Scolaris Content Display Scolaris Content Display

Monoterapia con pembrolizumab versus quimioterapia para el tratamiento del carcinoma urotelial avanzado con evolución de la enfermedad durante o después de la quimioterapia con platino. una revisión Cochrane rápida

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Bellmunt 2017 {published data only}

Bellmunt J, Sonpavde G, Wit R, Choueiri TK, Siefker‐Radtke AO, Plimack ER, et al. KEYNOTE‐045: randomized phase 3 trial of pembrolizumab (MK‐3475) versus paclitaxel, docetaxel, or vinflunine for previously treated metastatic urothelial cancer. Journal of Clinical Oncology2015 [Epub ahead of print]; Vol. 33, issue 15 Suppl. [DOI: 10.1200/jco.2015.33.15_suppl.tps4571]CENTRAL
Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee J‐L, Fong L, et al. Pembrolizumab as second‐line therapy for advanced urothelial carcinoma. New England Journal of Medicine 2017;376(11):1015‐26. [DOI: 10.1056/NEJMoa1613683]CENTRAL
De Wit R, Bajorin DF, Bellmunt J, Fradet Y, Lee J‐L, Fong L. Health‐related quality of life (HRQoL) of pembrolizumab (pembro) vs chemotherapy (chemo) for previously treated advanced urothelial cancer (UC) in KEYNOTE‐045. Journal of Clinical Oncology 2017;35(15 Suppl):4530. [DOI: 10.1200/JCO.2017.35.15_suppl.4530]CENTRAL
Merck Sharp, Dohme Corp. A study of pembrolizumab (MK‐3475) versus paclitaxel, docetaxel, or vinflunine for participants with advanced urothelial cancer (MK‐3475‐045/KEYNOTE‐045). clinicaltrials.gov/ct2/show/NCT02256436 (accessed 1 November 2017). CENTRAL
Quinn D, Bellmunt J, Wit R, Vaughn DJ, Fradet Y, Lee JL, et al. Keynote‐045: open‐label, phase 3 study of pembrolizumab versus investigator's choice of paclitaxel, docetaxel, or vinflunine for previously treated advanced urothelial cancer. Asia‐Pacific Journal of Clinical Oncology2017; Vol. 13, issue Suppl S1:35. CENTRAL
Vaughn DJ, Bellmunt J, Fradet Y, Lee JL, Fong L, Vogelzang NJ, et al. Health‐Related Quality‐of‐Life Analysis From KEYNOTE‐045: A Phase III Study of Pembrolizumab Versus Chemotherapy for Previously Treated Advanced Urothelial Cancer. Journal of Clinical Oncology 2018;36(16):1579‐1587. CENTRAL

References to studies excluded from this review

Acerta 2017 {published data only}

Acerta Pharma BV, Merck Sharp, Dohme Corp. Study of the combination of ACP‐196 and pembrolizumab in subjects with platinum resistant urothelial bladder cancer. clinicaltrials.gov/ct2/show/NCT02351739 (accessed 1 November 2017). CENTRAL

Alva 2016 {published data only}

Alva A, Gschwendt J, Loriot Y, Bellmunt J, Feng D, Poehlein C, et al. KEYNOTE‐361: randomized phase III study of pembrolizumab with or without chemotherapy versus chemotherapy alone in advanced urothelial carcinoma. Journal for ImmunoTherapy of Cancer2016; Vol. 4, issue Suppl 1. CENTRAL

Guo 2017 {published data only}

Guo T, Feng C. Pembrolizumab for advanced urothelial carcinoma. New England Journal of Medicine 2017;376(23):2303‐4. CENTRAL

Matthew 2015 {published data only}

Galsky M, Hoosier Cancer Research Network and Merck Sharp & Dohme Corp. Testing the PD‐1 inhibitor pembrolizumab as maintenance therapy after initial chemotherapy in metastatic bladder cancer. clinicaltrials.gov/ct2/show/NCT02500121 accessed 1 November 2017. CENTRAL

Mitchell 2017 {published data only}

Mitchell F. Pembrolizumab as second‐line treatment for urothelial cancer. Lancet Oncology 2017;18(4):e197. [DOI: 10.1016/S1470‐2045(17)30156‐0]CENTRAL

Powles 2017 {published data only}

Powles T, Gschwend JE, Loriot Y, Bellmunt J, Geczi L, Vulsteke C, et al. Phase 3 KEYNOTE‐361 trial: pembrolizumab (pembro) with or without chemotherapy versus chemotherapy alone in advanced urothelial cancer. Journal of Clinical Oncology2017 [Epub ahead of print]; Vol. 35, issue 15 Suppl. [DOI: 10.1200/JCO.2017.35.15_suppl.TPS4590]CENTRAL

Venniyoor 2017 {published data only}

Venniyoor A. Pembrolizumab for advanced urothelial carcinoma. New England Journal of Medicine 2017;376(23):2302‐3. [DOI: 10.1056/NEJMc1704612]CENTRAL

Bellmunt 2013

Bellmunt J, Fougeray R, Rosenberg JE, von der Maase H, Schutz FA, Salhi Y, et al. Long‐term survival results of a randomized phase III trial of vinflunine plus best supportive care versus best supportive care alone in advanced urothelial carcinoma patients after failure of platinum‐based chemotherapy. Annals of Oncology 2013;24(6):1466‐72.

Covidence [Computer program]

Veritas Health Innovation. Covidence. Version accessed 28 August 2017. Melbourne, Australia: Veritas Health Innovation, 2017.

EAU 2017

Witjes JA, Compérat E, Cowan NC, Gakis G, Hernández V, Lebret T, et al. EAU guidelines on muscle‐invasive and metastatic bladder cancer. uroweb.org/guideline/bladder‐cancer‐muscle‐invasive‐and‐metastatic/ (accessed 1 October 2017).

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 Cancer2009; Vol. 45, issue 2:228‐47.

Endnote 2011 [Computer program]

Thomson Reuters. EndNote X5. Version X5. Thomson Reuters, 2011.

Faraj 2015

Faraj SF, Munari E, Guner G, Taube J, Anders R, Hicks J, et al. Assessment of tumoral PD‐L1 expression and intratumoral CD8+ T cells in urothelial carcinoma. Urology 2015;85(3):703.e1‐6.

GLOBOCAN 2012

Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, et al. GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 [Internet]. Lyon, France: International Agency for Research on Cancer 2013;Available from: http://globocan.iarc.fr:accessed 10/2017.

GRADEpro GDT [Computer program]

McMaster University (developed by Evidence Prime). GRADEpro GDT. Version accessed 1 February 2017. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015.

Gupta 2015

Gupta S, O'Donnell P, Plimack ER, Berger R, Montgomery B, Heath K, et al. Mp68‐11 a Phase 1B Study of Pembrolizumab (Pembro; Mk‐3475) for Advanced Urothelial Cancer. The Journal of Urology 2015;193(4):e861‐2.

Guyatt 2008

Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck‐Ytter Y, Schünemann HJ, et al. GRADE: what is "quality of evidence" and why is it important to clinicians?. BMJ (Clinical Research Ed.) 2008;336(7651):995‐8.

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.

