interventions

Sorafenib 400 mg bid orally until progression or intolerable toxicity, followed by pazopanib 800 mg once daily orally until progression or intolerable toxicity or vice versa

Outcomes

PFS: primary outcome

OS: reported

AE: reported

QoL: planned

RR: planned

Other: time to first‐line treatment failure, biomarker

Funding Sources

Technische Universität München

Declarations of interest

Reported online

Notes

MSKCC PS low or intermediate patients only, non‐inferiority study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"377 patients were randomised"

Allocation concealment (selection bias)

Unclear risk

Data not found

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"Phase III randomized, sequential, open‐label study"; participants and personnel were not masked to treatment; no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"Phase III randomized, sequential, open‐label study"; participants and personnel were not masked to treatment; same drugs used in both arms

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Open label study design but no effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

Unclear risk

Data not found

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

Intention to treat population analysed

Incomplete outcome data (attrition bias)
Response rate

Low risk

Intention to treat population analysed

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

83/189 (96.8%) and 183/188 (97.3%) population analysed in each arm

Incomplete outcome data (attrition bias)
Quality of life

High risk

136/189 (71.9%) and 131/188 (69.6%) population analysed in each arm

Selective reporting (reporting bias)

Unclear risk

Quality of life outcome not published in conference abstract

Other bias

Low risk

Not detected

Rini 2008

Study characteristics

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: October 2003 to July 2005

Blinding: open label design

Strata: nephrectomy status; prognostic risk

IMC: Data Safety Monitoring Board

Crossover: data not found

Participants

Histology: clear cell

Prior systemic therapy: naïve

Measurable disease: measurable or non‐measurable disease

Non metastatic %: not specified

M/F %: 508/224

Eligible PS: Karnofsky; 100 to 70%

Age median: 62

Prior nephrectomy: 620

Prognostic strata: system, good/intermediate/poor risk %: MSKCC, 26/64/10

Interventions

INTERFERON alfa‐2b (Intron, Schering‐Plough) 9 MU SC TIW (1) with, or (2)
without, BEVACIZUMAB 10 mg/kg IV Q2weeks

Outcomes

PFS: secondary endpoint

OS: primary endpoint

AE: secondary endpoint, reported in toxicity table

QoL: not assessed

RR: secondary endpoint

Other:

Funding Sources

Independent sponsoring

Declarations of interest

Reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"A stratified random block design was used"; Central randomisation by cooperative groups (CALGB and NCI‐Canada)

Allocation concealment (selection bias)

Low risk

Central randomisation

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"There was no placebo infusion in this non blinded trial", participants and personnel were not blinded to treatment, no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"There was no placebo infusion in this non blinded trial", participants and personnel were not blinded to treatment

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

"no independent review of radiographs"; Investigator assessment of total population, overall survival primary outcome assumed reliable

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

"no independent review of radiographs", assessed by investigator

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

All patients accounted for with small symmetric losses

Incomplete outcome data (attrition bias)
Response rate

Low risk

All patients accounted for with small symmetric losses

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

On treated participant not reported

Selective reporting (reporting bias)

Low risk

All protocol outcomes reported

Other bias

Low risk

Planned accrual completed; independently sponsored and conducted

Rini 2014

Study characteristics

Methods

Study design: multicentre RCT
Phase: 3
Accrual period: April 2008 to October 2010
Blinding: open label design
Strata: nephrectomy status, MSKCC risk category
IMC: external data monitoring committee used
Crossover: not allowed

Participants

Histology: clear cell component
Prior systemic therapy: treatment‐naïve
Eligible PS: Karnofsky PS 70% or better
Measurable disease: yes
Non metastatic %: data not found
M/F: 556/235
Age median (range): 58 (22 to 87)
Prior nephrectomy: 673
Prognostic strata: system, good/intermediate/poor risk: MSKCC, 237/467/87

Interventions

Temsirolimus 25 mg iv weekly plus Bevacizumab 10 mg/kg iv every 2 weeks or IFN 9 million U[MIU] subcutaneously thrice weekly plus Bevacizumab 10 mg/kg IV every 2 weeks

Outcomes

PFS: independent assessment as primary endpoint
OS: secondary endpoint
AE: secondary endpoint
QoL: assessed as exploratory objective
RR: secondary endpoint
Other: "Disease‐related symptoms and quality of life were assessed as exploratory objectives."

Funding Sources

Industry sponsored

Declarations of interest

Reported in main publication

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"A computerized centrally located randomization system was used"

Allocation concealment (selection bias)

Low risk

"A computerized centrally located randomization system was used"

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"randomized, open‐label, multicenter, phase III study"; participants and personnel were not blinded to treatment, no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"randomized, open‐label, multicenter, phase III study"; participants and personnel were not blinded to treatment

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

"Secondary end points were investigator‐assessed PFS, independently assessed ORR, OS, and safety" no effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

"Radiographic evaluations were conducted at screening and every 8 weeks, and tumor progression was assessed both by investigators and by an independent blinded assessment"

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

ITT population reported, 7 patients in 1 arm did not receive treatment

Incomplete outcome data (attrition bias)
Response rate

Low risk

ITT population reported, 7 patients in 1 arm did not receive treatment

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants reported in toxicity table

Incomplete outcome data (attrition bias)
Quality of life

Low risk

"Completion rate for each questionnaire was uniformly high in both treatment arms, with rates above 90% among patients on treatment up to the end of treatment visit" assessed as exploratory outcome only

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

Unclear risk

Planned accrual almost completed (791 versus 800)

Rini 2016

Study characteristics

Methods

Study design: multicentre RCT

Phase: 3

Accrual period: December 2010 to December 2012

Blinding: open label design

Strata: IMDC risk group, region, nephrectomy status

IMC: data and safety monitored

Crossover: not allowed

Participants

Histology: clear cell component

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnowsky PS 80% or better

Measurable disease: required

Non metastatic %: ≤ 28

M/F: 230/109

Age median (range): 61 (54 to 69)

Prior nephrectomy: 306

Prognostic strata: system, good/intermediate/poor risk %: IMDC, 91/241/7

Interventions

Sunitinib 50 mg po 4 wks on, 2 wks off vs sunitinib 50 mg po 4 wks on, 2 wks off and Cyclophosphamide (1 dose of 300 mg/m² iv) at visit D (3 days before the first vaccination at visit 1) and the vaccination schedule (GM‐CSF 75 μg and IMA901 4.13 mg intradermally at each vaccination) included 6 vaccinations within the first 3 weeks (visits 1 to 6 on days 1, 2, 3, 8, 15, and 22, respectively) and a further 4 vaccinations at 3‐week intervals (visits 7 to 10 on days 43, 64, 85, and 106, respectively)

Outcomes

PFS: secondary outcome

OS: primary endpoint

AE: reported in toxicity table, secondary outcome

QoL: not assessed

RR: secondary outcome

Other: biomarker related OS

Funding Sources

Industry sponsored

Declarations of interest

Reported

Notes

Only favourable or intermediate risk according to the International Metastatic Database Consortium (IMDC) risk criteria patients included, all patients treated with sunitinib and evaluated for efficacy before randomisation.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"...randomised centrally with an interactive web response system..."

Allocation concealment (selection bias)

Low risk

"...randomised centrally with an interactive web response system..."

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"patients and investigators were not masked to treatment allocation", open label study design, no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"patients and investigators were not masked to treatment allocation", open label study design

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

"The primary outcome was overall survival from randomisation until death of any cause as determined by local investigators"; no effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

"progression‐free survival from randomisation according to blinded, independent central review", "Best tumour response was assessed according to RECIST 1.1 and was based on centrally reviewed tumour images"

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

ITT population reported, 8 patients did not receive treatment

Incomplete outcome data (attrition bias)
Response rate

Low risk

All randomised participants analysed

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants analysed

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

High risk

All participants treated with 1 of the study drugs in run in phase, only responders randomised

Rini 2019a

Study characteristics

Methods

Study design: multicentre RCT

Phase: 3

Accrual period: October 2016 to January 2018

Blinding: open label design

Strata: IMDC risk group (favourable, intermediate, or poor risk) and geographic region

IMC: an independent data and safety monitoring committee oversaw the trial

Crossover: not planned

Participants

Histology: clear‐cell renal‐cell carcinoma

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnowsky PS 70% or better

Measurable disease: required

Non metastatic %: < 99

M/F: 628/233

Age median (range): 62 (26 to 90)

Prior nephrectomy: 715

Prognostic strata: system, good/intermediate/poor risk %: IMDC, 269/484/108

Interventions

Pembrolizumab 200 mg intravenously every 3 weeks plus Axitinib 5 mg orally twice daily versus Sunitinib 50 mg orally once daily for 4 weeks and then are off treatment for 2 weeks

Outcomes

PFS: co‐primary outcome assessed by blinded, independent central review

OS: co‐primary outcome assessed by blinded, independent central review

AE: reported in toxicity table, secondary outcome

QoL: not assessed

RR: secondary outcome assessed by blinded, independent central review

Other: duration of response

Funding Sources

Industry sponsored

Declarations of interest

Reported online

Notes

Planned accrual completed, primary endpoint for OS not met because of short follow up

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Treatment randomization will occur centrally using an interactive voice response system / integrated web response system (IVRS/IWRS)" from protocol

Allocation concealment (selection bias)

Low risk

"Treatment randomization will occur centrally using an interactive voice response system / integrated web response system (IVRS/IWRS)" from protocol; central randomisation

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"In this open‐label, phase 3 trial..." participants and personnel were not blinded to treatment, no effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"In this open‐label, phase 3 trial..." different drugs and administration form, participants and personnel were not blinded to treatment

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

OS assessed by blinded, independent central review

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

Subjective primary and secondary efficacy endpoints assessed by blinded, independent central review

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

Intention to treat population analysed; < 1% in both groups did not receive treatment

Incomplete outcome data (attrition bias)
Response rate

Low risk

Intention to treat population analysed; < 1% in both groups did not receive treatment

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants reported in toxicity table

Selective reporting (reporting bias)

Low risk

All planned outcomes reported, no mature OS data available

Other bias

Low risk

Not detected

Rini 2019b

Study characteristics

Methods

Study design: multicentre RCT

Phase: 3

Accrual period: May 2015 to October 2016

Blinding: open label design

Strata: PD‐L1 expression, presence of liver metastasis, MSKCC risk category

IMC: "An independent data monitoring committee reviewed safety data during the study on a periodic basis"

Crossover: "No prespecified crossover was planned per protocol"

Participants

Histology: "clear‐cell histology and/or a component of sarcomatoid carcinoma"

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnowsky PS 70% or better

Measurable disease: required

Non metastatic %: not specified

M/F: 669/246

Age median (range): 61 (54 to 69)

Prior nephrectomy: 664

Prognostic strata: system, good/intermediate/poor risk %: MSKCC, 179/629/107

Interventions

Atezolizumab administered at a fixed dose of 1200 milligrams (mg) via intravenous (IV) infusion on Days 1 and 22 of each 42‐day cycle in combination with Bevacizumab administered at a dose of 15 milligrams per kilogram (mg/kg) via IV infusion on Days 1 and 22 of each 42‐day cycle versus Sunitinib administered at a dose of 50 mg once daily, orally via capsule, on Day 1 through Day 28 of each 42‐day cycle.

