Scolaris Content Display Scolaris Content Display

Cochrane Database of Systematic Reviews

Systemic treatments for metastatic cutaneous melanoma

Information

DOI:
https://doi.org/10.1002/14651858.CD011123.pub2Copy DOI
Database:
  1. Cochrane Database of Systematic Reviews
Version published:
  1. 06 February 2018see what's new
Type:
  1. Intervention
Stage:
  1. Review
Cochrane Editorial Group:
  1. Cochrane Skin Group

Copyright:
  1. Copyright © 2018 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.

Article metrics

Altmetric:

Cited by:

Cited 0 times via Crossref Cited-by Linking

Collapse

Authors

  • Sandro Pasquali

    Sarcoma Service, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy

  • Andreas V Hadjinicolaou

    Human Immunology Unit, Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK

  • Vanna Chiarion Sileni

    Medical Oncology Unit 2, Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy

  • Carlo Riccardo Rossi

    Melanoma and Sarcomas Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy

  • Simone Mocellin

    Correspondence to: Surgical Oncology Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy

    [email protected]

    [email protected]

    Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy

Contributions of authors

Simone Mocellin was the review contact person.
Sandro Pasquali and SImone Mocellin co‐ordinated contributions from co‐authors and wrote the final draft of the review.
Sandro Pasquali, Andreas V Hadjinicolaou and SImone Mocellin screened studies against eligibility criteria.
Sandro Pasquali obtained data on ongoing and unpublished studies.
Sandro Pasquali, Andreas V Hadjinicolaou and SImone Mocellin and appraised study quality.
Sandro Pasquali, Andreas V Hadjinicolaou and SImone Mocellin extracted data and sought additional information from trial authors.
Sandro Pasquali and Simone Mocellin entered data into RevMan.
Sandro Pasquali and Simone Mocellin analysed and interpreted data.
Sandro Pasquali and Simone Mocellin worked on the methods section.
Vanna Chiarion Sileni and Carlo Riccardo Rossi contributed to the writing of the review and critical revision.
Sandro Pasquali and Simone Mocellin drafted the clinical sections of the Background and responded to the clinical comments of the referees.
Sandro Pasquali and Simone Mocellin responded to methodology and statistics comments from external peer referees.
Simone Mocellin is the guarantor of the update.

Sources of support

Internal sources

  • University of Padova, Italy

External sources

  • The National Institute for Health Research (NIHR), UK

    The NIHR, UK, is the largest single funder of the Cochrane Skin Group.

Declarations of interest

Sandro Pasquali: nothing to declare.
Andreas V Hadjinicolaou: nothing to declare.
Vanna Chiarion Sileni: nothing to declare.
Carlo Riccardo Rossi: nothing to declare.
Simone Mocellin: nothing to declare.

Acknowledgements

We thank Cochrane Skin, and in particular Professor Hywel Williams, Miss Laura Prescott, and Dr Finola Delamere for their assistance.

The authors also thank Dr Mauro Apostolico for his help in retrieving articles and Dr Alessandra Costa for her assistance with uploading articles into RevMan.

The Cochrane Skin editorial base wishes to thank Bob Boyle, Cochrane Dermatology Editor for this review; Ben Carter, Statistical Editor; Ching‐Chi Chi, Methods Editor; the clinical referees, Laurence Le Cleach and Emilie Sbidian; the consumer referee, Kathie Godfrey; and Ann Jones, who copy‐edited the review.

Disclaimer

This project was supported by the National Institute for Health Research, via Cochrane Infrastructure funding to the Cochrane Skin Group. The views and opinions expressed therein are those of the authors and do not necessarily reflect those of the Systematic Reviews Programme, NIHR, NHS or the Department of Health. This project was supported by the Complex Reviews Support Unit, also funded by the National Institute for Health Research (project number 14/178/29).

Version history

Published

Title

Stage

Authors

Version

2018 Feb 06

Systemic treatments for metastatic cutaneous melanoma

Review

Sandro Pasquali, Andreas V Hadjinicolaou, Vanna Chiarion Sileni, Carlo Riccardo Rossi, Simone Mocellin

https://doi.org/10.1002/14651858.CD011123.pub2

2014 May 29

Systemic treatments for metastatic cutaneous melanoma

Protocol

Sandro Pasquali, Richard Kefford, Vanna Chiarion Sileni, Donato Nitti, Carlo Riccardo Rossi, Pierluigi Pilati, Simone Mocellin

https://doi.org/10.1002/14651858.CD011123

Differences between protocol and review

Network meta‐analysis

Given that direct comparisons between key therapies were unavailable (e.g. immune checkpoint inhibitors versus small‐molecule targeted drugs), we conducted a network meta‐analysis to compute estimates of indirect comparisons and to generate treatment rankings (Cipriani 2013; Mills 2013).

Study selection

We used the following criteria to assess randomised controlled trials (RCTs) for inclusion:

  1. studies reporting on the outcomes of interest, that is, progression‐free survival (as an efficacy outcome) and severe toxicity (as a harm outcome); and

  2. studies reporting on treatments for which high quality evidence of efficacy was available from direct comparisons and for which interventions are approved for routine use in clinical practice.

Further details on outcomes and treatments included in the network meta‐analysis are reported in the Effects of interventions section (see Network meta‐analysis findings).

We chose to include phase III and earlier phase studies because early phase trials were more likely to report on tumour response (which was a review secondary outcomes). Furthermore, early phase trials sometimes also describe survival findings (which was a review primary outcome). However, phase II trials are not designed to detect survival differences but rather tumour response differences.

We included trials with mixed disease stages if outcomes for metastatic disease were reported separately.

Evidence grading

We used the GRADE system adapted for network meta‐analysis to assess evidence quality according to four levels: high‐, moderate‐, low‐, and very low‐quality (Salanti 2014).

Quality was downgraded by one level (serious concern) or two levels (very serious concern) for study limitations (risk of bias), evidence for publication bias (assessed by inspecting a funnel plot dedicated to network meta‐analysis (Chaimani 2013)), indirectness (indirect population, intervention, control, outcomes; lack of transitivity assumption), inconsistency (between‐study statistical heterogeneity, as suggested by network meta‐analysis estimate of prediction interval crossing the null value), and imprecision (as suggested by wide confidence intervals estimated by network meta‐analysis).

Statistical analysis

Review primary outcomes were progression‐free survival and high‐grade toxicity. The outcome measure for survival data was hazard ratio (HR) and 95% confidence interval (CI). The outcome measure for toxicity was relative risk (RR) and 95% CI.