Higgins 2011a

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

Higgins 2011b

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

Higgins 2011c

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

Higgins 2011d

Deeks JJ, Higgins JP, Altman DG (editors). Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions. Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Hodi 2016

Hodi FS, Hwu WJ, Kefford R, Weber JS, Daud A, Hamid O, et al. Evaluation of immune‐related response criteria and RECIST v1.1 in patients with advanced melanoma treated with pembrolizumab. Journal of Clinical Oncology 2016;34(13):1510‐7.

Hróbjartsson 2013

Hróbjartsson A, Thomsen AS, Emanuelsson F, Tendal B, Hilden J, Boutron I, et al. Observer bias in randomized clinical trials with measurement scale outcomes: a systematic review of trials with both blinded and nonblinded assessors. Canadian Medical Association Journal 2013;185(4):E201‐11.

IQWiG 2017

Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen (IQWiG). Pembrolizumab (urothelial carcinoma) [Pembrolizumab (Urothelkarzinom)]. Institute for Quality and Efficiency in Health Care2017; Vol. Dossierbewertung A17‐46, issue Version 1.0:3‐9.

Iwai 2002

Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD‐L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD‐L1 blockade. Proceedings of the National Academy of Sciences of the United States of America 2002;99(19):12293‐7.

Kim 2015

Kim JW, Bellmunt J, Powles T, Loriot Y, Vogelzang MJ, Zambrano CC, et al. Clinical activity, safety, and biomarkers of MPDL3280A in metastatic urothelial bladder cancer: Additional analysis from phase IA study. Journal of Clinical Oncology 2015;33(7 Suppl):297.

Leitlinienprogramm Onkologie

Leitlinienprogramm Onkologie, Deutsche Krebsgesellschaft, Deutsche Krebshilfe, AWMF. S3 ‐ Guideline for early detection, diagnosis, therapy and aftercare for bladder cancer [S3 ‐ Leitlinie Früherkennung, Diagnose, Therapie und Nachsorge des Harnblasenkarzinoms]. leitlinienprogramm‐onkologie.de/Harnblasenkarzinom.92.0.html (accessed 1 October 2017).

Liang 2017

Liang F, Zhu J. Pembrolizumab for advanced urothelial carcinoma. New England Journal of Medicine 2017;376(23):2302.

Liberati 2009

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

Logothetis 1990

Logothetis CJ, Dexeus FH, Finn L, Sella A, Amato RJ, Ayala AG, et al. A prospective randomized trial comparing MVAC and CISCA chemotherapy for patients with metastatic urothelial tumors. Journal of Clinical Oncology 1990;8(6):1050‐5.

Morales 1976

Morales A, Eidinger D, Bruce AW. Intracavitary Bacillus Calmette‐Guerin in the treatment of superficial bladder tumors. The Journal of Urology 1976;116(2):180‐3.

NCCN Guideline 2017

Spiess PE, Agarwal N, Bangs R, Boorjian SA, Buyyounouski MK, Clark PE, et al. Bladder Cancer, Version 5.2017, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network 2017;15(10):1240‐67.

Park 2016

Park JC, Hahn NM. Emerging role of immunotherapy in urothelial carcinoma—Future directions and novel therapies. Urologic Oncology: Seminars and Original Investigations 2016;34(12):566‐76.

Parmar 1998

Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta‐analyses of the published literature for survival endpoints. Statistics in Medicine 1998;17(24):2815‐34. [PUBMED: 9921604]

Plimack 2017

Plimack ER, Bellmunt J, Gupta S, Berger R, Chow LQ, Juco J. Safety and activity of pembrolizumab in patients with locally advanced or metastatic urothelial cancer (KEYNOTE‐012): a non‐randomised, open‐label, phase 1b study. Lancet Oncology 2017;18(2):212‐20.

Review Manager 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Ribas 2015

Ribas A. Releasing the brakes on cancer immunotherapy. New England Journal of Medicine 2015;373(16):1490–2.

Rijnders 2017

Rijnders M, de Wit R, Boormans JL, Lolkema MP, van der Veldt AA. Systematic review of immune checkpoint inhibition in urological cancers. European Urology 2017;72(3):411‐23.

Rosenberg 2016

Rosenberg JE, Hoffman‐Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum‐based chemotherapy: a single‐arm, multicentre, phase 2 trial. The Lancet 2016;387(10031):1909‐20.

Schünemann 2011

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

Sharma 2015

Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015;348(6230):56‐61.

Sharma 2016

Sharma P, Callahan MK, Bono P, Kim J, Spiliopoulou P, Calvo E, et al. Nivolumab monotherapy in recurrent metastatic urothelial carcinoma (CheckMate 032): a multicentre, open‐label, two‐stage, multi‐arm, phase 1/2 trial. The Lancet Oncology 2016;17(11):1590‐8.

Tierney 2007

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

von der Maase 2000

von der Maase H, Hansen SW, Roberts JT, Dogliotti L, Oliver T, Moore MJ, et al. Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: results of a large, randomized, multinational, multicenter, phase III study. Journal of Clinical Oncology 2000;18(17):3068‐77.

Wang 2017

Wang PF, Chen Y, Song SY, Wang TJ, Ji WJ, Li SW, et al. Immune‐Related Adverse Events Associated with Anti‐PD‐1/PD‐L1 Treatment for Malignancies: A Meta‐Analysis. Frontiers in Pharmacology 2017;8:730.

References to other published versions of this review

Narayan 2017

Narayan V, Dahm P, Skoetz N, Risk MC, Bonfiorno C, Patel N, et al. Pembrolizumab monotherapy versus chemotherapy for treatment of advanced urothelial carcinoma with disease progression during or following platinum‐containing chemotherapy. A Cochrane Rapid Review. Cochrane Database of Systematic Reviews 2017, Issue 11. [DOI: 10.1002/14651858.CD012838]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Bellmunt 2017

Methods

Parallel RCT

Randomisation ratio: 1:1

Superiority design

Participants

Inclusion criteria:

  • Histologically or cytologically confirmed urothelial carcinoma of the renal pelvis, ureter, bladder, or urethra that showed predominantly transitional‐cell features on histologic testing.

  • Progression or recurrence of urothelial cancer following a first‐line platinum‐containing regimen (e.g. cisplatin, carboplatin) for metastatic or inoperable locally advanced disease; or adjuvant platinum‐based therapy following cystectomy for localized muscle‐invasive urothelial cancer with recurrence/progression <=12 months following completion of therapy; or neoadjuvant platinum‐containing therapy prior to cystectomy for localized muscle‐invasive urothelial cancer with recurrence <=12 months following completion of therapy.

  • No more than 2 prior lines of systemic chemotherapy for metastatic urothelial cancer.

  • Able to provide tissue for biomarker analysis from an archival tissue sample or newly obtained core or excisional biopsy of a tumour lesion not previously irradiated.

  • Measurable disease.

  • ECOG performance status of 0, 1, or 2.

  • Adequate organ function.

  • Female participants of childbearing potential have a negative urine or serum pregnancy test and willing to use 2 acceptable methods of birth control or abstain from heterosexual activity for the course of the study through 120 days after the last dose of pembrolizumab or 180 days after the last dose of paclitaxel, docetaxel, or vinflunine; or are surgically sterile.