Outcomes

PFS: co‐primary outcome by investigator assessment in patients with PD‐L1 positive disease, secondary outcome in the ITT population

OS: co‐primary outcome in the intention‐to‐treat population, secondary outcome in the PD‐L1 positive population

AE: secondary outcome

QoL: reported

RR: secondary outcome

Other:

Funding Sources

Industry sponsored

Declarations of interest

Reported in main publication

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomly assigned (1:1) via an interactive voice and web response system to receive Atezolizumab plus Bevacizumab or Sunitinib." central randomisation presumed

Allocation concealment (selection bias)

Low risk

Central randomisation presumed

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"The study was open label, and investigators and participants were not masked to treatment allocation" No effect on OS expected

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

"The study was open label, and investigators and participants were not masked to treatment allocation", different drugs and forms of administration used in experimental and control arm

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Assessment not specified but not effect on OS expected

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

"Co‐primary endpoints were progression‐free survival (RECIST 1.1) by investigator assessment in patients with PD‐L1 positive disease" PFS results in PD‐L1 population differ from independent assessment

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

Intention to treat population reported, 3 participants in experimental and 15 participants in control arm did not receive treatment

Incomplete outcome data (attrition bias)
Response rate

Low risk

Intention to treat population reported, 3 participants in experimental and 15 participants in control arm did not receive treatment

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants reported

Incomplete outcome data (attrition bias)
Quality of life

Unclear risk

"386 (86%) of 451 patients in the atezolizumab plus bevacizumab group and 369 (83%) of 446 patients in the sunitinib
group completed the MDASI at baseline"

Selective reporting (reporting bias)

Low risk

All planned outcomes reported

Other bias

Low risk

Planned accrual completed

Sternberg 2010

Study characteristics

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: April 2006 to April 2007

Blinding: double‐blind, placebo‐controlled

Strata: ECOG PS 0vs1; nephrectomy status; prior cytokine

IMC: responsible for safety monitoring and to review interim overall survival data

Crossover: allowed from placebo to active treatment

Participants

Histology: clear cell

Prior systemic therapy: 1 line of cytokines permitted.

Measurable disease: required

Non metastatic %: < 18

M/F: 307/128

Eligible PS: ECOG 0 to 1

Age median(range): 59 (25 to 85)

Prior nephrectomy: 385

Prognostic strata: system, good/intermediate/poor risk %: MSKCC; 39/54/3

Interventions

(1) PAZOPANIB 800 mg PO daily, vs (2) matched PLACEBO (2:1 randomization) Cross‐over 48%

Outcomes

PFS: primary endpoint

OS: principal secondary end point

AE: toxicity table available, additional secondary end point

QoL: reported

RR: additional secondary end point

Other:

Funding Sources

Industry sponsored

Declarations of interest

Reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were centrally randomly assigned in a 2:1 ratio..."; central randomisation

Allocation concealment (selection bias)

Low risk

"Patients were centrally randomly assigned in a 2:1 ratio..."; central randomisation

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

"was a placebo‐controlled, randomized, double‐blind, global, multicenter, phase III study"; participants and personnel were masked to treatment

Blinding of participants and personnel (performance bias)
Subjective outcomes

Low risk

"was a placebo‐controlled, randomized, double‐blind, global, multicenter, phase III study"; participants and personnel were masked to treatment

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

"All imaging scans were evaluated by an independent imaging‐review committee (IRC) blinded to study treatment"

Blinding of outcome assessment (detection bias)
Subjective outcomes

Low risk

"All imaging scans were evaluated by an independent imaging‐review committee (IRC) blinded to study treatment"

Incomplete outcome data (attrition bias)
PFS, OS

Low risk

All patients accounted for, losses 6% (investigational) and 3% (control) unlikely significant, “completion rates > 90% across most of the assessment time points”

Incomplete outcome data (attrition bias)
Response rate

Low risk

All patients accounted for, losses 6% (investigational) and 3% (control) unlikely significant, “completion rates > 90% across most of the assessment time points”

Incomplete outcome data (attrition bias)
Serious and minor adverse events

Low risk

All treated participants analysed

Incomplete outcome data (attrition bias)
Quality of life

Low risk

"Completion rates for QoL questionnaires were high across most of the assessment time points for each instrument"

Selective reporting (reporting bias)

Low risk

All protocol specified endpoints reported

Other bias

Low risk

Planned accrual completed

AE: adverse event; ECOG: Eastern Cooperative Oncology Group; F: female; IMC: independent monitoring committee; IMDC: International Metastatic Renal Cell Carcinoma Database Consortium; ITT: intention to treat; M: male; MSKCC: Memorial Sloan Kettering Cancer Center; OS: overall survival; PFS: progression‐free survival; PS: performance status; QoL: quality of life; RCT: randomised controlled trial; RR: response rate; wks: weeks

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Armstrong 2016

Non‐clear cell renal cell carcinoma participants only, less than 100 participants per treatment arm included

Atkins 2004

Same targeted drug used in all treatment arms, less than 100 participants per treatment arm included, cytokine pretreatment required

Bracarda 2010

Same targeted treatment used in both arms, less than 100 participants per treatment arm included

Broom 2015

Less than 100 participants per treatment arm included

Bukowski 2007

Less than 100 participants per treatment arm included

Choueiri 2015

Participants were not treatment‐naïve

Choueiri 2017

Less than 100 participants per treatment arm included

Cirkel 2016

Less than 100 participants per treatment arm included

Dorff 2015

Participants not treatment‐naïve, less than 100 participants per treatment arm included

Ebbinghaus 2007

Same drug used in both treatment arms, less than 100 participants per treatment arm included

Eisen 2015

Less than 100 participants per treatment arm included

Escudier 2007

Treatment is not considered as targeted therapy

Escudier 2009

Less than 100 participants per treatment arm included

Escudier 2010a

Participants were not treatment naïve

Flaherty 2015

Less than 100 participants per treatment arm included

Gordon 2004

No treatment that is considered as targeted therapy was used in trial

Hainsworth 2015

Less than 100 participants per treatment arm included

Hawkins 2016

Treatment not considered as targeted therapy, any renal cell carcinoma histologies included

Hutson 2013

Participants were not treatment naïve

Hutson 2014

Participants were not treatment naïve

Jonasch 2010

Less than 100 participants per treatment arm included

Jonasch 2017

Participants were not treatment naïve

Lee 2006

Treatment not considered as targeted therapy

Lee 2015

Same study drugs used in both arms, less than 100 participants per treatment arm included

Madhusudan 2004

Treatment not considered as targeted therapy

Motzer 2008

Participants were not treatment‐naïve

Motzer 2012

Same study drug used in both arms

Motzer 2014b

Participants were not treatment‐naïve

Motzer 2015a

Participants were not treatment‐naïve

Motzer 2015b

Participants were not treatment‐naïve

Motzer 2015c

Participants were not treatment‐naïve

Mulders 2012

Less than 100 participants per treatment arm included

Nosov 2012

Less than 100 participants per treatment arm included

Négrier 2011

Less than 100 participants per treatment arm included

Pal 2015

Less than 100 participants per treatment arm included, participants were not treatment‐naïve

Pili 2015

Less than 100 participants per treatment arm included, participants were not treatment‐naïve

Powles 2014

Less than 100 participants per treatment arm included, participants were not treatment‐naïve

Powles 2016a

Less than 100 participants per treatment arm included, participants were not treatment‐naïve

Powles 2016b

Less than 100 participants per treatment arm included, participants were not treatment‐naïve

Procopio 2011

Less than 100 participants per treatment arm included

Ratain 2006

Less than 100 participants per treatment arm included, 84% of participants pretreated

Ravaud 2008

Treatment in control arm not eligible for comparisons

Rini 2011

Participants were not treatment‐naïve

Rini 2012

Treatment in observational and control arm not eligible for comparison

Rini 2013

Less than 100 participants per treatment arm included

Srinivas 2005

Treatment not considered as targeted therapy

Stadler 2005

Treatment is not considered as targeted therapy

Tannir 2016

Less than 100 participants per treatment arm included, non‐clear cell renal cell carcinoma only

Tannir 2018

Less than 100 participants per treatment arm included

Tomita 2014

Less than 100 participants per treatment arm included

Twardowski 2015

Less than 100 participants per treatment arm included, participants were not treatment‐naïve

Yang 2003

Less than 100 participants per treatment arm included, participants were not treatment‐naïve

Characteristics of ongoing studies [ordered by study ID]

Choueiri 2018

Study name

NCT03141177, CheckMate 9ER

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: data not found

Blinding: open label study design

Strata: IMDC risk score, PD‐1 ligand 1 (PD‐L1) tumour expression and geographic region

IMC: data not found

Crossover: data not found

Participants

Histology: clear cell component

Prior systemic therapy: no prior systemic therapy for RCC

Measurable disease: required

Eligible PS: data not found

Non metastatic %: data not found

M/F %: data not found

Age median(range): data not found

Prior nephrectomy: data not found

Prognostic strata: system, good/intermediate/poor risk %: data not found

Interventions

Nivolumab and cabozantinib versus sunitinib

Outcomes

PFS: primary endpoint

OS: secondary end point

AE: secondary end point

QoL: not planned

RR: secondary end point

Other:

Starting date

July 2017

Contact information

Notes

Choueiri 2019

Study name

NCT03937219, Cosmic‐313

Methods

Study type: multicentre RCT

Phase: 3

Accrual period: 25 June 2019 ‐

Blinding: double‐blind study design

Strata: IMDC risk score and geographic region

IMC: data not found

Crossover: data not found

Participants

Histology: clear cell component

Prior systemic therapy: no prior systemic therapy for RCC

Measurable disease: required

Eligible PS: Karnofsky PS 70% or better

Non metastatic %: data not found

M/F %: data not found

Age median (range): data not found

Prior nephrectomy: data not found

Prognostic strata: system, good/intermediate/poor risk %: data not found

Interventions

Cabozantinib + nivolumab + ipilimumab (4 doses) followed by cabozantinib + nivolumab vs Cabozantinib‐matched placebo + nivolumab + ipilimumab (4 doses) followed by cabozantinib‐matched placebo + nivolumab

Outcomes

PFS: primary endpoint

OS: secondary end point

AE:

QoL:

RR:

Other:

Starting date

June 2019

Contact information

Notes

Grünwald 2018

Study name

NCT02959554, NIVOSWITCH

Methods

Study design: multicentre RCT

Phase: 2

Accrual period: data not found

Blinding: open label study design

Strata: data not found

IMC: data not found

Crossover: data not found

Participants

Histology: clear cell component

Prior systemic therapy: First‐line treatment with a TKI for 10‐12 weeks (limited to sunitinib or pazopanib)

Eligible PS: ECOG‐PS 0‐2

Measurable disease: required

Non metastatic %: data not found

M/F %: data not found

Age median, years: data not found

Prior nephrectomy: data not found

Prognostic strata: system, good/intermediate/poor risk %: data not found

Interventions

Nivolumab: 240 mg iv on D1 of every cycle (Q2W) for 16 weeks. After 16 weeks 480 mg iv on D1 of every cycle (Q4W) until disease progress, intolerable toxicity, withdrawal of consent or end of study versus Sunitinib: According to Standard of Care (SOC). Recommended dose is 50 mg PO once daily for 4 consecutive weeks followed by a 2‐week rest period (schedule 4/2) to comprise a complete cycle of 6 weeks (until disease progress, intolerable toxicity, withdrawal of consent or end of study) or Pazopanib: According to Standard of Care (SOC). Recommended dose is 800 mg PO daily continuously (until disease progress, intolerable toxicity, withdrawal of consent or end of study)

Outcomes

PFS: secondary endpoint

OS: primary end point

AE: secondary end point

QoL: secondary endpoint

RR: secondary end point

Other:

Starting date

December 2016

Contact information

Principal Investigator: Prof. Dr. Viktor Grünwald

Notes

Motzer 2018

Study name

NCT02811861

Methods

Study design: multicentre RCT

Phase: 3

Accrual period: data not found

Blinding: open label design

Strata: data not found

IMC: data not found

Crossover: data not found

Participants

Histology: clear cell carcinoma

Prior systemic therapy: treatment‐naïve

Eligible PS: Karnofsky PS 70% or better

Measurable disease: required

Non metastatic %: data not found

M/F %: data not found

Age median, years: data not found

Prior nephrectomy: data not found

Prognostic strata: system, good/intermediate/poor risk %: data not found

Interventions

lenvatinib 18 milligrams (mg) administered orally, once daily, plus everolimus 5 mg administered orally, once daily; Lenvatinib 20 mg administered orally, once daily, plus pembrolizumab 200 mg administered intravenously (IV), every 3 weeks; Sunitinib 50 mg administered orally, once daily, on a schedule of 4 weeks on treatment followed by 2 weeks off treatment

Outcomes

PFS: primary endpoint

OS: planned

AE: planned

QoL: planned

RR: primary endpoint

Other:

Starting date

Trial start date: 13 October 2016

Trial completion date: Estimated 15 January 2020

Contact information

Responsible party/ principal investigator: Eisai Inc.