Random‐effects network meta‐analysis was carried out within a frequentist setting (Hong 2013). A common heterogeneity parameter (Tau²) was assumed across all comparisons, allowing the inclusion of comparisons based on a single RCT. Summary effects are presented with 95% CIs and predictive interval. Predictive intervals were calculated using between‐study variance (Tau²) and represents the interval where the results of future studies are expected to be, thus providing information on the magnitude of heterogeneity. They are calculated as μ ± (tαdf) x √ (τ² + SE(μ)², where tαdf is the 100 x (1 ‐ α/2)% percentile of the t‐distribution with df degrees of freedom and μ is the meta‐analysis effect estimate (Chaimani 2013).

The key assumption of network meta‐analysis is transitivity (Donegan 2013). If information about comparisons A versus B and A versus C is available, then network meta‐analysis can derive information regarding the BC comparison based on the transitivity equation (A versus B – A versus C = B versus C). Transitivity holds assuming that:

  1. the common treatment, in this case conventional chemotherapy (used to compare different drug schedules indirectly), was similar when it appeared in different trials;

  2. pair‐wise comparisons did not differ substantially with respect to the distribution of effect modifiers; and

  3. in principle, participants could be randomised to any of the treatments compared in the network.

Lack of transitivity can manifest as inconsistency between direct and indirect estimates ('loop inconsistency') or between estimates deriving from different study designs ('design inconsistency', which can occur when the relative effectiveness of treatment A versus B is different when estimated in studies with different designs, such as A versus B and A versus B versus C). We investigated inconsistency using a design‐by‐treatment interaction model, which addresses both loop and design inconsistency (Higgins 2012; White 2012).

Inconsistencies of single loops can be assessed with an inconsistency plot, where a ratio of ratio can be calculated as the ratio between the relative risk estimated by the conventional pair‐wise meta‐analysis and that estimated by the network meta‐analysis. A ratio of ratio value close to the unit indicates that the results of the two techniques are in agreement; in general, values greater than 2 suggest high inconsistency (Chaimani 2013).

Network meta‐analysis also provides a ranking probability curve of each treatment (rankogram) by calculating the probability of each treatment to achieve the best rank amongst all treatments. The surface under the cumulative ranking (SUCRA) line for each treatment, which equals one when a treatment is certain to be the best and zero when a treatment is certain to be the worst, was used for treatment ranking (Chaimani 2013; Salanti 2011). We also generated a bivariate ranking plot including both efficacy (progression‐free survival) and acceptability (the inverse of toxicity: low toxicity rates are associated with high SUCRA values): an ideal treatment should be characterised by both high efficacy and high acceptability so should appear in the right upper corner of the ranking plot.

A dedicated funnel plot (comparison‐adjusted funnel plot) can be used to assess small‐study effects (which includes publication bias) (Chaimani 2013). This plot takes into consideration that included studies estimate effects for different comparisons: therefore, there cannot be a single reference line against which symmetry can be assessed. In the absence of small‐study effect the comparison‐adjusted funnel plot should be symmetrical around the zero line.

All statistical tests were two‐sided. Statistical analysis and graph generation was performed with Stata 11.2 (Stata 2017).

Notes

Small amendment to wording of background in PLS after a query via Cochrane Library feedback in consulation with lead author.

Keywords

MeSH

PICOs

Population
Intervention
Comparison
Outcome

The PICO model is widely used and taught in evidence-based health care as a strategy for formulating questions and search strategies and for characterizing clinical studies or meta-analyses. PICO stands for four different potential components of a clinical question: Patient, Population or Problem; Intervention; Comparison; Outcome.

See more on using PICO in the Cochrane Handbook.

RAS‐RAF‐MEK‐ERK pathway. Copyright © 2018 Claire Gorry: reproduced with permission.

Figures and Tables -
Figure 1

RAS‐RAF‐MEK‐ERK pathway. Copyright © 2018 Claire Gorry: reproduced with permission.

Study flow diagram.

Figures and Tables -
Figure 2

Study flow diagram.

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

Figures and Tables -
Figure 3

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

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

Figures and Tables -
Figure 4

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

Network plot

Figures and Tables -
Figure 5

Network plot

Interval plot: network meta‐analysis results for progression‐free survival. The network included eight treatment modalities. The effect measure is reported as hazard ratio (HR). CI: confidence interval; PrI: predictive interval.

Figures and Tables -
Figure 6

Interval plot: network meta‐analysis results for progression‐free survival. The network included eight treatment modalities. The effect measure is reported as hazard ratio (HR). CI: confidence interval; PrI: predictive interval.

Interval plot: network meta‐analysis results for high grade toxicity. The network included eight treatment modalities. The effect measure is reported as relative risk (RR). CI: confidence interval; PrI: predictive interval.

Figures and Tables -
Figure 7

Interval plot: network meta‐analysis results for high grade toxicity. The network included eight treatment modalities. The effect measure is reported as relative risk (RR). CI: confidence interval; PrI: predictive interval.

Ranking plot. Ranking plot representing simultaneously the efficacy (progression‐free survival) on the X axis and the acceptability (the inverse of toxicity) on the Y axis. The network included eight treatments for patients with metastatic melanoma. Optimal treatment should be characterised by both high efficacy and acceptability and should be in the right upper corner of this graph.

Figures and Tables -
Figure 8

Ranking plot. Ranking plot representing simultaneously the efficacy (progression‐free survival) on the X axis and the acceptability (the inverse of toxicity) on the Y axis. The network included eight treatments for patients with metastatic melanoma. Optimal treatment should be characterised by both high efficacy and acceptability and should be in the right upper corner of this graph.

Comparison adjusted funnel plot for network meta‐analysis of progression‐free survival

Figures and Tables -
Figure 9

Comparison adjusted funnel plot for network meta‐analysis of progression‐free survival

Comparison 1: Polychemotherapy versus single agent chemotherapy, Outcome 1: Overall survival

Figures and Tables -
Analysis 1.1

Comparison 1: Polychemotherapy versus single agent chemotherapy, Outcome 1: Overall survival

Comparison 1: Polychemotherapy versus single agent chemotherapy, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 1.2

Comparison 1: Polychemotherapy versus single agent chemotherapy, Outcome 2: Progression‐free survival

Comparison 1: Polychemotherapy versus single agent chemotherapy, Outcome 3: Tumour response

Figures and Tables -
Analysis 1.3

Comparison 1: Polychemotherapy versus single agent chemotherapy, Outcome 3: Tumour response

Comparison 1: Polychemotherapy versus single agent chemotherapy, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 1.4

Comparison 1: Polychemotherapy versus single agent chemotherapy, Outcome 4: Toxicity (≥ G3)

Comparison 2: Chemotherapy ± tamoxifen, Outcome 1: Overall survival

Figures and Tables -
Analysis 2.1

Comparison 2: Chemotherapy ± tamoxifen, Outcome 1: Overall survival

Comparison 2: Chemotherapy ± tamoxifen, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 2.2