  • Male participants must be willing to use an adequate method of contraception starting with the first dose of study medication through 120 days after the last dose of pembrolizumab or 180 days after the last dose of paclitaxel, docetaxel, or vinflunine.

Exclusion criteria:

  • Urothelial cancer that is suitable for local therapy administered with curative intent.

  • Currently participating in or has participated in a study of an investigational agent or using an investigational device within 4 weeks prior to the first dose of trial medication.

  • Diagnosis of immunodeficiency or receiving systemic steroid therapy or any other form of immunosuppressive therapy within 7 days prior to the first dose of study medication.

  • Anti‐cancer mAb within 4 weeks prior to study Day 1.

  • Not recovered from adverse events due to agents administered more than 4 weeks earlier, prior chemotherapy, targeted small molecule therapy, or radiation therapy within 2 weeks of study Day 1.

  • Not recovered from adverse events due to a previously administered agent or prior therapy with all choices of active comparator.

  • Known additional malignancy that is progressing or requires active treatment with the exception of basal cell carcinoma of the skin, squamous cell carcinoma of the skin that has undergone potentially curative therapy, or in situ cancer; or prostate cancer that was identified incidentally following cystoprostatectomy for bladder cancer that is Stage T2N0M0 or lower, Gleason score<= 6, or PSA undetectable.

  • Known active CNS metastases and/or carcinomatous meningitis.

  • Active autoimmune disease requiring systemic treatment within the past 3 months or a documented history of clinically severe autoimmune disease, or a syndrome that requires systemic or immunosuppressive agents, active cardiac disease, evidence of interstitial lung disease or active non‐infectious pneumonitis, or active infection requiring systemic therapy.

  • History of severe hypersensitivity reaction to paclitaxel, docetaxel, or to other drugs formulated with polysorbate 80 or polyoxyethylated castor oil, or to vinflunine or other vinca alkaloids.

  • Requires ongoing therapy with a medication that is a strong inhibitor or inducer of the cytochrome 3A4 (CYP3A4) enzymes.

  • Pregnant, breastfeeding, or expecting to conceive or father children within the projected duration of the trial, starting with the screening visit through 120 days after the last dose of pembrolizumab or 180 days after the last dose of paclitaxel, docetaxel, or vinflunine.

  • Prior therapy with an anti‐PD‐1 or anti‐PD‐ligand 1 agent, or with an agent directed to another co‐inhibitory T‐cell receptor.

  • HIV or active hepatitis B or hepatitis C.

  • Received a live virus vaccine within 30 days of planned start of trial treatment.

Characteristics:

  • Total number randomly assigned: 542.

  • Baseline imbalances: no.

  • Mean (range) age: pembrolizumab 67 (29‐88) years, chemotherapy 65 (26‐84) years.

  • Number of patients with ECOG 0: pembrolizumab 119, chemotherapy 106.

  • Number of patients with ECOG 1: pembrolizumab 143, chemotherapy 158.

  • Number of patients with ECOG ≥2: pembrolizumab 2, chemotherapy 4.

  • Number of patients with liver metastases: pembrolizumab 91, chemotherapy 95.

  • Number of patients with visceral metastases: pembrolizumab 240, chemotherapy 233.

  • Male sex: pembrolizumab 200 (74.1%), chemotherapy 202 (74.3%).

Interventions

Number of study centres: 748 patients were screened for enrolment in 120 sites in 29 countries (Australia, Austria, Belgium, Canada, Chile, Denmark, France, Germany, Hungary, Ireland, Israel, Italy, Japan, Republic of Korea, Netherlands, New Zealand, Norway, Peru, Poland, Portugal, Puerto Rico, Romania, Singapore, Spain, Sweden, Taiwan, Turkey, United Kingdom, United States).

Run‐in period: 11/2014‐11/2015.

Extension period: no.

Intervention: pembrolizumab 200 mg IV on Day 1 Q3W; 270 randomised patients, 266 patients received treatment.

Comparison: paclitaxel 175 mg/m2 IV or docetaxel 75 mg/m2 IV or vinflunine 320 mg/m2 IV, on Day 1 Q3W, 272 randomised patients, 255 received treatment, 84 patients received docetaxel, 84 received paclitaxel, and 87 received vinflunine.

Other co‐interventions for both groups: no.

Outcomes

Primary outcome measures

Overall survival:

  • time points measured: up to 30 months

  • time point reported: median duration of follow‐up was 14.1 months (range 9.9 to 22.1)

  • outcome definition: overall survival was defined as the time from randomisation to death from any cause

  • subgroups (of interest): performance status (ECOG 0 or 1 versus 2); time since last chemotherapy administration (< three months versus three months); degree of pretreatment; PDL‐1 tumour expression status (positive versus negative; 1% cutoff)

Progression‐free survival:

  • time points measured: up to 30 months

  • time point reported: median duration of follow‐up was 14.1 months (range 9.9 to 22.1)

  • outcome definition: progression‐free survival was defined as the time from randomisation to disease progression or death from any cause, per RECIST 1.1

  • subgroups: none (subgroup of participants with PD‐L1 positive score (10% cutoff) not relevant for this review)

Secondary outcome measures

Objective response rate:

  • time points measured: up to 30 months

  • time point reported: median duration of follow‐up was 14.1 months (range 9.9 to 22.1)

  • outcome definition: per RECIST 1.1; complete/partial response

  • subgroups: none

Progression‐free survival per modified RECIST 1.1:

  • time points measured: up to 30 months

  • time point reported: median duration of follow‐up was 14.1 months (range 9.9 to 22.1)

  • outcome definition: per modified RECIST 1.1

  • subgroups: none

Objective response reaction per modified RECIST 1.1:

  • time points measured: up to 30 months

  • time point reported: median duration of follow‐up was 14.1 months (range 9.9 to 22.1)

  • outcome definition: per modified RECIST 1.1

  • subgroups: none

Adverse event (time frame: up to 31 months):

  • time points measured: up to 30 months

  • time point reported: median duration of follow‐up was 14.1 months (range 9.9 to 22.1)

  • subgroups: none

Discontinuation of study drug due to an adverse event:

  • time points measured: up to 30 months

  • time point reported: median duration of follow‐up was 14.1 months (range 9.9 to 22.1)

  • subgroups: none

Quality of life:

  • was not listed as predefined outcome in ClinicalTrials.gov registry (NCT02256436)

Funding sources

Supported by Merck (Kenilworth, NJ)

Declarations of interest

  • Dr Bellmunt reports receiving consulting fees from Merck and Genentech.

  • Dr de Wit reports receiving fees for serving on an advisory board for Eli Lilly.

  • Dr Vaughn reports receiving fees for serving on a data and safety monitoring board from Astellas Pharma.

  • Dr Fradet reports receiving consulting fees from Astellas Pharma, Bayer, Amgen, Merck, Roche, Sanofi, and AstraZeneca, and grant support from Astellas Pharma, Amgen, and AstraZeneca.

  • Dr Lee reports receiving fees for serving on advisory boards for AstraZeneca, Astellas Pharma, Eisai, and Pfizer.

  • Dr Fong reports receiving grant support from Dendreon, Bristol‐Myers Squibb, Roche/Genentech, AbbVie, and Amgen

  • Dr. Climent reports receiving fees for serving on advisory boards for Roche, Pierre Fabre Laboratories, and Bristol‐Myers Squibb.