Notes

Study based on results of phase 2 study data

AE: adverse event; ECOG: Eastern Cooperative Oncology Group; F: female; IMC: independent monitoring committee; IMDC: International Metastatic Renal Cell Carcinoma Database Consortium; IV; intravenous; M: male; OS: overall survival; PFS: progression‐free survival; PO; per oral; PS: performance status; QoL: quality of life; RCC: renal cell carcinoma; RR: response rate; TKI: tyrosine kinase inhibitor

Data and analyses

Open in table viewer
Comparison 1. Sorafenib versus Sunitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 1.1

Comparison 1: Sorafenib versus Sunitinib, Outcome 1: Progression‐free survival

Comparison 1: Sorafenib versus Sunitinib, Outcome 1: Progression‐free survival

1.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 1.2

Comparison 1: Sorafenib versus Sunitinib, Outcome 2: Overall survival

Comparison 1: Sorafenib versus Sunitinib, Outcome 2: Overall survival

1.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 1.3

Comparison 1: Sorafenib versus Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 1: Sorafenib versus Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

1.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 1.4

Comparison 1: Sorafenib versus Sunitinib, Outcome 4: Response rate

Comparison 1: Sorafenib versus Sunitinib, Outcome 4: Response rate

1.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 1.5

Comparison 1: Sorafenib versus Sunitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 1: Sorafenib versus Sunitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 2. Pazopanib versus Sunitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 2.1

Comparison 2: Pazopanib versus Sunitinib, Outcome 1: Progression‐free survival

Comparison 2: Pazopanib versus Sunitinib, Outcome 1: Progression‐free survival

2.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 2.2

Comparison 2: Pazopanib versus Sunitinib, Outcome 2: Overall survival

Comparison 2: Pazopanib versus Sunitinib, Outcome 2: Overall survival

2.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 2.3

Comparison 2: Pazopanib versus Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 2: Pazopanib versus Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

2.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.4

Comparison 2: Pazopanib versus Sunitinib, Outcome 4: Health‐related quality of life

Comparison 2: Pazopanib versus Sunitinib, Outcome 4: Health‐related quality of life

2.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 2.5

Comparison 2: Pazopanib versus Sunitinib, Outcome 5: Response rate

Comparison 2: Pazopanib versus Sunitinib, Outcome 5: Response rate

2.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 2.6

Comparison 2: Pazopanib versus Sunitinib, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 2: Pazopanib versus Sunitinib, Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 3. Tivozanib versus Sorafenib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

3.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 3.1

Comparison 3: Tivozanib versus Sorafenib, Outcome 1: Progression‐free survival

Comparison 3: Tivozanib versus Sorafenib, Outcome 1: Progression‐free survival

3.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 3.2

Comparison 3: Tivozanib versus Sorafenib, Outcome 2: Overall survival

Comparison 3: Tivozanib versus Sorafenib, Outcome 2: Overall survival

3.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 3.3

Comparison 3: Tivozanib versus Sorafenib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 3: Tivozanib versus Sorafenib, Outcome 3: Serious adverse events (Grade 3 or 4)

3.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 3.4

Comparison 3: Tivozanib versus Sorafenib, Outcome 4: Health‐related quality of life

Comparison 3: Tivozanib versus Sorafenib, Outcome 4: Health‐related quality of life

3.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 3.5

Comparison 3: Tivozanib versus Sorafenib, Outcome 5: Response rate

Comparison 3: Tivozanib versus Sorafenib, Outcome 5: Response rate

3.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 3.6

Comparison 3: Tivozanib versus Sorafenib, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 3: Tivozanib versus Sorafenib, Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 4. Sorafenib versus Pazopanib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

4.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 4.1

Comparison 4: Sorafenib versus Pazopanib, Outcome 1: Progression‐free survival

Comparison 4: Sorafenib versus Pazopanib, Outcome 1: Progression‐free survival

4.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 4.2

Comparison 4: Sorafenib versus Pazopanib, Outcome 2: Overall survival

Comparison 4: Sorafenib versus Pazopanib, Outcome 2: Overall survival

4.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.3

Comparison 4: Sorafenib versus Pazopanib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 4: Sorafenib versus Pazopanib, Outcome 3: Serious adverse events (Grade 3 or 4)

4.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 4.4

Comparison 4: Sorafenib versus Pazopanib, Outcome 4: Health‐related quality of life

Comparison 4: Sorafenib versus Pazopanib, Outcome 4: Health‐related quality of life

4.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.5

Comparison 4: Sorafenib versus Pazopanib, Outcome 5: Response rate

Comparison 4: Sorafenib versus Pazopanib, Outcome 5: Response rate

4.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.6

Comparison 4: Sorafenib versus Pazopanib, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 4: Sorafenib versus Pazopanib, Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 5. Sunitinib versus Everolimus

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

5.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 5.1

Comparison 5: Sunitinib versus Everolimus, Outcome 1: Progression‐free survival

Comparison 5: Sunitinib versus Everolimus, Outcome 1: Progression‐free survival

5.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 5.2

Comparison 5: Sunitinib versus Everolimus, Outcome 2: Overall survival

Comparison 5: Sunitinib versus Everolimus, Outcome 2: Overall survival

5.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 5.3

Comparison 5: Sunitinib versus Everolimus, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 5: Sunitinib versus Everolimus, Outcome 3: Serious adverse events (Grade 3 or 4)

5.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 5.4

Comparison 5: Sunitinib versus Everolimus, Outcome 4: Health‐related quality of life

Comparison 5: Sunitinib versus Everolimus, Outcome 4: Health‐related quality of life

5.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 5.5

Comparison 5: Sunitinib versus Everolimus, Outcome 5: Response rate

Comparison 5: Sunitinib versus Everolimus, Outcome 5: Response rate

5.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 5.6

Comparison 5: Sunitinib versus Everolimus, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 5: Sunitinib versus Everolimus, Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 6. Sunitinib versus Avelumab + Axitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

6.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 6.1

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 1: Progression‐free survival

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 1: Progression‐free survival

6.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 6.2

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 2: Overall survival

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 2: Overall survival

6.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 6.3

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

6.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 6.4

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 4: Response rate

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 4: Response rate

6.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 6.5

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 7. Sunitinib versus Pembrolizumab + Axitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

7.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 7.1

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 1: Progression‐free survival

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 1: Progression‐free survival

7.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 7.2

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 2: Overall survival

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 2: Overall survival

7.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 7.3

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

7.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 7.4

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 4: Response rate

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 4: Response rate

7.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 7.5

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 8. Sunitinib versus Atezolizumab + Bevacizumab

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

8.1 Progression‐free survival Show forest plot

2

1117

Hazard Ratio (IV, Random, 95% CI)

1.18 [1.02, 1.36]

Analysis 8.1

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 1: Progression‐free survival

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 1: Progression‐free survival

8.2 Overall survival Show forest plot

2

1117

Hazard Ratio (IV, Random, 95% CI)

0.99 [0.73, 1.33]

Analysis 8.2

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 2: Overall survival

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 2: Overall survival

8.3 Serious adverse events (Grade 3 or 4) Show forest plot

2

1098

Risk Ratio (M‐H, Random, 95% CI)

1.22 [1.00, 1.49]

Analysis 8.3

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

8.4 Health‐related quality of life Show forest plot

2

691

Mean Difference (IV, Random, 95% CI)

1.00 [0.68, 1.32]

Analysis 8.4

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 4: Health‐related quality of life

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 4: Health‐related quality of life

8.5 Response rate Show forest plot

2

1117

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.77, 1.07]

Analysis 8.5

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 5: Response rate

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 5: Response rate

8.6 Minor adverse events (Grade 1 or 2) Show forest plot

2

1098

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.74, 0.97]

Analysis 8.6

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 9. Sunitinib versus IMA901 + Sunitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

9.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 9.1

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 1: Progression‐free survival

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 1: Progression‐free survival

9.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 9.2

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 2: Overall survival

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 2: Overall survival

9.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 9.3

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

9.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 9.4

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 4: Response rate

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 4: Response rate

9.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 9.5

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 10. Sunitinib versus Interferon‐α (IFN‐α)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

10.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 10.1

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 1: Progression‐free survival

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 1: Progression‐free survival

10.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 10.2

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 2: Overall survival

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 2: Overall survival

10.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 10.3

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 3: Serious adverse events (Grade 3 or 4)

10.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 10.4

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 4: Health‐related quality of life

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 4: Health‐related quality of life

10.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 10.5

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 5: Response rate

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 5: Response rate

10.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 10.6

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 11. Temsirolimus versus IFN‐α

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

11.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 11.1

Comparison 11: Temsirolimus versus IFN‐α, Outcome 1: Progression‐free survival

Comparison 11: Temsirolimus versus IFN‐α, Outcome 1: Progression‐free survival

11.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 11.2

Comparison 11: Temsirolimus versus IFN‐α, Outcome 2: Overall survival

Comparison 11: Temsirolimus versus IFN‐α, Outcome 2: Overall survival

11.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 11.3

Comparison 11: Temsirolimus versus IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 11: Temsirolimus versus IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

11.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 11.4

Comparison 11: Temsirolimus versus IFN‐α, Outcome 4: Health‐related quality of life

Comparison 11: Temsirolimus versus IFN‐α, Outcome 4: Health‐related quality of life

11.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 11.5

Comparison 11: Temsirolimus versus IFN‐α, Outcome 5: Response rate

Comparison 11: Temsirolimus versus IFN‐α, Outcome 5: Response rate

11.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 11.6

Comparison 11: Temsirolimus versus IFN‐α, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 11: Temsirolimus versus IFN‐α, Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 12. Sunitinib versus Atezolizumab

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

12.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 12.1

Comparison 12: Sunitinib versus Atezolizumab, Outcome 1: Progression‐free survival

Comparison 12: Sunitinib versus Atezolizumab, Outcome 1: Progression‐free survival

12.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 12.2

Comparison 12: Sunitinib versus Atezolizumab, Outcome 2: Overall survival

Comparison 12: Sunitinib versus Atezolizumab, Outcome 2: Overall survival

12.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 12.3

Comparison 12: Sunitinib versus Atezolizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 12: Sunitinib versus Atezolizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

12.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 12.4

Comparison 12: Sunitinib versus Atezolizumab, Outcome 4: Health‐related quality of life

Comparison 12: Sunitinib versus Atezolizumab, Outcome 4: Health‐related quality of life