Comparison 2: Chemotherapy ± tamoxifen, Outcome 2: Progression‐free survival

Comparison 2: Chemotherapy ± tamoxifen, Outcome 3: Tumour response

Figures and Tables -
Analysis 2.3

Comparison 2: Chemotherapy ± tamoxifen, Outcome 3: Tumour response

Comparison 2: Chemotherapy ± tamoxifen, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 2.4

Comparison 2: Chemotherapy ± tamoxifen, Outcome 4: Toxicity (≥ G3)

Comparison 3: Temozolomide versus dacarbazine, Outcome 1: Overall survival

Figures and Tables -
Analysis 3.1

Comparison 3: Temozolomide versus dacarbazine, Outcome 1: Overall survival

Comparison 3: Temozolomide versus dacarbazine, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 3.2

Comparison 3: Temozolomide versus dacarbazine, Outcome 2: Progression‐free survival

Comparison 3: Temozolomide versus dacarbazine, Outcome 3: Tumour response

Figures and Tables -
Analysis 3.3

Comparison 3: Temozolomide versus dacarbazine, Outcome 3: Tumour response

Comparison 3: Temozolomide versus dacarbazine, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 3.4

Comparison 3: Temozolomide versus dacarbazine, Outcome 4: Toxicity (≥ G3)

Comparison 4: Chemotherapy ± interferon‐alpha, Outcome 1: Overall survival

Figures and Tables -
Analysis 4.1

Comparison 4: Chemotherapy ± interferon‐alpha, Outcome 1: Overall survival

Comparison 4: Chemotherapy ± interferon‐alpha, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 4.2

Comparison 4: Chemotherapy ± interferon‐alpha, Outcome 2: Progression‐free survival

Comparison 4: Chemotherapy ± interferon‐alpha, Outcome 3: Tumour response

Figures and Tables -
Analysis 4.3

Comparison 4: Chemotherapy ± interferon‐alpha, Outcome 3: Tumour response

Comparison 4: Chemotherapy ± interferon‐alpha, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 4.4

Comparison 4: Chemotherapy ± interferon‐alpha, Outcome 4: Toxicity (≥ G3)

Comparison 5: Chemotherapy ± interleukin‐2, Outcome 1: Overall survival

Figures and Tables -
Analysis 5.1

Comparison 5: Chemotherapy ± interleukin‐2, Outcome 1: Overall survival

Comparison 5: Chemotherapy ± interleukin‐2, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 5.2

Comparison 5: Chemotherapy ± interleukin‐2, Outcome 2: Progression‐free survival

Comparison 5: Chemotherapy ± interleukin‐2, Outcome 3: Tumour response

Figures and Tables -
Analysis 5.3

Comparison 5: Chemotherapy ± interleukin‐2, Outcome 3: Tumour response

Comparison 6: Chemotherapy ± interferon‐alpha and interleukin‐2, Outcome 1: Overall survival

Figures and Tables -
Analysis 6.1

Comparison 6: Chemotherapy ± interferon‐alpha and interleukin‐2, Outcome 1: Overall survival

Comparison 6: Chemotherapy ± interferon‐alpha and interleukin‐2, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 6.2

Comparison 6: Chemotherapy ± interferon‐alpha and interleukin‐2, Outcome 2: Progression‐free survival

Comparison 6: Chemotherapy ± interferon‐alpha and interleukin‐2, Outcome 3: Tumour response

Figures and Tables -
Analysis 6.3

Comparison 6: Chemotherapy ± interferon‐alpha and interleukin‐2, Outcome 3: Tumour response

Comparison 6: Chemotherapy ± interferon‐alpha and interleukin‐2, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 6.4

Comparison 6: Chemotherapy ± interferon‐alpha and interleukin‐2, Outcome 4: Toxicity (≥ G3)

Comparison 7: Chemotherapy ± interferon‐alpha and interleukin‐2 (first line), Outcome 1: Overall survival

Figures and Tables -
Analysis 7.1

Comparison 7: Chemotherapy ± interferon‐alpha and interleukin‐2 (first line), Outcome 1: Overall survival

Comparison 7: Chemotherapy ± interferon‐alpha and interleukin‐2 (first line), Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 7.2

Comparison 7: Chemotherapy ± interferon‐alpha and interleukin‐2 (first line), Outcome 2: Progression‐free survival

Comparison 7: Chemotherapy ± interferon‐alpha and interleukin‐2 (first line), Outcome 3: Tumour response

Figures and Tables -
Analysis 7.3

Comparison 7: Chemotherapy ± interferon‐alpha and interleukin‐2 (first line), Outcome 3: Tumour response

Comparison 7: Chemotherapy ± interferon‐alpha and interleukin‐2 (first line), Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 7.4

Comparison 7: Chemotherapy ± interferon‐alpha and interleukin‐2 (first line), Outcome 4: Toxicity (≥ G3)

Comparison 8: Chemotherapy ± Bacille Calmette‐Guérin (BCG), Outcome 1: Overall survival

Figures and Tables -
Analysis 8.1

Comparison 8: Chemotherapy ± Bacille Calmette‐Guérin (BCG), Outcome 1: Overall survival

Comparison 8: Chemotherapy ± Bacille Calmette‐Guérin (BCG), Outcome 2: Tumour response

Figures and Tables -
Analysis 8.2

Comparison 8: Chemotherapy ± Bacille Calmette‐Guérin (BCG), Outcome 2: Tumour response

Comparison 9: Chemotherapy ± Corynebacterium parvum, Outcome 1: Overall survival

Figures and Tables -
Analysis 9.1

Comparison 9: Chemotherapy ± Corynebacterium parvum, Outcome 1: Overall survival

Comparison 9: Chemotherapy ± Corynebacterium parvum, Outcome 2: Tumour response

Figures and Tables -
Analysis 9.2

Comparison 9: Chemotherapy ± Corynebacterium parvum, Outcome 2: Tumour response

Comparison 10: Anti‐CTLA4 monoclonal antibodies (first line), Outcome 1: Overall survival

Figures and Tables -
Analysis 10.1

Comparison 10: Anti‐CTLA4 monoclonal antibodies (first line), Outcome 1: Overall survival

Comparison 10: Anti‐CTLA4 monoclonal antibodies (first line), Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 10.2

Comparison 10: Anti‐CTLA4 monoclonal antibodies (first line), Outcome 2: Progression‐free survival

Comparison 10: Anti‐CTLA4 monoclonal antibodies (first line), Outcome 3: Tumour response

Figures and Tables -
Analysis 10.3

Comparison 10: Anti‐CTLA4 monoclonal antibodies (first line), Outcome 3: Tumour response