  • Dr Petrylak reports receiving consulting fees from Millennium Pharmaceuticals, Dendreon, Sanofi Aventis, Roche, Bayer, Johnson & Johnson, Exelixis, Ferring Pharmaceuticals, Medivation, Pfizer, Bellicum Pharmaceuticals, and Tyme Pharmaceuticals; grant support from Merck, Celgene, Agensys, Eli Lilly, Millennium Pharmaceuticals, Dendreon, Sanofi Aventis, and Roche Laboratories; and having ownership interest or involvement in Bellicum Pharmaceuticals and Tyme Pharmaceuticals.

  • Dr Choueiri reports receiving fees for serving on an advisory board for Merck.

  • Dr Necchi reports receiving consulting fees from Merck Sharp & Dohme, Roche, AstraZeneca, and Bayer, and grant support from Millennium Pharmaceuticals, Amgen, Novartis Pharmaceuticals, Merck Sharp & Dohme, Roche, and AstraZeneca.

  • Dr Gerritsen reports receiving fees for serving on advisory boards from Amgen, Astellas Pharma, Janssen‐Cilag, CureVac, Merck Sharp & Dohme, and Bayer; lecture fees from Bristol‐Myers Squibb, Janssen‐Cilag, and Bayer; and grant support from Astellas Pharma and Janssen‐Cilag.

  • Dr Gurney reports receiving fees for serving on advisory boards for Bristol‐Myers Squibb, Astellas Pharma, and Roche, and grant support from Pfizer.

  • Dr Quinn reports receiving fees for serving on advisory boards for Merck, Bristol‐Myers Squibb, Genentech, AstraZeneca, Pfizer, Novartis Pharmaceuticals, Astellas Pharma, Peloton Therapeutics, Dendreon, Exelixis, Bayer, Piramal Life Sciences, and Sanofi.

  • Dr Sternberg reports receiving honoraria from OncoGeneX, Eli Lilly, Merck, and Bristol‐Myers Squibb, and grant support from Eli Lilly, Janssen Pharmaceuticals, AstraZeneca, and Roche/Genentech.

  • Drs Mai, Poehlein, and Perini report being employees of and holding stock or stock options in Merck.

  • Dr Poehlein reports having been employed in the past by and holding stock options in Bayer Health Care Oncology and holding a patent related to the augmentation of immune response to a cancer vaccine (U.S. patent number, 20090317407A1).

  • Dr Perini reports holding pending patents related to the combination of a PD‐1 antagonist and a listeria‐based vaccine for treating prostate cancer (U.S. patient application number 20160022814 A1) and treating cancer with a combination of a PD‐1 antagonist and a vascular endothelial growth factor receptor inhibitor (U.S. patent application number 15101409).

  • Dr Bajorin reports receiving lecture fees and travel support from Merck.

  • No other potential conflict of interest relevant to this article was reported.

  • The trial was designed by academic advisors and employees of the sponsor (Merck). Data were collected by investigators and their site personnel and analysed by statisticians who were employees of the sponsor. Results were interpreted by the academic authors, by authors who were employees of the sponsor, and by other employees of the sponsor who did not fulfil all the authorship criteria as outlined by the International Committee of Medical Journal Editors. An external data and safety monitoring committee oversaw the trial and assessed efficacy and safety at the time of prespecified interim analyses that were performed by statisticians from QuintilesIMS, with funding by the sponsor.

  • The first draft of the manuscript was written by the first and last authors, with input from authors who were employees of the sponsor. Assistance with manuscript preparation was provided by a medical writer employed by the sponsor.

Notes

  • "The data and safety monitoring committee reviewed the results of the second interim analysis on October 18, 2016, and recommended early termination of the trial because pembrolizumab met the superiority thresholds for overall survival in the co‐primary populations".

  • Manuscript was published in English.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from publication: "...central randomisation as described in the protocol".

Comment: Randomization was performed adequately.

Allocation concealment (selection bias)

Low risk

Quote from publication: "Centrally...interactive voice response system/integrated web response system".

Comment: allocation concealment was performed adequately.

Blinding of participants and personnel (performance bias)
Objective outcomes

Unclear risk

Quote from publication: "...there was no blinding".

Comment: overall survival was measured and reported. It might be conceivable that even objective outcomes are influenced by lack of blinding. We finally judge that there is an unclear risk of bias.

Blinding of participants and personnel (performance bias)
Subjective outcomes

High risk

Quote from publication: "...there was no blinding".

Comment: progression‐free survival, treatment‐related mortality, discontinuation due to adverse events, and adverse events were measured and reported (quality of life was not listed as a predefined outcome in ClinicalTrials.gov registry (NCT02256436) and was not reported in the full text publication, but results were presented in abstract form at ASCO GU 2017 conference (Bellmunt 2017)). We judge that subjective outcomes are likely to be influenced by lack of blinding leading to high risk of bias. For response rate, we are uncertain if lack of blinding of participants and personnel might have influenced results, and therefore judged that there is unclear risk of bias for this outcome.

Blinding of outcome assessment (detection bias)
Objective outcome (overall survival

Low risk

Quote from publication: no applicable quote.

Comment: Not likely that outcome assessment for overall survival would be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
Subjective outcomes (all except overall survival and response rate)

High risk

Quote from publication: no applicable quote.

Comment: We judged that the assessment of these subjective outcomes is likely to be influenced by the lack of blinding.

Blinding of outcome assessment (detection bias)
Subjective outcome (treatment response (partial or complete radiological)

Low risk

Quote from publication: "Imaging data were centrally reviewed, central imaging vendor were blinded to the subject treatment, imaging results were blinded to the clinical study team".

Comment: Adequate assurance of blinding

Incomplete outcome data (attrition bias)
Oncological outcomes

Low risk

All patients randomised were included in the analysis for overall survival, progression‐free survival, and response rate.

Incomplete outcome data (attrition bias)
Quality of life

Low risk

Missing outcome data are less than 10% in both groups (randomised: 270 pembrolizumab, 272 chemotherapy; in evaluation: 266 pembrolizumab, 253 chemotherapy). We judge that this number of withdrawals is not enough to have a clinically relevant effect.

Incomplete outcome data (attrition bias)
Treatment‐related mortality, Discontinuation due to adverse events, adverse events

Low risk

Missing outcome data are less than 10% in both groups (randomised: 270 pembrolizumab, 272 chemotherapy; in evaluation: 255 pembrolizumab, 255 chemotherapy). We judge that this number of withdrawals is not enough to have a clinically relevant effect.

Selective reporting (reporting bias)

Unclear risk

Quality of life was not listed as a predefined outcome in the ClinicalTrials.gov registry (NCT02256436)

Other bias

Low risk

No other potential bias identified.

ASCO GU: Genitourinary Cancers Symposium of the American Society of Clinical Oncology; CNS: central nervous system; ECOG: Eastern Cooperative Oncology Group; HIV: human immunodeficiency virus; IV: intravenous; mAb: monoclonal antibody; NCT: ClinicalTrials.gov identifier; PD‐1: programmed cell death protein 1; PD‐L1: programmed cell death protein 1 ligand; PSA: prostatic‐specific antigen; Q3W: every three weeks; RCT: randomised controlled trial; RECIST: Response Evaluation Criteria in Solid Tumors.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Acerta 2017

Wrong comparator.