12.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 12.5

Comparison 12: Sunitinib versus Atezolizumab, Outcome 5: Response rate

Comparison 12: Sunitinib versus Atezolizumab, Outcome 5: Response rate

12.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 12.6

Comparison 12: Sunitinib versus Atezolizumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 12: Sunitinib versus Atezolizumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 13. Bevacizumab + IFN versus IFN (+ placebo)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

13.1 Progression‐free survival Show forest plot

2

1381

Hazard Ratio (IV, Random, 95% CI)

0.68 [0.60, 0.77]

Analysis 13.1

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 1: Progression‐free survival

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 1: Progression‐free survival

13.2 Overall survival Show forest plot

2

1381

Hazard Ratio (IV, Random, 95% CI)

0.88 [0.79, 0.99]

Analysis 13.2

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 2: Overall survival

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 2: Overall survival

13.3 Serious adverse events (Grade 3 or 4) Show forest plot

2

1356

Risk Ratio (M‐H, Random, 95% CI)

1.31 [1.20, 1.42]

Analysis 13.3

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 3: Serious adverse events (Grade 3 or 4)

13.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 13.4

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 4: Response rate

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 4: Response rate

13.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 13.5

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 5: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 14. Temsirolimus + IFN‐α versus IFN‐α

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

14.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 14.1

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 1: Progression‐free survival

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 1: Progression‐free survival

14.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 14.2

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 2: Overall survival

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 2: Overall survival

14.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 14.3

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

14.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 14.4

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 4: Health‐related quality of life

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 4: Health‐related quality of life

14.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 14.5

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 5: Response rate

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 5: Response rate

14.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 14.6

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 15. Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

15.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 15.1

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 1: Progression‐free survival

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 1: Progression‐free survival

15.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 15.2

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 2: Overall survival

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 2: Overall survival

15.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 15.3

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

15.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 15.4

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 4: Response rate

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 4: Response rate

15.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 15.5

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 5: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 16. Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

16.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 16.1

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 1: Progression‐free survival

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 1: Progression‐free survival

16.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 16.2

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 2: Overall survival

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 2: Overall survival

16.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 16.3

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

16.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 16.4

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 4: Response rate

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 4: Response rate

16.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 16.5

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 5: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 17. Sunitinib versus Nivolumab + Ipilimumab

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

17.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 17.1

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 1: Progression‐free survival

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 1: Progression‐free survival

17.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 17.2

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 2: Overall survival

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 2: Overall survival

17.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 17.3

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 3: Serious adverse events (Grade 3 or 4)

17.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 17.4

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 4: Health‐related quality of life

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 4: Health‐related quality of life

17.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 17.5

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 5: Response rate

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 5: Response rate

17.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 17.6

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Open in table viewer
Comparison 18. Pazopanib versus Placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

18.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 18.1

Comparison 18: Pazopanib versus Placebo, Outcome 1: Progression‐free survival

Comparison 18: Pazopanib versus Placebo, Outcome 1: Progression‐free survival

18.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 18.2

Comparison 18: Pazopanib versus Placebo, Outcome 2: Overall survival

Comparison 18: Pazopanib versus Placebo, Outcome 2: Overall survival

18.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 18.3

Comparison 18: Pazopanib versus Placebo, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 18: Pazopanib versus Placebo, Outcome 3: Serious adverse events (Grade 3 or 4)

18.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 18.4

Comparison 18: Pazopanib versus Placebo, Outcome 4: Health‐related quality of life

Comparison 18: Pazopanib versus Placebo, Outcome 4: Health‐related quality of life

18.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 18.5

Comparison 18: Pazopanib versus Placebo, Outcome 5: Response rate

Comparison 18: Pazopanib versus Placebo, Outcome 5: Response rate

18.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 18.6

Comparison 18: Pazopanib versus Placebo, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 18: Pazopanib versus Placebo, Outcome 6: Minor adverse events (Grade 1 or 2)

Study flow diagram.

Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study. Categories: green point (+) = low risk of bias; yellow point (?) = unclear risk of bias; red point (‐) = high risk of bias.

Figuras y tablas -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study. Categories: green point (+) = low risk of bias; yellow point (?) = unclear risk of bias; red point (‐) = high risk of bias.

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

Figuras y tablas -
Figure 3

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

Comparison 1: Sorafenib versus Sunitinib, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 1.1

Comparison 1: Sorafenib versus Sunitinib, Outcome 1: Progression‐free survival

Comparison 1: Sorafenib versus Sunitinib, Outcome 2: Overall survival

Figuras y tablas -
Analysis 1.2

Comparison 1: Sorafenib versus Sunitinib, Outcome 2: Overall survival

Comparison 1: Sorafenib versus Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 1.3

Comparison 1: Sorafenib versus Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 1: Sorafenib versus Sunitinib, Outcome 4: Response rate

Figuras y tablas -
Analysis 1.4

Comparison 1: Sorafenib versus Sunitinib, Outcome 4: Response rate

Comparison 1: Sorafenib versus Sunitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 1.5

Comparison 1: Sorafenib versus Sunitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 2: Pazopanib versus Sunitinib, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 2.1

Comparison 2: Pazopanib versus Sunitinib, Outcome 1: Progression‐free survival

Comparison 2: Pazopanib versus Sunitinib, Outcome 2: Overall survival

Figuras y tablas -
Analysis 2.2

Comparison 2: Pazopanib versus Sunitinib, Outcome 2: Overall survival

Comparison 2: Pazopanib versus Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 2.3

Comparison 2: Pazopanib versus Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 2: Pazopanib versus Sunitinib, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 2.4

Comparison 2: Pazopanib versus Sunitinib, Outcome 4: Health‐related quality of life

Comparison 2: Pazopanib versus Sunitinib, Outcome 5: Response rate

Figuras y tablas -
Analysis 2.5

Comparison 2: Pazopanib versus Sunitinib, Outcome 5: Response rate

Comparison 2: Pazopanib versus Sunitinib, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 2.6

Comparison 2: Pazopanib versus Sunitinib, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 3: Tivozanib versus Sorafenib, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 3.1

Comparison 3: Tivozanib versus Sorafenib, Outcome 1: Progression‐free survival

Comparison 3: Tivozanib versus Sorafenib, Outcome 2: Overall survival

Figuras y tablas -
Analysis 3.2

Comparison 3: Tivozanib versus Sorafenib, Outcome 2: Overall survival

Comparison 3: Tivozanib versus Sorafenib, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 3.3

Comparison 3: Tivozanib versus Sorafenib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 3: Tivozanib versus Sorafenib, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 3.4

Comparison 3: Tivozanib versus Sorafenib, Outcome 4: Health‐related quality of life

Comparison 3: Tivozanib versus Sorafenib, Outcome 5: Response rate

Figuras y tablas -
Analysis 3.5

Comparison 3: Tivozanib versus Sorafenib, Outcome 5: Response rate

Comparison 3: Tivozanib versus Sorafenib, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 3.6

Comparison 3: Tivozanib versus Sorafenib, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 4: Sorafenib versus Pazopanib, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 4.1

Comparison 4: Sorafenib versus Pazopanib, Outcome 1: Progression‐free survival

Comparison 4: Sorafenib versus Pazopanib, Outcome 2: Overall survival

Figuras y tablas -
Analysis 4.2

Comparison 4: Sorafenib versus Pazopanib, Outcome 2: Overall survival

Comparison 4: Sorafenib versus Pazopanib, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 4.3

Comparison 4: Sorafenib versus Pazopanib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 4: Sorafenib versus Pazopanib, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 4.4

Comparison 4: Sorafenib versus Pazopanib, Outcome 4: Health‐related quality of life

Comparison 4: Sorafenib versus Pazopanib, Outcome 5: Response rate

Figuras y tablas -
Analysis 4.5

Comparison 4: Sorafenib versus Pazopanib, Outcome 5: Response rate

Comparison 4: Sorafenib versus Pazopanib, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 4.6

Comparison 4: Sorafenib versus Pazopanib, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 5: Sunitinib versus Everolimus, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 5.1

Comparison 5: Sunitinib versus Everolimus, Outcome 1: Progression‐free survival

Comparison 5: Sunitinib versus Everolimus, Outcome 2: Overall survival

Figuras y tablas -
Analysis 5.2

Comparison 5: Sunitinib versus Everolimus, Outcome 2: Overall survival

Comparison 5: Sunitinib versus Everolimus, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 5.3

Comparison 5: Sunitinib versus Everolimus, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 5: Sunitinib versus Everolimus, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 5.4

Comparison 5: Sunitinib versus Everolimus, Outcome 4: Health‐related quality of life

Comparison 5: Sunitinib versus Everolimus, Outcome 5: Response rate

Figuras y tablas -
Analysis 5.5

Comparison 5: Sunitinib versus Everolimus, Outcome 5: Response rate

Comparison 5: Sunitinib versus Everolimus, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 5.6

Comparison 5: Sunitinib versus Everolimus, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 6.1

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 1: Progression‐free survival

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 2: Overall survival

Figuras y tablas -
Analysis 6.2

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 2: Overall survival

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 6.3

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 4: Response rate

Figuras y tablas -
Analysis 6.4

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 4: Response rate

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 6.5

Comparison 6: Sunitinib versus Avelumab + Axitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 7.1

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 1: Progression‐free survival

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 2: Overall survival

Figuras y tablas -
Analysis 7.2

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 2: Overall survival

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 7.3

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 4: Response rate

Figuras y tablas -
Analysis 7.4

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 4: Response rate

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 7.5

Comparison 7: Sunitinib versus Pembrolizumab + Axitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 8.1

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 1: Progression‐free survival

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 2: Overall survival

Figuras y tablas -
Analysis 8.2

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 2: Overall survival

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 8.3

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 8.4

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 4: Health‐related quality of life

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 5: Response rate

Figuras y tablas -
Analysis 8.5

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 5: Response rate

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 8.6

Comparison 8: Sunitinib versus Atezolizumab + Bevacizumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 9.1

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 1: Progression‐free survival

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 2: Overall survival

Figuras y tablas -
Analysis 9.2

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 2: Overall survival

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 9.3

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 4: Response rate

Figuras y tablas -
Analysis 9.4

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 4: Response rate

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 9.5

Comparison 9: Sunitinib versus IMA901 + Sunitinib, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 10.1

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 1: Progression‐free survival

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 2: Overall survival

Figuras y tablas -
Analysis 10.2

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 2: Overall survival

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 10.3

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 10.4

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 4: Health‐related quality of life

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 5: Response rate

Figuras y tablas -
Analysis 10.5

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 5: Response rate

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 10.6

Comparison 10: Sunitinib versus Interferon‐α (IFN‐α), Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 11: Temsirolimus versus IFN‐α, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 11.1

Comparison 11: Temsirolimus versus IFN‐α, Outcome 1: Progression‐free survival

Comparison 11: Temsirolimus versus IFN‐α, Outcome 2: Overall survival

Figuras y tablas -
Analysis 11.2

Comparison 11: Temsirolimus versus IFN‐α, Outcome 2: Overall survival

Comparison 11: Temsirolimus versus IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 11.3

Comparison 11: Temsirolimus versus IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 11: Temsirolimus versus IFN‐α, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 11.4

Comparison 11: Temsirolimus versus IFN‐α, Outcome 4: Health‐related quality of life

Comparison 11: Temsirolimus versus IFN‐α, Outcome 5: Response rate

Figuras y tablas -
Analysis 11.5

Comparison 11: Temsirolimus versus IFN‐α, Outcome 5: Response rate

Comparison 11: Temsirolimus versus IFN‐α, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 11.6