Comparison 10: Anti‐CTLA4 monoclonal antibodies (first line), Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 10.4

Comparison 10: Anti‐CTLA4 monoclonal antibodies (first line), Outcome 4: Toxicity (≥ G3)

Comparison 11: Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line), Outcome 1: Overall survival

Figures and Tables -
Analysis 11.1

Comparison 11: Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line), Outcome 1: Overall survival

Comparison 11: Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line), Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 11.2

Comparison 11: Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line), Outcome 2: Progression‐free survival

Comparison 11: Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line), Outcome 3: Tumour response

Figures and Tables -
Analysis 11.3

Comparison 11: Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line), Outcome 3: Tumour response

Comparison 11: Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line), Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 11.4

Comparison 11: Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line), Outcome 4: Toxicity (≥ G3)

Comparison 12: Anti‐PD1 monoclonal antibodies versus chemotherapy, Outcome 1: Overall survival

Figures and Tables -
Analysis 12.1

Comparison 12: Anti‐PD1 monoclonal antibodies versus chemotherapy, Outcome 1: Overall survival

Comparison 12: Anti‐PD1 monoclonal antibodies versus chemotherapy, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 12.2

Comparison 12: Anti‐PD1 monoclonal antibodies versus chemotherapy, Outcome 2: Progression‐free survival

Comparison 12: Anti‐PD1 monoclonal antibodies versus chemotherapy, Outcome 3: Tumour response

Figures and Tables -
Analysis 12.3

Comparison 12: Anti‐PD1 monoclonal antibodies versus chemotherapy, Outcome 3: Tumour response

Comparison 12: Anti‐PD1 monoclonal antibodies versus chemotherapy, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 12.4

Comparison 12: Anti‐PD1 monoclonal antibodies versus chemotherapy, Outcome 4: Toxicity (≥ G3)

Comparison 13: Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies, Outcome 1: Overall survival

Figures and Tables -
Analysis 13.1

Comparison 13: Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies, Outcome 1: Overall survival

Comparison 13: Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 13.2

Comparison 13: Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies, Outcome 2: Progression‐free survival

Comparison 13: Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies, Outcome 3: Tumour response

Figures and Tables -
Analysis 13.3

Comparison 13: Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies, Outcome 3: Tumour response

Comparison 13: Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 13.4

Comparison 13: Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies, Outcome 4: Toxicity (≥ G3)

Comparison 14: Anti‐PD1 monoclonal antibodies and anti‐CTLA4 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies alone, Outcome 1: Progression‐free survival

Figures and Tables -
Analysis 14.1

Comparison 14: Anti‐PD1 monoclonal antibodies and anti‐CTLA4 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies alone, Outcome 1: Progression‐free survival

Comparison 14: Anti‐PD1 monoclonal antibodies and anti‐CTLA4 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies alone, Outcome 2: Tumour response

Figures and Tables -
Analysis 14.2

Comparison 14: Anti‐PD1 monoclonal antibodies and anti‐CTLA4 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies alone, Outcome 2: Tumour response

Comparison 14: Anti‐PD1 monoclonal antibodies and anti‐CTLA4 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies alone, Outcome 3: Toxicity (≥ G3)

Figures and Tables -
Analysis 14.3

Comparison 14: Anti‐PD1 monoclonal antibodies and anti‐CTLA4 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies alone, Outcome 3: Toxicity (≥ G3)

Comparison 15: Chemotherapy ± sorafenib, Outcome 1: Overall survival

Figures and Tables -
Analysis 15.1

Comparison 15: Chemotherapy ± sorafenib, Outcome 1: Overall survival

Comparison 15: Chemotherapy ± sorafenib, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 15.2

Comparison 15: Chemotherapy ± sorafenib, Outcome 2: Progression‐free survival

Comparison 15: Chemotherapy ± sorafenib, Outcome 3: Tumour response

Figures and Tables -
Analysis 15.3

Comparison 15: Chemotherapy ± sorafenib, Outcome 3: Tumour response

Comparison 15: Chemotherapy ± sorafenib, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 15.4

Comparison 15: Chemotherapy ± sorafenib, Outcome 4: Toxicity (≥ G3)

Comparison 16: Chemotherapy ± elesclomol, Outcome 1: Overall survival

Figures and Tables -
Analysis 16.1

Comparison 16: Chemotherapy ± elesclomol, Outcome 1: Overall survival

Comparison 16: Chemotherapy ± elesclomol, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 16.2

Comparison 16: Chemotherapy ± elesclomol, Outcome 2: Progression‐free survival

Comparison 16: Chemotherapy ± elesclomol, Outcome 3: Tumour response

Figures and Tables -
Analysis 16.3

Comparison 16: Chemotherapy ± elesclomol, Outcome 3: Tumour response

Comparison 16: Chemotherapy ± elesclomol, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 16.4

Comparison 16: Chemotherapy ± elesclomol, Outcome 4: Toxicity (≥ G3)

Comparison 17: Chemotherapy ± anti‐angiogenic drugs, Outcome 1: Overall survival

Figures and Tables -
Analysis 17.1

Comparison 17: Chemotherapy ± anti‐angiogenic drugs, Outcome 1: Overall survival

Comparison 17: Chemotherapy ± anti‐angiogenic drugs, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 17.2

Comparison 17: Chemotherapy ± anti‐angiogenic drugs, Outcome 2: Progression‐free survival

Comparison 17: Chemotherapy ± anti‐angiogenic drugs, Outcome 3: Tumour response

Figures and Tables -
Analysis 17.3

Comparison 17: Chemotherapy ± anti‐angiogenic drugs, Outcome 3: Tumour response

Comparison 17: Chemotherapy ± anti‐angiogenic drugs, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 17.4

Comparison 17: Chemotherapy ± anti‐angiogenic drugs, Outcome 4: Toxicity (≥ G3)

Comparison 18: Single agent BRAF inhibitor, Outcome 1: Overall survival

Figures and Tables -
Analysis 18.1

Comparison 18: Single agent BRAF inhibitor, Outcome 1: Overall survival

Comparison 18: Single agent BRAF inhibitor, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 18.2

Comparison 18: Single agent BRAF inhibitor, Outcome 2: Progression‐free survival

Comparison 18: Single agent BRAF inhibitor, Outcome 3: Tumour response

Figures and Tables -
Analysis 18.3

Comparison 18: Single agent BRAF inhibitor, Outcome 3: Tumour response

Comparison 18: Single agent BRAF inhibitor, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 18.4

Comparison 18: Single agent BRAF inhibitor, Outcome 4: Toxicity (≥ G3)

Comparison 19: Single agent MEK inhibitor, Outcome 1: Overall survival

Figures and Tables -
Analysis 19.1

Comparison 19: Single agent MEK inhibitor, Outcome 1: Overall survival

Comparison 19: Single agent MEK inhibitor, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 19.2