Alva 2016

First‐line therapy.

Guo 2017

Comment to another article.

Matthew 2015

Wrong intervention.

Mitchell 2017

Comment to another article.

Powles 2017

First‐line therapy.

Venniyoor 2017

Comment to another article.

Data and analyses

Open in table viewer
Comparison 1. Pembrolizumab versus chemotherapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall survival Show forest plot

1

542

Hazard Ratio (Random, 95% CI)

0.73 [0.59, 0.90]

Analysis 1.1

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 1 Overall survival.

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 1 Overall survival.

2 Quality of life (change from baseline to week 15) Show forest plot

1

520

Mean Difference (IV, Random, 95% CI)

9.05 [4.41, 13.69]

Analysis 1.2

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 2 Quality of life (change from baseline to week 15).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 2 Quality of life (change from baseline to week 15).

3 Progression‐free survival Show forest plot

1

542

Hazard Ratio (Random, 95% CI)

0.98 [0.81, 1.19]

Analysis 1.3

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 3 Progression‐free survival.

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 3 Progression‐free survival.

4 Response rate (partial and complete response) Show forest plot

1

542

Risk Ratio (M‐H, Random, 95% CI)

1.85 [1.24, 2.77]

Analysis 1.4

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 4 Response rate (partial and complete response).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 4 Response rate (partial and complete response).

5 Treatment‐related mortality Show forest plot

1

521

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.24, 3.79]

Analysis 1.5

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 5 Treatment‐related mortality.

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 5 Treatment‐related mortality.

6 Discontinuation due to adverse event (any grade) Show forest plot

1

521

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.39, 1.10]

Analysis 1.6

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 6 Discontinuation due to adverse event (any grade).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 6 Discontinuation due to adverse event (any grade).

7 Serious adverse events (irrespective of attribution to treatment) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.7

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 7 Serious adverse events (irrespective of attribution to treatment).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 7 Serious adverse events (irrespective of attribution to treatment).

7.1 Serious adverse events (Grade 3, 4, or 5)

1

521

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.72, 0.97]

8 Serious adverse events (irrespective of attribution to treatment) Show forest plot

1

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

Analysis 1.8

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 8 Serious adverse events (irrespective of attribution to treatment).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 8 Serious adverse events (irrespective of attribution to treatment).

8.1 Pruritus (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.13 [0.00, 6.54]

8.2 Fatigue (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.63 [0.28, 1.40]

8.3 Diarrhoea (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.96 [0.24, 3.87]

8.4 Anaemia (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.65 [0.37, 1.15]

8.5 Constipation (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.38 [0.11, 1.25]

8.6 Peripheral sensory neuropathy (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.13 [0.02, 0.74]

8.7 Neutropenia (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.11 [0.06, 0.22]

8.8 Alopecia (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.13 [0.01, 1.24]

8.9 Hypothyroidism (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.10 Skin reaction (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.23 [0.05, 1.00]

Open in table viewer
Comparison 2. Pembrolizumab versus chemotherapy (predefined subgroup analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall survival based on performance status Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.74 [0.59, 0.92]

Analysis 2.1

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 1 Overall survival based on performance status.

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 1 Overall survival based on performance status.

1.1 ECOG 0‐1

1

Hazard Ratio (Random, 95% CI)

0.74 [0.59, 0.93]

1.2 ECOG 2

1

Hazard Ratio (Random, 95% CI)

0.43 [0.04, 4.62]

2 Overall survival based on time since last chemotherapy Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.58, 0.91]

Analysis 2.2

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 2 Overall survival based on time since last chemotherapy.

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 2 Overall survival based on time since last chemotherapy.

2.1 Less than 3 months

1

Hazard Ratio (Random, 95% CI)

0.82 [0.58, 1.16]

2.2 Equal or greater than 3 months

1

Hazard Ratio (Random, 95% CI)

0.66 [0.49, 0.89]

3 Overall survival based on degree of pretreatment Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.57, 0.91]

Analysis 2.3

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 3 Overall survival based on degree of pretreatment.

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 3 Overall survival based on degree of pretreatment.

3.1 Two prior treatments for metastatic disease

1

Hazard Ratio (Random, 95% CI)

0.83 [0.52, 1.32]

3.2 One prior treatment for metastatic disease

1

Hazard Ratio (Random, 95% CI)

0.72 [0.54, 0.96]

3.3 Neoadjuvant

1

Hazard Ratio (Random, 95% CI)

0.53 [0.20, 1.40]

3.4 Adjuvant

1

Hazard Ratio (Random, 95% CI)

0.53 [0.18, 1.56]

4 Overall survival based on PD‐L1 tumour expression status Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.75 [0.51, 1.08]

Analysis 2.4

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 4 Overall survival based on PD‐L1 tumour expression status.

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 4 Overall survival based on PD‐L1 tumour expression status.

4.1 PD‐L1 positive (>1% cut off)

1

Hazard Ratio (Random, 95% CI)

0.61 [0.43, 0.87]

4.2 PD‐L1 negative (<1% cut off)

1

Hazard Ratio (Random, 95% CI)

0.89 [0.66, 1.20]

Open in table viewer
Comparison 3. Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall survival based on age Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.76 [0.60, 0.95]

Analysis 3.1

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 1 Overall survival based on age.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 1 Overall survival based on age.

1.1 < 65 years

1

Hazard Ratio (Random, 95% CI)

0.75 [0.53, 1.06]

1.2 ≥ 65 years

1

Hazard Ratio (Random, 95% CI)

0.76 [0.56, 1.03]

2 Overall survival based on sex Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.74 [0.59, 0.93]

Analysis 3.2

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 2 Overall survival based on sex.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 2 Overall survival based on sex.

2.1 Male

1

Hazard Ratio (Random, 95% CI)

0.73 [0.56, 0.95]

2.2 Female

1

Hazard Ratio (Random, 95% CI)

0.78 [0.49, 1.24]

3 Overall survival based on smoking status Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.68 [0.41, 1.15]

Analysis 3.3

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 3 Overall survival based on smoking status.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 3 Overall survival based on smoking status.

3.1 Current

1

Hazard Ratio (Random, 95% CI)

0.32 [0.15, 0.68]

3.2 Former

1

Hazard Ratio (Random, 95% CI)

0.71 [0.52, 0.97]

3.3 Never

1

Hazard Ratio (Random, 95% CI)

1.06 [0.72, 1.56]

4 Overall survival based on histologic type Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.54, 0.97]

Analysis 3.4

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 4 Overall survival based on histologic type.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 4 Overall survival based on histologic type.

4.1 Transitional cell

1

Hazard Ratio (Random, 95% CI)

0.80 [0.62, 1.03]

4.2 Mixed

1

Hazard Ratio (Random, 95% CI)

0.58 [0.37, 0.91]

5 Overall survival based on PD‐L1 tumour expression status (10% cutoff) Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.71 [0.51, 0.97]

Analysis 3.5

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 5 Overall survival based on PD‐L1 tumour expression status (10% cutoff).