Comparison 11: Temsirolimus versus IFN‐α, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 12: Sunitinib versus Atezolizumab, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 12.1

Comparison 12: Sunitinib versus Atezolizumab, Outcome 1: Progression‐free survival

Comparison 12: Sunitinib versus Atezolizumab, Outcome 2: Overall survival

Figuras y tablas -
Analysis 12.2

Comparison 12: Sunitinib versus Atezolizumab, Outcome 2: Overall survival

Comparison 12: Sunitinib versus Atezolizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 12.3

Comparison 12: Sunitinib versus Atezolizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 12: Sunitinib versus Atezolizumab, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 12.4

Comparison 12: Sunitinib versus Atezolizumab, Outcome 4: Health‐related quality of life

Comparison 12: Sunitinib versus Atezolizumab, Outcome 5: Response rate

Figuras y tablas -
Analysis 12.5

Comparison 12: Sunitinib versus Atezolizumab, Outcome 5: Response rate

Comparison 12: Sunitinib versus Atezolizumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 12.6

Comparison 12: Sunitinib versus Atezolizumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 13.1

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 1: Progression‐free survival

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 2: Overall survival

Figuras y tablas -
Analysis 13.2

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 2: Overall survival

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 13.3

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 4: Response rate

Figuras y tablas -
Analysis 13.4

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 4: Response rate

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 5: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 13.5

Comparison 13: Bevacizumab + IFN versus IFN (+ placebo), Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 14.1

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 1: Progression‐free survival

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 2: Overall survival

Figuras y tablas -
Analysis 14.2

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 2: Overall survival

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 14.3

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 14.4

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 4: Health‐related quality of life

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 5: Response rate

Figuras y tablas -
Analysis 14.5

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 5: Response rate

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 14.6

Comparison 14: Temsirolimus + IFN‐α versus IFN‐α, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 15.1

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 1: Progression‐free survival

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 2: Overall survival

Figuras y tablas -
Analysis 15.2

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 2: Overall survival

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 15.3

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 4: Response rate

Figuras y tablas -
Analysis 15.4

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 4: Response rate

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 5: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 15.5

Comparison 15: Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 16.1

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 1: Progression‐free survival

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 2: Overall survival

Figuras y tablas -
Analysis 16.2

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 2: Overall survival

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 16.3

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 4: Response rate

Figuras y tablas -
Analysis 16.4

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 4: Response rate

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 5: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 16.5

Comparison 16: Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab, Outcome 5: Minor adverse events (Grade 1 or 2)

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 17.1

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 1: Progression‐free survival

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 2: Overall survival

Figuras y tablas -
Analysis 17.2

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 2: Overall survival

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 17.3

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 17.4

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 4: Health‐related quality of life

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 5: Response rate

Figuras y tablas -
Analysis 17.5

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 5: Response rate

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 17.6

Comparison 17: Sunitinib versus Nivolumab + Ipilimumab, Outcome 6: Minor adverse events (Grade 1 or 2)

Comparison 18: Pazopanib versus Placebo, Outcome 1: Progression‐free survival

Figuras y tablas -
Analysis 18.1

Comparison 18: Pazopanib versus Placebo, Outcome 1: Progression‐free survival

Comparison 18: Pazopanib versus Placebo, Outcome 2: Overall survival

Figuras y tablas -
Analysis 18.2

Comparison 18: Pazopanib versus Placebo, Outcome 2: Overall survival

Comparison 18: Pazopanib versus Placebo, Outcome 3: Serious adverse events (Grade 3 or 4)

Figuras y tablas -
Analysis 18.3

Comparison 18: Pazopanib versus Placebo, Outcome 3: Serious adverse events (Grade 3 or 4)

Comparison 18: Pazopanib versus Placebo, Outcome 4: Health‐related quality of life

Figuras y tablas -
Analysis 18.4

Comparison 18: Pazopanib versus Placebo, Outcome 4: Health‐related quality of life

Comparison 18: Pazopanib versus Placebo, Outcome 5: Response rate

Figuras y tablas -
Analysis 18.5

Comparison 18: Pazopanib versus Placebo, Outcome 5: Response rate

Comparison 18: Pazopanib versus Placebo, Outcome 6: Minor adverse events (Grade 1 or 2)

Figuras y tablas -
Analysis 18.6

Comparison 18: Pazopanib versus Placebo, Outcome 6: Minor adverse events (Grade 1 or 2)

Summary of findings 1. Sorafenib compared to sunitinib (targeted agent versus targeted agent)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (any cell type)
Setting: Germany and the Netherlands/muticentre/likely outpatient
Intervention: Sorafenib
Comparison: Sunitinib

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Sunitinib

Risk difference with Sorafenib

Progression‐free survival

(absolute effect size estimates based on survival rate at 10 months)
follow‐up: mean 10.3 months

365
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

HR 1.19
(0.92 to 1.53)

Study population

340 per 1000

63 fewer per 1000
(148 fewer to 31 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)
follow‐up: mean 10.3 months

365
(1 RCT)

⊕⊝⊝⊝
VERY LOW 3 4

HR 0.99
(0.74 to 1.33)

Study population

550 per 1000

3 more per 1000
(98 fewer to 92 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v3.0

353
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 4

RR 0.99
(0.85 to 1.14)

Study population

670 per 1000

7 fewer per 1000
(101 fewer to 94 more)

Health‐related quality of life5

not reported

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for study limitations; high risk of performance and detection bias and unclear risk of other bias

2 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included harm and no harm)

3 Downgraded by 1 level for study limitations; unclear risk of other bias

4 Downgraded by 2 levels for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference: wide confidence interval (included both benefit and harm)

5 Health‐related quality of life: no available data

Figuras y tablas -
Summary of findings 1. Sorafenib compared to sunitinib (targeted agent versus targeted agent)
Summary of findings 2. Pazopanib compared to sunitinib (targeted agent versus targeted agent)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Pazopanib
Comparison: Sunitinib

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Sunitinib

Risk difference with Pazopanib

Progression‐free survival

(absolute effect size estimates based on survival rate at 12 months)

follow‐up: not reported

1110
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

HR 1.05
(0.90 to 1.23)

Study population

420 per 1000

18 fewer per 1000
(76 fewer to 38 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)

follow‐up: not reported

1110
(1 RCT)

⊕⊕⊝⊝
LOW 3 4

HR 0.92
(0.80 to 1.06)

Study population

550 per 1000

27 more per 1000
(19 fewer to 70 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v3.0

1102
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

RR 1.01
(0.94 to 1.09)

Study population

734 per 1000

7 more per 1000
(44 fewer to 66 more)

Health‐related quality of life (mean change value)
assessed with: FACIT‐F (higher scores indicating less fatigue)
Scale from: 0 to 52

follow‐up: after 4 cycle

467
(1 RCT)

⊕⊕⊝⊝
LOW 5 6

The mean health‐related quality of life (mean change value) was ‐6.5

MD 3.6 higher
(1.76 higher to 5.44 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;FACIT‐F: Functional Assessment of Chronic Illness Therapy–Fatigue scale; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for study limitations; high risk of performance and detection bias and unclear risk of other bias

2 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included harm and no harm)

3 Downgraded by 1 level for study limitations; unclear risk of other bias

4 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included no benefit and benefit)

5 Downgraded by 1 level for study limitations; high risk of performance, detection and attrition bias and unclear risk of other bias

6 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (3 points, included benefit and little benefit)

Figuras y tablas -
Summary of findings 2. Pazopanib compared to sunitinib (targeted agent versus targeted agent)
Summary of findings 3. Tivozanib compared to sorafenib (targeted agent versus targeted agent)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Tivozanib
Comparison: Sorafenib

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Sorafenib

Risk difference with Tivozanib

Progression‐free survival

(absolute effect size estimates based on survival rate at 12 months)

follow‐up: not reported

517
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

HR 0.79
(0.64 to 0.99)

Study population

360 per 1000

86 more per 1000
(4 more to 160 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)

follow‐up: not reported

517
(1 RCT)

⊕⊕⊝⊝
LOW 3 4

HR 1.25
(0.95 to 1.64)

Study population

620 per 1000

70 fewer per 1000
(163 fewer to 15 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v3.0

516
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

RR 0.85
(0.74 to 0.97)

Study population

689 per 1000

103 fewer per 1000
(179 fewer to 21 fewer)

Health‐related quality of life
assessed with: EQ‐5D Health State Index
Scale from: ‐0.59 (worst health state) to 1 (best health state)
follow‐up: 12 months

506
(1 RCT)

⊕⊕⊝⊝
LOW 2 5

The mean health‐related quality of life was ‐0.06

MD 0.01 higher
(0.05 lower to 0.07 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;EQ‐5D: EuroQol‐5D; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (included benefit and little benefit)

2 Downgraded by 1 level for study limitations; high risk of performance, detection and other bias

3 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included harm and no harm)

4 Downgraded by 1 level for study limitations; high risk of other bias

5 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (0.06 points, included benefit and no benefit)

Figuras y tablas -
Summary of findings 3. Tivozanib compared to sorafenib (targeted agent versus targeted agent)
Summary of findings 4. Sorafenib compared to pazopanib (targeted agent versus targeted agent)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (any cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Sorafenib
Comparison: Pazopanib

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Pazopanib

Risk difference with Sorafenib

Progression‐free survival

(absolute effect size estimates based on survival rate at 12 months)

follow‐up: not reported

377
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

HR 1.92
(1.74 to 2.11)

Study population

380 per 1000

224 fewer per 1000
(250 fewer to 194 fewer)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)

follow‐up: not reported

377
(1 RCT)

⊕⊕⊝⊝
LOW 2 3

HR 1.22
(0.91 to 1.64)

Study population

520 per 1000

70 fewer per 1000
(178 fewer to 32 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v4.03

366
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 4

RR 0.92
(0.78 to 1.09)

Study population

639 per 1000

51 fewer per 1000
(141 fewer to 58 more)

Health‐related quality of life
(mean change value)
assessed with: FACIT‐F (higher scores indicating less fatigue)
Scale from: 0 to 52

follow‐up: not reported

267
(1 RCT)

⊕⊕⊝⊝
LOW 5 6

The mean health‐related quality of life was ‐9.9

MD 3.1 higher
(1.82 lower to 8.02 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;FACIT‐F: Functional Assessment of Chronic Illness Therapy–Fatigue scale; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for study limitations; unclear risk of selection, detection, and reporting bias and high risk of performance bias

2 Downgraded by 1 level for study limitations; unclear risk of selection, and reporting bias

3 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included harm and no harm)

4 Downgraded by 2 levels for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference: wide confidence interval (included both benefit and harm)

5 Downgraded by 1 level for study limitations; unclear risk of selection, detection, and reporting bias and high risk of performance and attrition bias

6 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (3 points, included benefit and no benefit)

Figuras y tablas -
Summary of findings 4. Sorafenib compared to pazopanib (targeted agent versus targeted agent)
Summary of findings 5. Sunitinib compared to everolimus (targeted agent versus targeted agent)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (any cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Sunitinib
Comparison: Everolimus

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Everolimus

Risk difference with Sunitinib

Progression‐free survival

(absolute effect size estimates based on survival rate at 12 months)

follow‐up: not reported

471
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

HR 0.71
(0.59 to 0.87)

Study population

300 per 1000

125 more per 1000
(51 more to 191 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)

follow‐up: not reported

471
(1 RCT)

⊕⊕⊝⊝
LOW 2 3

HR 0.90
(0.72 to 1.11)

Study population

470 per 1000

37 more per 1000
(37 fewer to 111 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v3.0