Comparison 19: Single agent MEK inhibitor, Outcome 2: Progression‐free survival

Comparison 19: Single agent MEK inhibitor, Outcome 3: Tumour response

Figures and Tables -
Analysis 19.3

Comparison 19: Single agent MEK inhibitor, Outcome 3: Tumour response

Comparison 19: Single agent MEK inhibitor, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 19.4

Comparison 19: Single agent MEK inhibitor, Outcome 4: Toxicity (≥ G3)

Comparison 20: Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor, Outcome 1: Overall survival

Figures and Tables -
Analysis 20.1

Comparison 20: Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor, Outcome 1: Overall survival

Comparison 20: Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 20.2

Comparison 20: Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor, Outcome 2: Progression‐free survival

Comparison 20: Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor, Outcome 3: Tumour response

Figures and Tables -
Analysis 20.3

Comparison 20: Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor, Outcome 3: Tumour response

Comparison 20: Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 20.4

Comparison 20: Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor, Outcome 4: Toxicity (≥ G3)

Comparison 21: Immunostimulating agents, Outcome 1: Overall survival

Figures and Tables -
Analysis 21.1

Comparison 21: Immunostimulating agents, Outcome 1: Overall survival

Comparison 21: Immunostimulating agents, Outcome 2: Progression‐free survival

Figures and Tables -
Analysis 21.2

Comparison 21: Immunostimulating agents, Outcome 2: Progression‐free survival

Comparison 21: Immunostimulating agents, Outcome 3: Tumour response

Figures and Tables -
Analysis 21.3

Comparison 21: Immunostimulating agents, Outcome 3: Tumour response

Comparison 21: Immunostimulating agents, Outcome 4: Toxicity (≥ G3)

Figures and Tables -
Analysis 21.4

Comparison 21: Immunostimulating agents, Outcome 4: Toxicity (≥ G3)

Summary of findings 1. Anti‐PD1 monoclonal antibodies versus chemotherapy

Anti‐PD1 monoclonal antibodies compared with chemotherapy for the treatment of metastatic melanoma

Patient or population: people with cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: anti‐PD1 monoclonal antibodies

Comparison: chemotherapy

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Chemotherapy

Anti‐PD1

Overall survival

600 per 1000

320 per 1000
(290 to 360)

HR 0.42

(0.37 to 0.48)

N = 418
(n = 1)

⊕⊕⊕⊕
higha

Progression‐free survival

850 per 1000

610 per 1000
(520 to 690)

HR 0.49 (0.39 to 0.61)

N = 957
(n = 2)

⊕⊕⊕⊝
moderateb

Tumour response

81 per 1000

277 per 1000
(193 to 398)

RR 3.42

(2.38 to 4.92)

N = 1367
(n = 3)

⊕⊕⊕⊕
higha

Toxicity (≥ G3)

300 per 1000

165 per 1000
(93 to 291)

RR0.55 (0.31 to 0.97)

N = 1360
(n = 3)

⊕⊕⊝⊝
lowc

* The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. death rates and progression rates).

CI: confidence interval; HR: hazard ratio

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

Assumed risk in the control population: 1‐year overall survival rate = 40%.

Assumed risk in the control population: 1‐year progression‐free survival rate = 15%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Not downgraded: high‐quality evidence.

b Downgraded by one level: inconsistency (between‐study heterogeneity).

c Downgraded by two levels: inconsistency (between‐study heterogeneity) and imprecision (CI includes both a meaningful benefit (relative risk reduction > 25%) and a small/null benefit (relative risk reduction < 10%)).

Figures and Tables -
Summary of findings 1. Anti‐PD1 monoclonal antibodies versus chemotherapy
Summary of findings 2. Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies

Anti‐PD1 monoclonal antibodies compared with anti‐CTLA4 monoclonal antibodies for the treatment of metastatic melanoma

Patient or population: people with cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: anti‐PD1 monoclonal antibodies

Comparison: anti‐CTLA4 monoclonal antibodies

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Anti‐CTLA4

Anti‐PD1

Overall survival

600 per 1000

438 per 1000
(423 to 454)

HR 0.63

(0.60 to 0.66)

N = 764
(n = 1)

⊕⊕⊕⊕
higha

Progression‐free survival

850 per 1000

641 per 1000
(612 to 679)

HR 0.54

(0.50 to 0.60)

n = 1465
(n = 2)

⊕⊕⊕⊕
higha

Tumour response

157 per 1000

388 per 1000
(315 to 477)

RR 2.47

(2.01 to 3.04)

N = 1465
(n = 2)

⊕⊕⊕⊕
higha

Toxicity (≥ G3)

398 per 1000

278 per 1000
(215 to 362)

RR 0.70

(0.54 to 0.91)

N = 1465
(n = 2)

⊕⊕⊝⊝
lowb

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. death rates and progression rates).

CI: confidence interval; RR: risk ratio; HR: hazard ratio.

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

Assumed risk in the control population: 1‐year overall survival rate = 40%.

Assumed risk in the control population: 1‐year progression‐free survival rate = 15%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Not downgraded: high‐quality evidence.

b Downgraded by two levels: inconsistency (between‐study heterogeneity) and imprecision (CI includes both a meaningful benefit (relative risk reduction > 25%) and a small/null benefit (relative risk reduction < 10%).

Figures and Tables -
Summary of findings 2. Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies
Summary of findings 3. Anti‐CTLA4 monoclonal antibodies plus chemotherapy versus chemotherapy

Anti‐CTLA4 monoclonal antibodies plus chemotherapy compared with chemotherapy for the treatment of metastatic melanoma

Patient or population: people with cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: anti‐CTLA4 monoclonal antibodies plus chemotherapy (combo)

Comparison: chemotherapy

Outcomes

Illustrative comparative risks* (95% CI)

Relative Effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Chemotherapy

Combo

Overall survival

600 per 1000

524 per 1000
(449 to 604)

HR 0.81 (0.65 to 1.01)

N = 1157
(n = 2)

⊕⊕⊝⊝
lowa

Progression‐free survival

850 per 1000

763 per 1000
(697 to 825)

HR 0.76 (0.63 to 0.92)

N = 502
(n = 1)

⊕⊕⊕⊝
moderateb

Tumour response

100 per 1000

128 per 1000
(92 to 177)

RR 1.28 (0.92 to 1.77)

N = 1157
(n = 2)

⊕⊕⊕⊝
moderatec

Toxicity (≥ G3)

352 per 1000

595 per 1000
(419 to 852)

RR 1.69 (1.19 to 2.42)

N = 1142
(n = 2)

⊕⊕⊕⊝
moderated

* The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. death rates and progression rates).