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 5 Overall survival based on PD‐L1 tumour expression status (10% cutoff).

5.1 < 10%

1

Hazard Ratio (Random, 95% CI)

0.80 [0.61, 1.05]

5.2 ≥ 10%

1

Hazard Ratio (Random, 95% CI)

0.57 [0.37, 0.88]

6 Overall survival based on location of primary tumour Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.54, 0.95]

Analysis 3.6

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 6 Overall survival based on location of primary tumour.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 6 Overall survival based on location of primary tumour.

6.1 Upper tract

1

Hazard Ratio (Random, 95% CI)

0.53 [0.28, 1.00]

6.2 Lower tract

1

Hazard Ratio (Random, 95% CI)

0.77 [0.60, 0.99]

7 Overall survival based on location of metastases Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.73 [0.59, 0.91]

Analysis 3.7

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 7 Overall survival based on location of metastases.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 7 Overall survival based on location of metastases.

7.1 Lymph node only

1

Hazard Ratio (Random, 95% CI)

0.46 [0.18, 1.18]

7.2 Visceral disease

1

Hazard Ratio (Random, 95% CI)

0.75 [0.60, 0.94]

8 Overall survival based on liver metastases Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.74 [0.59, 0.94]

Analysis 3.8

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 8 Overall survival based on liver metastases.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 8 Overall survival based on liver metastases.

8.1 Liver metastases

1

Hazard Ratio (Random, 95% CI)

0.85 [0.61, 1.18]

8.2 No liver metastases

1

Hazard Ratio (Random, 95% CI)

0.67 [0.50, 0.90]

9 Overall survival based on haemoglobin concentration Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.57, 0.90]

Analysis 3.9

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 9 Overall survival based on haemoglobin concentration.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 9 Overall survival based on haemoglobin concentration.

9.1 < 10 g/dl

1

Hazard Ratio (Random, 95% CI)

0.75 [0.46, 1.22]

9.2 ≥ 10 g/dl

1

Hazard Ratio (Random, 95% CI)

0.71 [0.55, 0.92]

10 Overall survival based on number of risk factors Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.78 [0.62, 0.98]

Analysis 3.10

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 10 Overall survival based on number of risk factors.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 10 Overall survival based on number of risk factors.

10.1 No risk factor

1

Hazard Ratio (Random, 95% CI)

0.82 [0.42, 1.60]

10.2 1 risk factor

1

Hazard Ratio (Random, 95% CI)

0.73 [0.49, 1.09]

10.3 2 risk factors

1

Hazard Ratio (Random, 95% CI)

0.84 [0.56, 1.26]

10.4 3 or 4 risk factors

1

Hazard Ratio (Random, 95% CI)

0.76 [0.47, 1.23]

11 Overall survival based on previous platinum therapy Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.73 [0.58, 0.92]

Analysis 3.11

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 11 Overall survival based on previous platinum therapy.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 11 Overall survival based on previous platinum therapy.

11.1 Cisplatin

1

Hazard Ratio (Random, 95% CI)

0.73 [0.56, 0.95]

11.2 Carboplatin

1

Hazard Ratio (Random, 95% CI)

0.74 [0.47, 1.17]

12 Overall survival based on investigator's choice of chemotherapy Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.73 [0.61, 0.88]

Analysis 3.12

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 12 Overall survival based on investigator's choice of chemotherapy.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 12 Overall survival based on investigator's choice of chemotherapy.

12.1 Paclitaxel

1

Hazard Ratio (Random, 95% CI)

0.76 [0.55, 1.05]

12.2 Docetaxel

1

Hazard Ratio (Random, 95% CI)

0.76 [0.55, 1.05]

12.3 Vinflunine

1

Hazard Ratio (Random, 95% CI)

0.69 [0.51, 0.93]

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.
Figuras y tablas -
Figure 2

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

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

Forest plot of comparison: 1 Pembrolizumab versus chemotherapy, outcome: 1.1 Overall survival.
Figuras y tablas -
Figure 4

Forest plot of comparison: 1 Pembrolizumab versus chemotherapy, outcome: 1.1 Overall survival.

Forest plot of comparison: 1 Pembrolizumab versus chemotherapy, outcome: 1.2 Quality of life (change from baseline to week 15).
Figuras y tablas -
Figure 5

Forest plot of comparison: 1 Pembrolizumab versus chemotherapy, outcome: 1.2 Quality of life (change from baseline to week 15).

Forest plot of comparison: 1 Pembrolizumab versus chemotherapy, outcome: 1.8 Serious adverse events (irrespective of attribution to treatment).
Figuras y tablas -
Figure 6

Forest plot of comparison: 1 Pembrolizumab versus chemotherapy, outcome: 1.8 Serious adverse events (irrespective of attribution to treatment).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 1 Overall survival.
Figuras y tablas -
Analysis 1.1

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 1 Overall survival.

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 2 Quality of life (change from baseline to week 15).
Figuras y tablas -
Analysis 1.2

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 2 Quality of life (change from baseline to week 15).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 3 Progression‐free survival.
Figuras y tablas -
Analysis 1.3

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 3 Progression‐free survival.

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 4 Response rate (partial and complete response).
Figuras y tablas -
Analysis 1.4

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 4 Response rate (partial and complete response).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 5 Treatment‐related mortality.
Figuras y tablas -
Analysis 1.5

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 5 Treatment‐related mortality.

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 6 Discontinuation due to adverse event (any grade).
Figuras y tablas -
Analysis 1.6

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 6 Discontinuation due to adverse event (any grade).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 7 Serious adverse events (irrespective of attribution to treatment).
Figuras y tablas -
Analysis 1.7

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 7 Serious adverse events (irrespective of attribution to treatment).

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 8 Serious adverse events (irrespective of attribution to treatment).
Figuras y tablas -
Analysis 1.8

Comparison 1 Pembrolizumab versus chemotherapy, Outcome 8 Serious adverse events (irrespective of attribution to treatment).

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 1 Overall survival based on performance status.
Figuras y tablas -
Analysis 2.1

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 1 Overall survival based on performance status.

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 2 Overall survival based on time since last chemotherapy.
Figuras y tablas -
Analysis 2.2

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 2 Overall survival based on time since last chemotherapy.

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 3 Overall survival based on degree of pretreatment.
Figuras y tablas -
Analysis 2.3

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 3 Overall survival based on degree of pretreatment.

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 4 Overall survival based on PD‐L1 tumour expression status.
Figuras y tablas -
Analysis 2.4

Comparison 2 Pembrolizumab versus chemotherapy (predefined subgroup analyses), Outcome 4 Overall survival based on PD‐L1 tumour expression status.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 1 Overall survival based on age.
Figuras y tablas -
Analysis 3.1

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 1 Overall survival based on age.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 2 Overall survival based on sex.
Figuras y tablas -
Analysis 3.2

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 2 Overall survival based on sex.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 3 Overall survival based on smoking status.
Figuras y tablas -
Analysis 3.3

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 3 Overall survival based on smoking status.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 4 Overall survival based on histologic type.
Figuras y tablas -
Analysis 3.4

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 4 Overall survival based on histologic type.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 5 Overall survival based on PD‐L1 tumour expression status (10% cutoff).
Figuras y tablas -
Analysis 3.5

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 5 Overall survival based on PD‐L1 tumour expression status (10% cutoff).