469
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

RR 1.34
(1.14 to 1.59)

Study population

471 per 1000

160 more per 1000
(66 more to 278 more)

Health‐related quality of life
assessed with: EORTC QLQ‐C30 (Global health status scale: high score represent better functioning)
Scale from: 0 to 100
follow‐up: 16 weeks

288
(1 RCT)

⊕⊕⊝⊝
LOW 1 4

The mean health‐related quality of life was 65.5

MD 5 lower
(10.4 lower to 0.4 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;EORTC QLQ‐C30: European Organization for Research and Treatment of Cancer Quality of Life Questionnaire; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for study limitations; high risk of performance, detection and other bias

2 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included benefit and no benefit)

3 Downgraded by 1 level for study limitations; high risk of other bias

4 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (10 points, included harm and no harm)

Figuras y tablas -
Summary of findings 5. Sunitinib compared to everolimus (targeted agent versus targeted agent)
Summary of findings 6. Sunitinib compared to avelumab + axitinib (targeted agent versus immunotherapy + targeted agent)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Sunitinib
Comparison: Avelumab + Axitinib

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Avelumab + Axitinib

Risk difference with Sunitinib

Progression‐free survival
(absolute effect size estimates based on survival rate at 12 months)

follow‐up: median 10.8 months

886
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

HR 1.45
(1.17 to 1.80)

Study population

550 per 1000

130 fewer per 1000
(209 fewer to 53 fewer)

Overall survival

(absolute effect size estimates based on survival rate at 12 months)
follow‐up: median 12.0 months

886
(1 RCT)

⊕⊕⊝⊝
LOW 2 3

HR 1.28
(0.92 to 1.79)

Study population

890 per 1000

29 fewer per 1000
(78 fewer to 8 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v4.03

873
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

RR 1.01
(0.93 to 1.10)

Study population

705 per 1000

7 more per 1000
(49 fewer to 71 more)

Health‐related quality of life4

Not reported

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for study limitations: high risk of performance bias and unclear risk of reporting bias

2 Downgraded by 1 level for imprecision: confidence interval crossed the assumed threshold of a clinically important difference (included no benefit and harm)

3 Downgraded by 1 level for study limitations: unclear risk of reporting bias

4 Health‐related quality of life: no available data

Figuras y tablas -
Summary of findings 6. Sunitinib compared to avelumab + axitinib (targeted agent versus immunotherapy + targeted agent)
Summary of findings 7. Sunitinib compared to pembrolizumab + axitinib (targeted agent versus immunotherapy + targeted agent)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Sunitinib
Comparison: Pembrolizumab + Axitinib

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Pembrolizumab + Axitinib

Risk difference with Sunitinib

Progression‐free survival
(absolute effect size estimates based on survival rate at 12 months)

follow‐up: median 12.8 months

861
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

HR 1.45
(1.19 to 1.76)

Study population

590 per 1000

125 fewer per 1000
(195 fewer to 56 fewer)

Overall survival

(absolute effect size estimates based on survival rate at 12 months)
follow‐up: median 12.8 months

861
(1 RCT)

⊕⊕⊕⊝
MODERATE 2

HR 1.90
(1.36 to 2.65)

Study population

880 per 1000

96 fewer per 1000
(167 fewer to 40 fewer)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v4.0

854
(1 RCT)

⊕⊕⊝⊝
LOW 1 3

RR 0.90
(0.81 to 1.02)

Study population

604 per 1000

60 fewer per 1000
(115 fewer to 12 more)

Health‐related quality of life4

Not reported

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for study limitations; high risk of performance bias

2 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (included harm and little harm)

3 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included benefit and no benefit)

4 Health‐related quality of life: no available data

Figuras y tablas -
Summary of findings 7. Sunitinib compared to pembrolizumab + axitinib (targeted agent versus immunotherapy + targeted agent)
Summary of findings 8. Sunitinib compared to atezolizumab + bevacizumab (targeted agent versus immunotherapy + targeted agent)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Sunitinib
Comparison: Atezolizumab + Bevacizumab

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Atezolizumab + Bevacizumab

Risk difference with Sunitinib

Progression‐free survival
(absolute effect size estimates based on survival rate at 12 months)

follow‐up: range 15 months to 20.7 months

1117
(2 RCTs)

⊕⊕⊝⊝
LOW 1 2

HR 1.18
(1.02 to 1.36)

Study population

480 per 1000

59 fewer per 1000
(111 fewer to 7 fewer)

Overall survival
(absolute effect size estimates based on survival rate at 24 months)

follow‐up: range 20.7 months to 24 months

1117
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 2 3

HR 0.99
(0.73 to 1.33)

Study population

630 per 1000

3 more per 1000
(89 fewer to 84 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v4.0

1098
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 2 4 5

RR 1.22
(1.00 to 1.49)

Study population

446 per 1000

98 more per 1000
(0 fewer to 218 more)

Health‐related quality of life
assessed with: MDASI (high score indicates worse QoL)
Scale from: 0 to 10
follow‐up: 12 weeks

691
(2 RCTs)

⊕⊕⊝⊝
LOW 1 2

The mean health‐related quality of life ranged from 0.56 to 1.57

MD 1 higher
(0.68 higher to 1.32 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;HR: Hazard ratio; MDASI: MD Anderson Symptom Inventory; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (included harm and little harm)

2 Downgraded by 1 level for study limitations; high and unclear risk of 1 or more domains.

3 Downgraded by 2 levels for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference: wide confidence interval (included both benefit and harm)

4 Downgraded by 1 level for inconsistency; moderate to substantial heterogeneity: unexplained differences between study results

5 Downgraded by 1 level for imprecision; confidence interval reached the line of no difference and crossed the assumed threshold of a clinically important difference (included harm and no harm)

Figuras y tablas -
Summary of findings 8. Sunitinib compared to atezolizumab + bevacizumab (targeted agent versus immunotherapy + targeted agent)
Summary of findings 9. Sunitinib compared to IMA901 + sunitinib (targeted agent versus tumour vaccine + targeted agent)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Sunitinib
Comparison: IMA901 + Sunitinib

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with IMA901 + Sunitinib

Risk difference with Sunitinib

Progression‐free survival

(absolute effect size estimates based on survival rate at 12 months)
follow‐up: median 33.27 months

339
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

HR 0.95
(0.70 to 1.30)

Study population

590 per 1000

16 more per 1000
(86 fewer to 101 more)

Overall survival

(absolute effect size estimates based on survival rate at 12 months)
follow‐up: median 33.27 months

339
(1 RCT)

⊕⊕⊝⊝
LOW 3 4

HR 0.75
(0.54 to 1.04)

Study population

800 per 1000

46 more per 1000
(7 fewer to 86 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v4.0

334
(1 RCT)

⊕⊕⊝⊝
LOW 2 5

RR 0.74
(0.59 to 0.95)

Study population

550 per 1000

143 fewer per 1000
(225 fewer to 27 fewer)

Health‐related quality of life6

Not reported

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 2 levels for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference: wide confidence interval (included both benefit and harm)

2 Downgraded by 1 level for study limitations; high risk of performance and other bias

3 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included benefit and no benefit)

4 Downgraded by 1 level for study limitations: high risk of other bias

5 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (included benefit and little benefit)

6 Health‐related quality of life: no available data

Figuras y tablas -
Summary of findings 9. Sunitinib compared to IMA901 + sunitinib (targeted agent versus tumour vaccine + targeted agent)
Summary of findings 10. Sunitinib compared to interferon‐α (IFN‐α) (targeted agent versus classic immunotherapy)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Sunitinib
Comparison: Interferon‐α (IFN‐α)

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Interferon‐α (IFN‐α)

Risk difference with Sunitinib

Progression‐free survival

(absolute effect size estimates based on survival rate at 6 months)
follow‐up: median 31 months

750
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

HR 0.54
(0.45 to 0.64)

Study population

400 per 1000

210 more per 1000
(156 more to 262 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)
follow‐up: median 31 months

750
(1 RCT)

⊕⊕⊝⊝
LOW 2 3

HR 0.82
(0.67 to 1.00)

Study population

480 per 1000

68 more per 1000
(0 fewer to 132 more)

Serious adverse events (Grade 3 or 4)
assessed as: CTCAE v3.0

735
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

RR 1.75
(1.43 to 2.16)

Study population

258 per 1000

194 more per 1000
(111 more to 300 more)

Health‐related quality of life
assessed with: EQ‐5D Health State Index
Scale from: ‐0.59 (worst health state) to 1 (best health state)

follow‐up: after 2 cycle

544
(1 RCT)

⊕⊕⊕⊝
MODERATE 4

The mean health‐related quality of life was 0.74

MD 0.01 lower
(0.05 lower to 0.03 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval;EQ‐5D: EuroQol‐5D; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for study limitations; unclear risk of selection bias and high risk of performance and other bias

2 Downgraded by 1 level for study limitations; unclear risk of selection bias and high risk of other bias

3 Downgraded by 1 level for imprecision; confidence interval reached the line of no difference and crossed the assumed threshold of a clinically important difference (included benefit and no benefit)

4 Downgraded by 1 level for study limitations; unclear risk of selection and attrition bias and high risk of performance and other bias

Figuras y tablas -
Summary of findings 10. Sunitinib compared to interferon‐α (IFN‐α) (targeted agent versus classic immunotherapy)
Summary of findings 11. Temsirolimus compared to IFN‐α (targeted agent versus classic immunotherapy)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (any cell type [80% clear cell])
Setting: Multinational muticentre/likely outpatient
Intervention: Temsirolimus
Comparison: IFN‐α

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with IFN‐α

Risk difference with Temsirolimus

Progression‐free survival (absolute effect size estimates based on survival rate at 12 months)

follow‐up: up to 80 months

416
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

HR 0.74
(0.60 to 0.91)

Study population

100 per 1000

82 more per 1000
(23 more to 151 more)

Overall survival survival (absolute effect size estimates based on survival rate at 12 months )

follow‐up: up to 80 months

416
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

HR 0.78
(0.63 to 0.97)

Study population

300 per 1000

91 more per 1000
(11 more to 168 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE version: not reported

408
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

RR 0.86
(0.76 to 0.97)

Study population

780 per 1000

109 fewer per 1000
(187 fewer to 23 fewer)

Health‐related quality of life
assessed with: EQ‐5D Health State Index
Scale from: ‐0.59 (worst health state) to 1 (best health state)

follow‐up: not reported

401
(1 RCT)

⊕⊕⊝⊝
LOW 3 4

The mean health‐related quality of life was 0.66

MD 0.03 higher
(0.01 lower to 0.07 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;EQ‐5D: EuroQol‐5D; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (included benefit and no benefit)

2 Downgraded by 1 level for study limitations; high risk of performance and detection bias

3 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included benefit and no benefit)

4 Downgraded by 1 level for study limitations; high risk of performance, detection and attrition bias

Figuras y tablas -
Summary of findings 11. Temsirolimus compared to IFN‐α (targeted agent versus classic immunotherapy)
Summary of findings 12. Sunitinib compared to atezolizumab (targeted therapy versus immunotherapy)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Sunitinib
Comparison: Atezolizumab

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Atezolizumab

Risk difference with Sunitinib

Progression‐free survival

(absolute effect size estimates based on survival rate at 12 months)
follow‐up: median 20.7 months

204
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

HR 0.84
(0.58 to 1.22)

Study population

420 per 1000

63 more per 1000
(73 fewer to 185 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)
follow‐up: median 20.7 months

204
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 3

HR 0.94
(0.58 to 1.54)