CI: confidence interval; HR: hazard ratio.

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

Assumed risk in the control population: 1‐year overall survival rate = 40%.

Assumed risk in the control population: 1‐year progression‐free survival rate = 15%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Downgraded by two levels: inconsistency (between‐study heterogeneity) and imprecision (CI includes both a meaningful benefit (relative risk reduction > 25%) and a harmful effect).

b Downgraded by one level: imprecision (CI includes both a meaningful benefit (relative risk reduction > 25%) and a small/null benefit (relative risk reduction < 10%)).

c Downgraded by one level: imprecision (CI includes both a meaningful benefit (relative risk increase > 25%) and a harmful effect).

d Downgraded by one level: inconsistency (between‐study heterogeneity).

Figures and Tables -
Summary of findings 3. Anti‐CTLA4 monoclonal antibodies plus chemotherapy versus chemotherapy
Summary of findings 4. Anti‐CTLA4 monoclonal antibodies with versus without anti‐PD1 monoclonal antibodies

Anti‐CTLA4 plus anti‐PD1 monoclonal antibodies compared with anti‐CTLA4 monoclonal antibodies for the treatment of metastatic melanoma

Patient or population: people with cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: Anti‐CTLA4 plus Anti‐PD1 monoclonal antibodies (combo)

Comparison: Anti‐CTLA4 monoclonal antibodies

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Anti‐CTLA4

Combo

Overall survival

See comment

See comment

See comment

See comment

See comment

Outcome not measured

Progression‐free survival

750 per 1000

425 per 1000
(375 to 478)

HR 0.40

(0.35 to 0.46)

N = 738
(n = 2)

⊕⊕⊕⊕
higha

Tumour response

182 per 1000

636 per 1000
(376 to 1073)

RR 3.50 (2.07 to 5.92)

N = 738
(n = 2)

⊕⊕⊕⊕
higha

Toxicity (≥ G3)

521 per 1000

818 per 1000
(442 to 1521)

RR 1.57 (0.85 to 2.92)

N = 764
(n = 2)

⊕⊕⊝⊝
lowb

* The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. progression rates).

CI: confidence interval

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

Assumed risk in the control population: 1‐year progression‐free survival rate = 25%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Not downgraded: high‐quality evidence.

b Downgraded by two levels: inconsistency (between‐study heterogeneity) and imprecision (CI includes both a meaningful harm (relative risk increase > 25%) and a beneficial effect)

Figures and Tables -
Summary of findings 4. Anti‐CTLA4 monoclonal antibodies with versus without anti‐PD1 monoclonal antibodies
Summary of findings 5. BRAF inhibitors versus chemotherapy

BRAF inhibitors compared with chemotherapy for the treatment of metastatic melanoma

Patient or population: people with cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: BRAF inhibitors

Comparison: chemotherapy

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Chemotherapy

BRAF inhibitors

Overall survival

600 per 1000

307 per 1000
(226 to 407)

HR 0.40

(0.28 to 0.57)

N = 925
(n = 2)

⊕⊕⊕⊕
higha

Progression‐free survival

850 per 1000

401 per 1000
(328 to 475)

HR 0.27

(0.21 to 0.34)

N = 925
(n = 2)

⊕⊕⊕⊕
higha

Tumour response

82 per 1000

556 per 1000
(397 to 778)

RR 6.78

(4.84 to 9.49)

N = 925
(n = 2)

⊕⊕⊕⊕
higha

Toxicity (≥ G3)

341 per 1000

433 per 1000
(163 to 1135)

RR 1.27 (0.48 to 3.33)

N = 408
(n = 2)

⊕⊕⊝⊝
lowb

* The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. death rates and progression rates).
CI: confidence interval; HR: hazard ratio.

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

Assumed risk in the control population: 1‐year overall survival rate = 40%.

Assumed risk in the control population: 1‐year progression‐free survival rate = 15%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Not downgraded: high‐quality evidence.

b Downgraded by two levels: inconsistency (between‐study heterogeneity) and imprecision (CI includes both a meaningful harm (relative risk increase > 25%) and a meaningful benefit (relative risk reduction > 25%)).

Figures and Tables -
Summary of findings 5. BRAF inhibitors versus chemotherapy
Summary of findings 6. MEK inhibitors versus chemotherapy

MEK inhibitors compared with chemotherapy for the treatment of metastatic melanoma

Patient or population: people with cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: MEK inhibitors

Comparison: chemotherapy

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Chemotherapy

MEK inhibitors

Overall survival

600 per 1000

541 per 1000
(412 to 682)

HR 0.85

(0.58 to 1.25)

N = 496
(n = 3)

⊕⊕⊝⊝
lowa

Progression‐free survival

850 per 1000

667 per 1000
(549 to 781)

HR 0.58

(0.42 to 0.80)

N = 496
(n = 3)

⊕⊕⊕⊝
moderateb

Tumour response

138 per 1000

277 per 1000
(186 to 413)

RR 2.01

(1.35 to 2.99)

N = 496
(n = 3)

⊕⊕⊕⊕
highc

Toxicity (≥ G3)

413 per 1000

665 per 1000
(446 to 995)

RR 1.61

(1.08 to 2.41)

N = 91
(n = 1)

⊕⊕⊕⊝
moderated

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. death rates and progression rates).
CI: confidence interval; RR: risk ratio; HR: hazard ratio.

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

Assumed risk in the control population: 1‐year overall survival rate = 40%.

Assumed risk in the control population: 1‐year progression‐free survival rate = 15%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Downgraded by two levels: inconsistency (between‐study heterogeneity) and imprecision (CI includes both a meaningful benefit (relative risk reduction > 25%) and a harmful effect).

b Downgraded by one level: inconsistency (between‐study heterogeneity).

c Not downgraded: high‐quality evidence.

d Downgraded by one level: imprecision (sample size lower than optimal information size, calculated to be equal to 400 participants).

Figures and Tables -
Summary of findings 6. MEK inhibitors versus chemotherapy
Summary of findings 7. BRAF plus MEK inhibitors versus BRAF inhibitors

BRAF plus MEK inhibitors compared with BRAF inhibitors for the treatment of metastatic melanoma

Patient or population: people cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: BRAF inhibitor plus MEK inhibitor (combo)

Comparison: BRAF inhibitor

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

BRAF inhibitor

Combo

Overall survival

350 per 1000

260 per 1000
(204 to 321)

HR 0.70

(0.59 to 0.82)

N = 1784
(n = 4)

⊕⊕⊕⊕
higha

Progression‐free survival

700 per 1000

490 per 1000
(411 to 574)

HR 0.56 (0.44 to 0.71)

N = 1784
(n = 4)

⊕⊕⊕⊝
moderateb

Tumour response

494 per 1000

652 per 1000
(593 to 721)

RR 1.32

(1.20 to 1.46)

N = 1784
(n = 4)

⊕⊕⊕⊕
higha

Toxicity (≥ G3)

495 per 1000

500 per 1000
(421 to 594)

RR 1.01 (0.85 to 1.20)

N = 1774
(n = 4)

⊕⊕⊕⊝
moderateb

* The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. death rates and progression rates).