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 6 Overall survival based on location of primary tumour.
Figuras y tablas -
Analysis 3.6

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 6 Overall survival based on location of primary tumour.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 7 Overall survival based on location of metastases.
Figuras y tablas -
Analysis 3.7

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 7 Overall survival based on location of metastases.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 8 Overall survival based on liver metastases.
Figuras y tablas -
Analysis 3.8

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 8 Overall survival based on liver metastases.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 9 Overall survival based on haemoglobin concentration.
Figuras y tablas -
Analysis 3.9

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 9 Overall survival based on haemoglobin concentration.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 10 Overall survival based on number of risk factors.
Figuras y tablas -
Analysis 3.10

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 10 Overall survival based on number of risk factors.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 11 Overall survival based on previous platinum therapy.
Figuras y tablas -
Analysis 3.11

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 11 Overall survival based on previous platinum therapy.

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 12 Overall survival based on investigator's choice of chemotherapy.
Figuras y tablas -
Analysis 3.12

Comparison 3 Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses), Outcome 12 Overall survival based on investigator's choice of chemotherapy.

Summary of findings for the main comparison. Pembrolizumab compared to chemotherapy for treatment of advanced urothelial carcinoma with disease progression during or following platinum‐containing chemotherapy. A Cochrane Rapid Review

Pembrolizumab compared to chemotherapy for treatment of advanced urothelial carcinoma with disease progression during or following platinum‐containing chemotherapy. A Cochrane Rapid Review

Participants: people with advanced urothelial carcinoma with disease progression during or following platinum‐containing chemotherapy

Setting: multicentre; 120 sites in 29 countries

Intervention: pembrolizumab

Control: chemotherapy (vinflunine or docetaxel or paclitaxel)

Outcomes

Number of participants
(studies)

Certainty of the evidence
(GRADE)

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Risk with chemotherapy

Risk difference with pembrolizumab

Time to death from any cause (here: overall mortality at 12‐month follow‐up)

542
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

HR 0.73
(0.59 to 0.90)

Study population

695 per 1000 a

115 fewer per 1000
(191 fewer to 38 fewer)

Quality of life (Change from baseline to week 15)
Assessed with: EORTC QLQ‐C30
Scale from 0 to 100 (a higher score represents better quality of life)
Follow‐up: from baseline to week 15

519
(1 RCT)

⊕⊕⊝⊝
LOW 2 3 4

MD 9.05 (4.61 to 13.50)

The mean quality of life (change from baseline to week 15) was ‐8.3 score change b

MD 9.05 score change higher
(4.61 higher to 13.50 higher)

Response rate (partial and complete response)

Follow‐up: median 14.1 months

542
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

RR 1.85
(1.24 to 2.77)

Study population

114 per 1000

97 more per 1000
(27 more to 202 more)

Treatment‐related mortality
Follow‐up: median 14.1 months

521
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

RR 0.96
(0.24 to 3.79)

Study population

16 per 1000

1 less per 1000
(12 fewer to 44 more)

Discontinuation due to adverse event
Follow‐up: median 14.1 months

521
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

RR 0.66
(0.39 to 1.10)

Study population

110 per 1000

37 fewer per 1000
(67 fewer to 11 more)

Serious adverse events (irrespective of attribution to treatment)

Follow‐up: median 14.1 months

521
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

RR 0.83

(0.72 to 0.97)

Study population

627 per 1000

107 fewer per 1000
(176 fewer to 19 fewer)

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

RR 0.66, 95% CI 0.39 to 1.10

CI: confidence interval; HR: hazard ratio; MD: mean difference; RR: risk ratio; RCT: randomised controlled trial; EORTC QLQ‐C30:.European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire‐C30

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 for imprecision due to wide confidence intervals.

2 Downgraded for study limitations (performance and detection bias)

3 Additonal concerns about selective reporting bias but not downgraded further.

4 Downgraded for imprecision; 95% CI crosses minimal clinically important difference of 10.

a The baseline risk for death of any cause in the chemotherapy group was assumed to be 69.3% at 12 months as reported by Bellmunt 2017 (at 12 months, the estimated overall survival rate was 43.9% (95% CI 37.8 to 49.9) for participants treated with pembrolizumab and 30.7% (95% CI 25.9 to 36.7) for participants treated with chemotherapy).

b Baseline risk for the chemotherapy group at 15 weeks as reported by Bellmunt 2017 ("From baseline to week 15, scores were stable for pembrolizumab (n = 266) (least squares [LS] mean +0.75 [95% CI –2.34 to +3.83]) but worsened for chemotherapy (n = 254) (LS mean –8.30 [95% CI –11.76 to –4.83]); the difference in LS means between arms was 9.05 (95% CI 4.61‐13.48; nominal 2‐sided P < 0.001)").

Figuras y tablas -
Summary of findings for the main comparison. Pembrolizumab compared to chemotherapy for treatment of advanced urothelial carcinoma with disease progression during or following platinum‐containing chemotherapy. A Cochrane Rapid Review
Table 1. Baseline characteristics

Intervention(s) and comparator(s)

Duration of follow‐up

Description of participants

Trial period (year to year)

Country

Setting

Ethnic groups (%)

Bellmunt 2017

I: pembrolizumab

Median 14.1 months (for quality of life: from randomisation to week 15)

Participants with advanced urothelial carcinoma with disease progression during or following platinum‐containing chemotherapy

2014‐2015

International

Multicentre

C: paclitaxel or docetaxel or vinflunine

C: comparator; I: intervention; : not reported.

Figuras y tablas -
Table 1. Baseline characteristics
Table 2. Participant disposition

Intervention(s) and comparator(s)

Sample size

N screened/eligible

N randomised

N ITT

N analysed for overall survival, progression‐frees survival, and response rate

N analysed for quality of life

N analysed for treatment‐related mortality, discontinuation due to adverse events, and adverse events

Follow‐upa

Bellmunt 2017

I1: Pembrolizumab

542

748

270

270

270

266

266

Median 14.1 months (9.9‐22.1; for quality of life: from randomisation to week 15)

C1: Paclitaxel or docetaxel or vinflunine

272

272

272

254

255

Total:

542

542

542

520

521

Grand total

All interventions

270

All comparators

272

All interventions and comparators

542

aFollow‐up under randomised conditions until end of trial or, if not available, duration of intervention.

C: comparator; I: intervention; ITT: intention‐to‐treat.