Moderate

630 per 1000 6

18 more per 1000
(139 fewer to 135 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v4.0

203
(1 RCT)

⊕⊕⊕⊝
MODERATE 2

RR 1.73
(1.32 to 2.27)

Study population

398 per 1000

291 more per 1000
(127 more to 506 more)

Health‐related quality of life
assessed with: MDASI (high score indicates worse QoL)
Scale from: 0 to 10
follow‐up: 12 weeks

157
(1 RCT)

⊕⊕⊝⊝
LOW 4 5

The mean health‐related quality of life was 1.04

MD 1.46 higher
(0.8 higher to 2.12 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;HR: Hazard ratio; MDASI: MD Anderson Symptom Inventory; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 2 levels for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference: wide confidence interval (included both benefit and harm)

2 Downgraded by 1 level for study limitations; high risk of selection, performance and detection bias and unclear risk of other bias

3 Downgraded by 1 level for study limitations; high risk of selection and unclear risk of other bias

4 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (1 point, included harm and little harm)

5 Downgraded by 1 level for study limitations; high risk of selection, performance and detection bias and unclear risk of other bias

6 Baseline risk for overall survival in the atezolizumab group was assumed to be 63% (moderate risk) at 24 months as reported in Rini 2019b

Figuras y tablas -
Summary of findings 12. Sunitinib compared to atezolizumab (targeted therapy versus immunotherapy)
Summary of findings 13. Bevacizumab + IFN compared to IFN (+ placebo) (targeted agent + classic immunotherapy versus classic immunotherapy)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Bevacizumab + IFN
Comparison: IFN (+ placebo)

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with IFN (+ placebo)

Risk difference with Bevacizumab + IFN

Progression‐free survival

(absolute effect size estimates based on survival rate at 12 months)

follow‐up: intervention: 13.3 months

comparator: 12.8 months

1381
(2 RCTs)

⊕⊕⊕⊝
MODERATE 1

HR 0.68
(0.60 to 0.77)

Study population

200 per 1000

135 more per 1000
(90 more to 181 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)

follow‐up: intervention: 23 months

comparator: 21 months

1381
(2 RCTs)

⊕⊕⊝⊝
LOW 1 2

HR 0.88
(0.79 to 0.99)

Study population

500 per 1000

43 more per 1000
(3 more to 78 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v3.0

1356
(2 RCTs)

⊕⊕⊕⊝
MODERATE 1

RR 1.31
(1.20 to 1.42)

Study population

536 per 1000

166 more per 1000
(107 more to 225 more)

Health‐related quality of life3

Not reported

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for study limitations; high and unclear risk of 1 or more domains

2 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (included benefit and little benefit)

3 Health‐related quality of life: no available data

Figuras y tablas -
Summary of findings 13. Bevacizumab + IFN compared to IFN (+ placebo) (targeted agent + classic immunotherapy versus classic immunotherapy)
Summary of findings 14. Temsirolimus + IFN‐α compared to IFN‐α (targeted agent + classic immunotherapy versus classic immunotherapy)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (any cell type [80% clear cell])
Setting: Multinational muticentre/likely outpatient
Intervention: Temsirolimus + IFN‐α
Comparison: IFN‐α

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with IFN‐α

Risk difference with Temsirolimus + IFN‐α

Progression‐free survival (absolute effect size estimates based on survival rate at 12 months) follow‐up: up to 80 months

417
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

HR 0.76
(0.62 to 0.93)

Study population

100 per 1000

74 more per 1000
(17 more to 140 more)

Overall survival

(absolute effect size estimates based on survival rate at 12 months) follow‐up: up to 80 months

417
(1 RCT)

⊕⊕⊝⊝
LOW 3

HR 0.93
(0.75 to 1.15)

Study population

300 per 1000

26 more per 1000
(50 fewer to 105 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE version: not reported

408
(1 RCT)

⊕⊕⊝⊝
LOW 2 4

RR 1.12
(1.02 to 1.22)

Study population

780 per 1000

94 more per 1000
(16 more to 172 more)

Health‐related quality of life
assessed with: EQ‐5D Health State Index
Scale from: ‐0.59 (worst health state) to 1 (best health state)

follow‐up: not reported

394
(1 RCT)

⊕⊕⊝⊝
LOW 5 6

The mean health‐related quality of life was 0.66

MD 0.03 higher
(0.01 lower to 0.07 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;EQ‐5D: EuroQol‐5D; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (included benefit and no benefit)

2 Downgraded by 1 level for study limitations; high risk of performance and detection bias

3 Downgraded by 2 levels for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference: wide confidence interval (included both benefit and harm)

4 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (included harm and no harm)

5 Downgraded by 1 level for study limitations; high risk of performance, detection and attrition bias

6 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included benefit and no benefit)

Figuras y tablas -
Summary of findings 14. Temsirolimus + IFN‐α compared to IFN‐α (targeted agent + classic immunotherapy versus classic immunotherapy)
Summary of findings 15. Temsirolimus + bevacizumab compared to bevacizumab + IFN‐α (targeted agent + targeted agent versus targeted agent + classic immunotherapy)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (any cell type [80% clear cell])
Setting: Multinational muticentre/likely outpatient
Intervention: Temsirolimus + Bevacizumab
Comparison: Bevacizumab + IFN‐α

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Bevacizumab + IFN‐α

Risk difference with Temsirolimus + Bevacizumab

Progression‐free survival

(absolute effect size estimates based on survival rate at 12 months)

follow‐up: not reported

791
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

HR 1.10
(0.90 to 1.34)

Study population

420 per 1000

35 fewer per 1000
(107 fewer to 38 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)

follow‐up: not reported

791
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

HR 1.08
(0.90 to 1.30)

Study population

550 per 1000

26 fewer per 1000
(90 fewer to 34 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v3.0

784
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

RR 1.05
(0.98 to 1.13)

Study population

760 per 1000

38 more per 1000
(15 fewer to 99 more)

Health‐related quality of life3

assessed with: FKSI–15

Scale from: 0 to 60

no available data

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;FKSI: Functional Assessment of Cancer Therapy–Kidney Symptom Index; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included harm and no harm)

2 Downgraded by 1 level for study limitations; high risk of performance bias (we are not concerned with unclear risk of other bias)

3 Health‐related quality of life: no available data

Figuras y tablas -
Summary of findings 15. Temsirolimus + bevacizumab compared to bevacizumab + IFN‐α (targeted agent + targeted agent versus targeted agent + classic immunotherapy)
Summary of findings 16. Everolimus + bevacizumab compared to IFN α‐2a + bevacizumab (targeted agent + targeted agent versus targeted agent + classic immunotherapy)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (any cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Everolimus + Bevacizumab
Comparison: IFN α‐2a + Bevacizumab

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with IFN α‐2a + Bevacizumab

Risk difference with Everolimus + Bevacizumab

Progression‐free survival

(absolute effect size estimates based on survival rate at 18 months)

follow‐up: not reported

365
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

HR 0.91
(0.69 to 1.20)

Study population

250 per 1000

33 more per 1000
(61 fewer to 134 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)

follow‐up: not reported

365
(1 RCT)

⊕⊝⊝⊝
VERY LOW 2 3

HR 1.01
(0.75 to 1.36)

Study population

533 per 1000

3 fewer per 1000
(108 fewer to 91 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v3.0

361
(1 RCT)

⊕⊕⊝⊝
LOW 1 4

RR 1.06
(0.95 to 1.18)

Study population

762 per 1000

46 more per 1000
(38 fewer to 137 more)

Health‐related quality of life5
assessed with: EORTC QLQ‐C30 (Global health status scale: high score represent better functioning)
Scale from: 0 to 100

no available data

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;EORTC QLQ‐C30: European Organization for Research and Treatment of Cancer Quality of Life Questionnaire; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for study limitations; unclear risk of selection, performance and other bias and high risk of detection bias

2 Downgraded by 2 levels for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference: wide confidence interval (included both benefit and harm)

3 Downgraded by 1 level for study limitations; unclear risk of selection and other bias

4 Downgraded by 1 level for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included harm and no harm)

5 Health‐related quality of life: no available data

Figuras y tablas -
Summary of findings 16. Everolimus + bevacizumab compared to IFN α‐2a + bevacizumab (targeted agent + targeted agent versus targeted agent + classic immunotherapy)
Summary of findings 17. Sunitinib compared to nivolumab + ipilimumab (targeted agent versus combinations of immunotherapy)

Patient or population: Treatment‐naïve metastatic renal cell carcinoma (clear cell type); IMDC intermediate, poor risk patients only.
Setting: Multinational muticentre/likely outpatient
Intervention: Sunitinib
Comparison: Nivolumab + Ipilimumab

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Nivolumab + Ipilimumab

Risk difference with Sunitinib

Progression‐free survival

(absolute effect size estimates based on survival rate at 30 months)
follow‐up: median 32.4 months

847
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

HR 1.30
(1.11 to 1.52)

Study population

280 per 1000

89 fewer per 1000
(136 fewer to 37 fewer)

Overall survival

(absolute effect size estimates based on survival rate at 30 months)
follow‐up: median 32.4 months

847
(1 RCT)

⊕⊕⊕⊕
HIGH

HR 1.52
(1.23 to 1.89)

Study population

600 per 1000

140 fewer per 1000
(219 fewer to 67 fewer)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v4.0

1082
(1 RCT)

⊕⊕⊕⊝
MODERATE 2

RR 1.37
(1.22 to 1.53)

Study population

457 per 1000

169 more per 1000
(101 more to 242 more)

Health‐related quality of life
assessed with: FKSI‐19 (higher scores indicating fewer symptoms)

Scale from: 0 to 76
follow‐up: 24 weeks

460
(1 RCT)

⊕⊕⊕⊝
MODERATE 3

The mean health‐related quality of life was 2.6

MD 4.1 lower
(5.75 lower to 2.45 lower)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;FKSI: Functional Assessment of Cancer Therapy–Kidney Symptom IndexHR: Hazard ratio; IMDC: International Metastatic Renal Cell Carcinoma Database Consortium; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 1 level for imprecision; confidence interval crossed the assumed threshold of a clinically important difference (included harm and no harm)

2 Downgraded by 1 level for study limitations; high risk of performance and detection bias

3 Downgraded by 1 level for study limitations; high risk of performance and detection bias and unclear risk of attrition bias

Figuras y tablas -
Summary of findings 17. Sunitinib compared to nivolumab + ipilimumab (targeted agent versus combinations of immunotherapy)
Summary of findings 18. Pazopanib compared to placebo (targeted agent versus placebo)

Patient or population: Previous treated and treatment‐naïve (54%) metastatic renal cell carcinoma (clear cell type)
Setting: Multinational muticentre/likely outpatient
Intervention: Pazopanib
Comparison: Placebo

Outcomes

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with Placebo

Risk difference with Pazopanib

Progression‐free survival

(absolute effect size estimates based on survival rate at 12 months)

follow‐up: not reported

435
(1 RCT)

⊕⊕⊕⊕
HIGH

HR 0.46
(0.34 to 0.62)

Study population

180 per 1000

274 more per 1000
(165 more to 378 more)

Overall survival

(absolute effect size estimates based on survival rate at 24 months)

follow‐up: not reported

435
(1 RCT)

⊕⊕⊝⊝
LOW 1

HR 0.91
(0.72 to 1.16)

Study population

480 per 1000

33 more per 1000
(53 fewer to 110 more)

Serious adverse events (Grade 3 or 4)
assessed with: CTCAE v3.0

435
(1 RCT)

⊕⊕⊕⊕
HIGH

RR 2.00
(1.40 to 2.85)