CI confidence interval; HR: hazard ratio

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

Assumed risk in the control population: 1‐year overall survival rate = 65%.

Assumed risk in the control population: 1‐year progression‐free survival rate = 30%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Not downgraded: high‐quality evidence.

b Downgraded by one level: inconsistency (between‐study heterogeneity).

Figures and Tables -
Summary of findings 7. BRAF plus MEK inhibitors versus BRAF inhibitors
Summary of findings 8. Anti‐angiogenic drugs plus chemotherapy versus chemotherapy

Anti‐angiogenic drugs plus chemotherapy compared with chemotherapy for the treatment of metastatic melanoma

Patient or population: people with cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: Anti‐angiogenic drug plus chemotherapy (combo)

Comparison: chemotherapy

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Chemotherapy

Combo

Overall survival

600 per 1000

423 per 1000
(338 to 524)

HR 0.60

(0.45 to 0.81)

N = 324
(n = 2)

⊕⊕⊕⊝
moderatea

Progression‐free survival

850 per 1000

730 per 1000
(627 to 825)

HR 0.69

(0.52 to 0.92)

N = 324
(n = 2)

⊕⊕⊕⊝
moderatea

Tumour response

104 per 1000

178 per 1000
(100 to 315)

RR 1.71 (0.96 to 3.03)

N = 324
(n = 2)

⊕⊕⊕⊝
moderatea

Toxicity (≥ G3)

272 per 1000

185 per 1000
(25 to 1447)

RR 0.68 (0.09 to 5.32)

N = 324
(n = 2)

⊕⊕⊝⊝
lowb

* The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. death rates and progression rates).
CI: confidence interval; HR: hazard ratio.

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

Assumed risk in the control population: 1‐year overall survival rate = 40%.

Assumed risk in the control population: 1‐year progression‐free survival rate = 15%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Downgraded by one level: imprecision (sample size lower than optimal information size, calculated to be equal to 400 participants).

b Downgraded by two levels: inconsistency (between‐study heterogeneity) and imprecision (sample size lower than optimal information size, calculated to be equal to 400 participants).

Figures and Tables -
Summary of findings 8. Anti‐angiogenic drugs plus chemotherapy versus chemotherapy
Summary of findings 9. Biochemotherapy versus chemotherapy

Biochemotherapy compared with chemotherapy for the treatment of metastatic melanoma

Patient or population: people with cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: biochemotherapy (chemotherapy combined with both interferon‐alpha and interleukin‐2)

Comparison: chemotherapy

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Chemotherapy

Biochemotherapy

Overall survival

600 per 1000

577 per 1000
(537 to 621)

HR 0.94

(0.84 to 1.06)

N = 1317
(n = 7)

⊕⊕⊕⊕
higha

Progression‐free survival

850 per 1000 °

818 per 1000
(793 to 847)

HR 0.90

(0.83 to 0.99)

N = 964
(n = 6)

⊕⊕⊕⊕
higha

Tumour response

192 per 1000

262 per 1000
(214 to 321)

RR 1.36

(1.12 to 1.66)

N = 770
(n = 7)

⊕⊕⊕⊕
higha

Toxicity (≥ G3)

631 per 1000

852 per 1000
(719 to 1000)

RR 1.35

(1.14 to 1.61)

N = 631
(n = 2)

⊕⊕⊕⊕
higha

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. death rates and progression rates).
CI: confidence interval; RR: risk ratio; HR: hazard ratio.

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

Assumed risk in the control population: 1‐year overall survival rate = 40%.

Assumed risk in the control population: 1‐year progression‐free survival rate = 15%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Not downgraded: high‐quality evidence.

Figures and Tables -
Summary of findings 9. Biochemotherapy versus chemotherapy
Summary of findings 10. Polychemotherapy versus chemotherapy

Polychemotherapy compared with chemotherapy for the treatment of metastatic melanoma

Patient or population: people with cutaneous melanoma

Settings: hospital (metastatic disease)

Intervention: polychemotherapy

Comparison: chemotherapy

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Chemotherapy

Polychemotherapy

Overall survival

600 per 1000

596 per 1000
(541 to 655)

HR 0.99

(0.85 to 1.16)

N = 594
(n = 6)

⊕⊕⊕⊕
higha

Progression‐freesurvival

850 per 1000

869 per 1000

(822 to 907)

HR 1.07

(0.91 to 1.25)

N = 398

(n = 5)

⊕⊕⊕⊕
higha

Tumour response

143 per 1000

182 per 1000
(146 to 226)

RR 1.27

(1.02 to 1.58)

N = 1885
(n = 5)

⊕⊕⊕⊝
moderateb

Toxicity (≥ G3)

189 per 1000

372 per 1000
(272 to 512)

RR 1.97

(1.44 to 2.71)

N = 390
(n = 3)

⊕⊕⊕⊝
moderatec

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

† Numbers presented refer to event rates (i.e. death rates and progression rates).
CI: confidence interval; RR: risk ratio; HR: hazard ratio

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

Assumed risk in the control population: 1‐year overall survival rate = 40%.

Assumed risk in the control population: 1‐year progression‐free survival rate = 15%.

Assumed risk in the control population: tumour response rate across control arms of included trials.

Assumed risk in the control population: toxicity rate across control arms of included trials.

a Not downgraded: high‐quality evidence.

b Downgraded by one level: imprecision (CI includes both a meaningful benefit (relative risk increase > 25%) and a small/null benefit (relative risk increase < 10%)).

c Downgraded by one level: imprecision (sample size lower than optimal information size, calculated to be equal to 400 participants).