Figuras y tablas -
Table 2. Participant disposition
Comparison 1. Pembrolizumab versus chemotherapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall survival Show forest plot

1

542

Hazard Ratio (Random, 95% CI)

0.73 [0.59, 0.90]

2 Quality of life (change from baseline to week 15) Show forest plot

1

520

Mean Difference (IV, Random, 95% CI)

9.05 [4.41, 13.69]

3 Progression‐free survival Show forest plot

1

542

Hazard Ratio (Random, 95% CI)

0.98 [0.81, 1.19]

4 Response rate (partial and complete response) Show forest plot

1

542

Risk Ratio (M‐H, Random, 95% CI)

1.85 [1.24, 2.77]

5 Treatment‐related mortality Show forest plot

1

521

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.24, 3.79]

6 Discontinuation due to adverse event (any grade) Show forest plot

1

521

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.39, 1.10]

7 Serious adverse events (irrespective of attribution to treatment) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

7.1 Serious adverse events (Grade 3, 4, or 5)

1

521

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.72, 0.97]

8 Serious adverse events (irrespective of attribution to treatment) Show forest plot

1

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

8.1 Pruritus (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.13 [0.00, 6.54]

8.2 Fatigue (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.63 [0.28, 1.40]

8.3 Diarrhoea (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.96 [0.24, 3.87]

8.4 Anaemia (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.65 [0.37, 1.15]

8.5 Constipation (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.38 [0.11, 1.25]

8.6 Peripheral sensory neuropathy (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.13 [0.02, 0.74]

8.7 Neutropenia (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.11 [0.06, 0.22]

8.8 Alopecia (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.13 [0.01, 1.24]

8.9 Hypothyroidism (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.10 Skin reaction (Grade 3, 4, or 5)

1

521

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.23 [0.05, 1.00]

Figuras y tablas -
Comparison 1. Pembrolizumab versus chemotherapy
Comparison 2. Pembrolizumab versus chemotherapy (predefined subgroup analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall survival based on performance status Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.74 [0.59, 0.92]

1.1 ECOG 0‐1

1

Hazard Ratio (Random, 95% CI)

0.74 [0.59, 0.93]

1.2 ECOG 2

1

Hazard Ratio (Random, 95% CI)

0.43 [0.04, 4.62]

2 Overall survival based on time since last chemotherapy Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.58, 0.91]

2.1 Less than 3 months

1

Hazard Ratio (Random, 95% CI)

0.82 [0.58, 1.16]

2.2 Equal or greater than 3 months

1

Hazard Ratio (Random, 95% CI)

0.66 [0.49, 0.89]

3 Overall survival based on degree of pretreatment Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.57, 0.91]

3.1 Two prior treatments for metastatic disease

1

Hazard Ratio (Random, 95% CI)

0.83 [0.52, 1.32]

3.2 One prior treatment for metastatic disease

1

Hazard Ratio (Random, 95% CI)

0.72 [0.54, 0.96]

3.3 Neoadjuvant

1

Hazard Ratio (Random, 95% CI)

0.53 [0.20, 1.40]

3.4 Adjuvant

1

Hazard Ratio (Random, 95% CI)

0.53 [0.18, 1.56]

4 Overall survival based on PD‐L1 tumour expression status Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.75 [0.51, 1.08]

4.1 PD‐L1 positive (>1% cut off)

1

Hazard Ratio (Random, 95% CI)

0.61 [0.43, 0.87]

4.2 PD‐L1 negative (<1% cut off)

1

Hazard Ratio (Random, 95% CI)

0.89 [0.66, 1.20]

Figuras y tablas -
Comparison 2. Pembrolizumab versus chemotherapy (predefined subgroup analyses)
Comparison 3. Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall survival based on age Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.76 [0.60, 0.95]

1.1 < 65 years

1

Hazard Ratio (Random, 95% CI)

0.75 [0.53, 1.06]

1.2 ≥ 65 years

1

Hazard Ratio (Random, 95% CI)

0.76 [0.56, 1.03]

2 Overall survival based on sex Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.74 [0.59, 0.93]

2.1 Male

1

Hazard Ratio (Random, 95% CI)

0.73 [0.56, 0.95]

2.2 Female

1

Hazard Ratio (Random, 95% CI)

0.78 [0.49, 1.24]

3 Overall survival based on smoking status Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.68 [0.41, 1.15]

3.1 Current

1

Hazard Ratio (Random, 95% CI)

0.32 [0.15, 0.68]

3.2 Former

1

Hazard Ratio (Random, 95% CI)

0.71 [0.52, 0.97]

3.3 Never

1

Hazard Ratio (Random, 95% CI)

1.06 [0.72, 1.56]

4 Overall survival based on histologic type Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.54, 0.97]

4.1 Transitional cell

1

Hazard Ratio (Random, 95% CI)

0.80 [0.62, 1.03]

4.2 Mixed

1

Hazard Ratio (Random, 95% CI)

0.58 [0.37, 0.91]

5 Overall survival based on PD‐L1 tumour expression status (10% cutoff) Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.71 [0.51, 0.97]

5.1 < 10%

1

Hazard Ratio (Random, 95% CI)

0.80 [0.61, 1.05]

5.2 ≥ 10%

1

Hazard Ratio (Random, 95% CI)

0.57 [0.37, 0.88]

6 Overall survival based on location of primary tumour Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.54, 0.95]

6.1 Upper tract

1

Hazard Ratio (Random, 95% CI)

0.53 [0.28, 1.00]

6.2 Lower tract

1

Hazard Ratio (Random, 95% CI)

0.77 [0.60, 0.99]

7 Overall survival based on location of metastases Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.73 [0.59, 0.91]

7.1 Lymph node only

1

Hazard Ratio (Random, 95% CI)

0.46 [0.18, 1.18]

7.2 Visceral disease

1

Hazard Ratio (Random, 95% CI)

0.75 [0.60, 0.94]

8 Overall survival based on liver metastases Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.74 [0.59, 0.94]

8.1 Liver metastases

1

Hazard Ratio (Random, 95% CI)

0.85 [0.61, 1.18]

8.2 No liver metastases

1

Hazard Ratio (Random, 95% CI)

0.67 [0.50, 0.90]

9 Overall survival based on haemoglobin concentration Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.72 [0.57, 0.90]

9.1 < 10 g/dl

1

Hazard Ratio (Random, 95% CI)

0.75 [0.46, 1.22]

9.2 ≥ 10 g/dl

1

Hazard Ratio (Random, 95% CI)

0.71 [0.55, 0.92]

10 Overall survival based on number of risk factors Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.78 [0.62, 0.98]

10.1 No risk factor

1

Hazard Ratio (Random, 95% CI)

0.82 [0.42, 1.60]

10.2 1 risk factor

1

Hazard Ratio (Random, 95% CI)

0.73 [0.49, 1.09]

10.3 2 risk factors

1

Hazard Ratio (Random, 95% CI)

0.84 [0.56, 1.26]

10.4 3 or 4 risk factors

1

Hazard Ratio (Random, 95% CI)

0.76 [0.47, 1.23]

11 Overall survival based on previous platinum therapy Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.73 [0.58, 0.92]

11.1 Cisplatin

1

Hazard Ratio (Random, 95% CI)

0.73 [0.56, 0.95]

11.2 Carboplatin

1

Hazard Ratio (Random, 95% CI)

0.74 [0.47, 1.17]

12 Overall survival based on investigator's choice of chemotherapy Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.73 [0.61, 0.88]

12.1 Paclitaxel

1

Hazard Ratio (Random, 95% CI)

0.76 [0.55, 1.05]

12.2 Docetaxel

1

Hazard Ratio (Random, 95% CI)

0.76 [0.55, 1.05]

12.3 Vinflunine

1

Hazard Ratio (Random, 95% CI)

0.69 [0.51, 0.93]

Figuras y tablas -
Comparison 3. Pembrolizumab versus chemotherapy (post‐hoc included subgroup analyses)