Study population

200 per 1000

200 more per 1000
(80 more to 370 more)

Health‐related quality of life
assessed with: EORTC QLQ‐C30 (Global health status scale: high score represent better functioning but negative change from baseline represents a worsening condition)
Scale from: 0 to 100
follow‐up: 12 weeks

300
(1 RCT)

⊕⊕⊕⊕
HIGH

The mean health‐related quality of life was ‐0.5

MD 3.1 lower
(7.76 lower to 1.56 higher)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; CTCAE: Common Terminology Criteria for Adverse Events;EORTC QLQ‐C30: European Organization for Research and Treatment of Cancer Quality of Life Questionnaire; HR: Hazard ratio; RCT: Randomized controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Downgraded by 2 levels for imprecision; confidence interval crossed the line of no difference and the assumed threshold of a clinically important difference (included both benefit and harm)

Figuras y tablas -
Summary of findings 18. Pazopanib compared to placebo (targeted agent versus placebo)
Table 1. Individual targeted agents to be searched

Axitinib

Bevacizumab

Dovitinib

Erlotinib

Everolimus

Lapatinib

Pazopanib

Sorafenib

Sunitinib

Temsirolimus

Tivozanib

Other agents identified during search

Figuras y tablas -
Table 1. Individual targeted agents to be searched
Table 2. Participants disposition

Studies

Intervention(s)/ Comparator(s)

Randomised (N)

Received treatment (N)

Discontinued treatment (N)

Efficacy analysis (N)

Safety analysis (N)

Eichelberg 2015

Soreafenib/ Sunitinib

182

177

161

182

177

Sunitinib/ Sorafenib

183

176

156

183

176

Escudier 2010

Bevacizumab + IFN‐a2a

327

325

206

327

337

IFN‐a2a + Placebo

322

316

274

322

304

Escudier 2017 1

Sunitinib

546

535

438

546

535

Nivolumab + Ipilimumab

550

547

419

550

547

Hudes 2007

Temsirolimus

209

208

199

209

208

Temsirolimus + Interferon

210

208

193

210

208

Interferon

207

200

194

207

200

McDermott 2017

Sunitinib

101

100

83

101

100

Atezolizumab + Bevacizumab

101

101

69

101

101

Atezolizumab

103

103

80

103

103

Motzer 2010

Sunitinib

375

375

127

375

375

IFN‐a2a

375

360

234

375

360

Motzer 2013a

Pazopanib

557

554

486

557

554

Sunitinib

553

548

483

553

548

Motzer 2013b

Tivozanib

260

259

154

260

259

Sorafenib

257

257

192

257

257

Motzer 2014

Sunitinib/ Everolimus

233

231

192

233

231

Everolimus/ Sunitinib

238

238

201

238

238

Motzer 2019

Sunitinib

444

439

227

444

439

Avelumab + Axitinib

442

434

187

442

434

Ravaud 2015

Everolimus + Bevacizumab

182

180

175

182

180

Interferon + Bevacizumab

183

181

175

183

181

Retz 2019

Sorafenib/ Pazopanib

189

183

115

189

183

Pazopanib/ Sorafenib

188

183

110

188

183

Rini 2008

Bevacizumab + IFN‐a2b

369

366

355

369

366

IFN‐a2b

363

350

355

363

349

Rini 2014

Temsirolimus + Bevacizumab

400

393

372

400

393

Temsirolimus + Interferon

391

391

354

391

391

Rini 2016

Sunitinib

135

130

23

135

132

IMA901 + Sunitinib

204

185

28

204

202

Rini 2019a

Sunitinib

429

425

242

429

425

Pembrolizumab + Axitinib

432

429

176

432

429

Rini 2019b

Sunitinib

461

446

308

461

446

Atezolizumab + Bevacizumab

454

451

265

454

451

Sternberg 2010

Pazopanib

290

290

227

290

290

Placebo

145

145

131

145

145

Total

11590

11419

8366

11590

11437

1 Included overall population; but in the data and analyses section and summary of findings table, we used IMDC intermediate and poor risk patients for efficacy analysis.

Figuras y tablas -
Table 2. Participants disposition
Table 3. Baseline characteristics

Studies

Phase of study

Accrual

Blinding

RCC subtype

Prior therapy

Intervention

Comparator

Eichelberg 2015

3

Feb 2009 to Dec 2011

open label study

any, 87% clear cell

naïve

Soreafenib/Sunitinib

Sunitinib/Sorafenib

Escudier 2010

3

Jun 2004 to Oct 2005

double‐blind study

clear cell

naïve

Bevacizumab + IFN‐a2a

IFN‐a2a + Placebo

Escudier 2017

3

Oct 2014 to Feb 2016

open label study

clear cell

naïve

Sunitinib

Nivolumab + Ipilimumab

Hudes 2007

3

Jul 2003 to Apr 2005

open label study

any, 80% clear cell

naïve

Temsirolimus

Temsirolimus + Interferon

Inferferon

McDermott 2017

2

Jan 2014 to Mar 2015

open label study

clear cell

naïve

Sunitinib

Atezolizumab + Bevacizumab

Atezolizumab

Motzer 2010

3

Aug 2004 to Oct 2005

radiologic assessment

clear cell

naïve

Sunitinib

IFN‐a2a

Motzer 2013a

3

Aug 2008 to Sep 2011

open label study

clear cell

naïve

Pazopanib

Sunitinib

Motzer 2013b

2

Feb 2010 to Aug 2010

open label study

clear cell

naïve

Tivozanib

Sorafenib

Motzer 2014

2

Sep 2009 to Jun 2012

open label study

any, 85% clear cell

naïve

Sunitinib/ Everolimus

Everolimus/ Sunitinib

Motzer 2019

3

Mar 2016 to Dec 2017

open label study

clear cell

naïve

Sunitinib

Avelumab + Axitinib

Ravaud 2015

2

open label study

any 96% clear cell

naïve

Everolimus + Bevacizumab

Interferon + Bevacizumab

Retz 2019

3

Jun 2012 to Nov 2016

open label study

any, 87% clear cell

naïve

Sorafenib/ Pazopanib

Pazopanib/ Sorafenib

Rini 2008

3

Oct 2003 to Jul 2005

open label study

clear cell

naïve

Bevacizumab + IFN‐a2b

IFN‐a2b

Rini 2014

3

Apr 2008 to Oct 2010

open label study

any, 80% clear cell

naïve

Temsirolimus + Bevacizumab

Bevacizumab + Inferferon

Rini 2016

3

Dec 2010 to Dec 2012

open label study

clear cell

naïve

Sunitinib

IMA901 + Sunitinib

Rini 2019a

3

Oct 2016 to Jan 2018

open label study

clear cell

naïve

Sunitinib

Pembrolizumab + Axitinib

Rini 2019b

3

May 2015 to Oct 2016

open label study

clear cell

naïve

Sunitinib

Atezolizumab + Bevacizumab

Sternberg 2010

3

Apr 2006 to Apr 2007

double‐blind study

clear cell

54% naive

Pazopanib

Placebo

‐ denotes not reported

IFN: interferon; RCC: renal cell carcinoma

Figuras y tablas -
Table 3. Baseline characteristics
Comparison 1. Sorafenib versus Sunitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

1.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

1.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

1.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

1.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 1. Sorafenib versus Sunitinib
Comparison 2. Pazopanib versus Sunitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

2.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

2.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 2. Pazopanib versus Sunitinib
Comparison 3. Tivozanib versus Sorafenib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

3.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

3.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

3.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 3. Tivozanib versus Sorafenib
Comparison 4. Sorafenib versus Pazopanib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

4.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

4.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

4.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

4.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

4.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

4.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 4. Sorafenib versus Pazopanib
Comparison 5. Sunitinib versus Everolimus

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

5.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

5.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

5.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

5.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

5.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

5.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 5. Sunitinib versus Everolimus
Comparison 6. Sunitinib versus Avelumab + Axitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

6.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

6.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

6.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

6.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

6.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 6. Sunitinib versus Avelumab + Axitinib
Comparison 7. Sunitinib versus Pembrolizumab + Axitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

7.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

7.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

7.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

7.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

7.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 7. Sunitinib versus Pembrolizumab + Axitinib
Comparison 8. Sunitinib versus Atezolizumab + Bevacizumab

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

8.1 Progression‐free survival Show forest plot

2

1117

Hazard Ratio (IV, Random, 95% CI)

1.18 [1.02, 1.36]

8.2 Overall survival Show forest plot

2

1117

Hazard Ratio (IV, Random, 95% CI)

0.99 [0.73, 1.33]

8.3 Serious adverse events (Grade 3 or 4) Show forest plot

2

1098

Risk Ratio (M‐H, Random, 95% CI)

1.22 [1.00, 1.49]

8.4 Health‐related quality of life Show forest plot

2

691

Mean Difference (IV, Random, 95% CI)

1.00 [0.68, 1.32]

8.5 Response rate Show forest plot

2

1117

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.77, 1.07]

8.6 Minor adverse events (Grade 1 or 2) Show forest plot

2

1098

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.74, 0.97]

Figuras y tablas -
Comparison 8. Sunitinib versus Atezolizumab + Bevacizumab
Comparison 9. Sunitinib versus IMA901 + Sunitinib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

9.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

9.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

9.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

9.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

9.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 9. Sunitinib versus IMA901 + Sunitinib
Comparison 10. Sunitinib versus Interferon‐α (IFN‐α)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

10.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

10.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

10.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

10.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

10.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

10.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 10. Sunitinib versus Interferon‐α (IFN‐α)
Comparison 11. Temsirolimus versus IFN‐α

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

11.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

11.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

11.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

11.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

11.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

11.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 11. Temsirolimus versus IFN‐α
Comparison 12. Sunitinib versus Atezolizumab

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

12.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

12.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

12.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

12.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

12.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

12.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 12. Sunitinib versus Atezolizumab
Comparison 13. Bevacizumab + IFN versus IFN (+ placebo)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

13.1 Progression‐free survival Show forest plot

2

1381

Hazard Ratio (IV, Random, 95% CI)

0.68 [0.60, 0.77]

13.2 Overall survival Show forest plot

2

1381

Hazard Ratio (IV, Random, 95% CI)

0.88 [0.79, 0.99]

13.3 Serious adverse events (Grade 3 or 4) Show forest plot

2

1356

Risk Ratio (M‐H, Random, 95% CI)

1.31 [1.20, 1.42]

13.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

13.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 13. Bevacizumab + IFN versus IFN (+ placebo)
Comparison 14. Temsirolimus + IFN‐α versus IFN‐α

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

14.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

14.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

14.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

14.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

14.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

14.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 14. Temsirolimus + IFN‐α versus IFN‐α
Comparison 15. Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

15.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

15.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

15.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

15.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

15.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 15. Temsirolimus + Bevacizumab versus Bevacizumab + IFN‐α
Comparison 16. Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

16.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

16.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

16.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

16.4 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

16.5 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 16. Everolimus + Bevacizumab versus IFN α‐2a + Bevacizumab
Comparison 17. Sunitinib versus Nivolumab + Ipilimumab

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

17.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

17.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

17.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

17.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

17.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

17.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 17. Sunitinib versus Nivolumab + Ipilimumab
Comparison 18. Pazopanib versus Placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

18.1 Progression‐free survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

18.2 Overall survival Show forest plot

1

Hazard Ratio (IV, Random, 95% CI)

Totals not selected

18.3 Serious adverse events (Grade 3 or 4) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

18.4 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

18.5 Response rate Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

18.6 Minor adverse events (Grade 1 or 2) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 18. Pazopanib versus Placebo