Figures and Tables -
Summary of findings 10. Polychemotherapy versus chemotherapy
Table 1. Glossary of terms used

Term

Explanation

Actinomycin‐D

A polypeptide used as an antibiotic and antineoplastic agent as a result of its ability to inhibit transcription

AJCC TNM staging

This is the most widely used tumour staging classification system, which has been developed and constantly updated by the American Joint Committee on Cancer (AJCC) for describing the extent of disease progression in people with cancer. It uses in part the TNM scoring system: tumour size, lymph nodes affected, metastases. Individuals affected by specific tumour type are assigned to categories describing risk of death

AJCC TNM stage III

People at this disease stage have melanoma metastasis in their regional lymph node (i.e. the first lymph nodes draining the skin area affected by the melanoma)

AJCC TNM stage IIIC

Stage IIIC is a higher risk subgroup among people with lymph node metastasis. The category includes people with all primary tumour stages (T stages) and those with clinically positive lymph nodes, or 4 or more positive lymph nodes

AJCC TNM stage IV

People with this disease stage have melanoma metastasis to distant sites (e.g. lung, liver, brain, bone)

Anti‐angiogenic agents

Drugs aimed to disrupt tumour vascularisation and reduce blood supply to malignant cells; examples include bevacizumab and endostar

Antigen

A substance that invokes the body's immune response

Aranoza

An alkylating agent that is used as a chemotherapy drug for various cancers including melanoma as part of combination chemotherapy regimens

Bacille Calmette‐Guérin (BCG)

BCG is a vaccine used in the prevention of tuberculosis. However, it is also a form of cancer immunotherapy with established effects in superficial (non‐muscle invading) bladder cancer

Bevacizumab

Bevacizumab (Avastin) is an angiogenesis inhibitor approved for use for people with various metastatic cancers. Bevacizumab acts through blockade of vascular endothelial growth factor A (VEGF‐A) that prevents development of new vessels necessary for tumours to grow

Bleomycin

An antineoplastic agent used in chemotherapy regimens for various tumours. Belomycin acts through cleavage of DNA within cells

Biochemotherapy

A combination of chemotherapy plus immunostimulating cytokines, such as interleukin‐2 and interferon‐alpha

Bosentan

An endothelin receptor inhibitor that causes reduced DNA synthesis and promotes apoptosis through competitive antagonism with the anti‐apoptotic factor endothelin‐1, often secreted by cancer cells in an autocrine or paracrine manner

BRAF

A gene that makes a protein called B‐Raf. BRAF is involved in sending signals within cells that direct their growth. In some cancers, this gene has mutated (Melanoma Institute Australia 2017)

Carmustine

An alkylating agent that prevents DNA replication and cell proliferation used in chemotherapy for various cancers

Cobimetinib

An inhibitor of MAPK kinase (MEK) approved for use in metastatic melanoma with BRAF V600E/K mutation usually in combination with a BRAF inhibitor

Corynebacterium parvum

C parvum is an aerobic, gram positive bacterium that has been reported to have antineoplastic potential

Cyclophosphamide

An alkylating agent used in auto‐immune diseases and various tumours as a chemotherapy drug

Cytokine

Small proteins produced by a broad range of cells that are important in cell signalling; they are immunostimulating agents

Cytotoxic

Cell killing

CTLA4 (cytotoxic T‐cell lymphocyte‐associated antigen‐4)

CTLA4 is a receptor located on the surface of T‐cells that down regulates the immune system (an immune checkpoint). The inhibition of this receptor with monoclonal antibodies, such as ipilimumab and tremelimumab, 'unleashes' the immune response to fight against malignant cells

Dabrafenib

An inhibitor of the BRAF kinase that has been approved for people with advanced melanoma carrying the BRAF V600E mutation

Dacarbazine

A chemotherapy drug that belongs to the family of alkylating agents that is used in the treatment of various cancers, including melanoma

Dendritic cell

These are antigen‐presenting cells that link the innate to the adaptive immune systems via processing antigens and presenting them to T‐lymphocytes. Their role is crucial for proper functioning of vaccines, including cancer vaccines

Elesclomol

A drug that causes the accumulation of reactive oxygen species to trigger apoptosis in cancer cells via oxidative stress. It is approved for use for people with metastatic melanoma

Endostar

A modified recombinant human endostatin that acts as an anti‐angiogenic agent to prevent the formation of new blood vessels that are necessary for tumour growth and survival

Fotemustine

A chemotherapy drug that belongs to the family of alkylating agents and has been approved for the treatment of metastatic melanoma

G3 and G4

G3 (grade 3) and G4 (grade 4) toxicity refers to the highest degree of adverse events due to a systemic treatment. This system grades the toxicity related to a given system or organ (e.g. hepatic, cardiac, haematologic)

gp100

A known melanoma antigen that can be applied to develop a cancer vaccine through processing and presentation by dendritic cells to lymphocytes

Granulocyte macrophage ‐ colony‐stimulating factor (GM‐CSF)

A cytokine that stimulates stem cells to give rise to granulocytes and monocytes and boosts the immune system

Hydroxyurea

A chemotherapy agent that acts through reducing the generation of deoxyribonucleotides, the building blocks of DNA, to inhibit adequate synthesis of DNA. It is used as a chemotherapy drug for people with myeloproliferative disorders

Immune checkpoints

Signalling proteins that protect against auto‐immunity and regulate the immune response; these checkpoints can be hijacked by cancer cells to evade T‐cell‐mediated death, i.e. stopping an immune response to the tumour. CTLA4 and PD1 are both immune checkpoints

Immune checkpoint inhibitors

Drugs that override the signalling/activation of immune checkpoints to encourage cytotoxic T‐cell recognition of cancer (i.e. an immune response). These are monoclonal antibodies blocking either CTLA4 or PD1 (two immune checkpoints), known as anti‐CTLA4 and anti‐PD1 monoclonal antibodies

Immunomodulating

Stimulates or suppresses the immune system

Immunostimulating

Stimulates an immune response

Interferon‐alpha

Interferon‐alpha is used for the postoperative treatment of people with AJCC TNM stages II (primary tumour at high risk of disease progression with negative lymph nodes) and III (positive lymph nodes) and to enhance the efficacy of chemotherapy in those who have metastatic melanoma

Interleukin‐2

Interleukin‐2 is a protein that regulates the activities of leucocytes (particularly lymphocytes) that are responsible for immunity. The receptor for interleukin‐2 is expressed by lymphocytes. A recombinant form of human interleukin‐2 has been approved by the FDA for the treatment of melanoma and renal cell cancer

Lomustine

An oral alkylating chemotherapeutic agent used mainly to treat brain tumours because it crosses the blood‐brain barrier

MEK

Mitogen‐activated protein kinase (MEK) is part of the MAPK signalling pathway (see 'RAS‐RAF‐MEK‐ERK pathway' below), which is activated in melanoma

Monoclonal antibodies

Monoclonal antibodies are a type of targeted drug therapy; they work by recognising and finding specific proteins on cancer cells (they work in different ways depending on the protein they are targeting) (Cancer Research UK 2017)

Oblimersen

A bcl‐2 antisense oligodeoxynucleotide that reduces cancer cell survival and proliferation by blocking the generation of the anti‐apoptotic protein bcl‐2 thus promoting programmed cell death in cancer cells

Oncogene

A gene thats activation or over expression favours cancer growth

Paclitaxel

A chemotherapy agent targeting the protein tubulin. The drug interferes with the dynamics of microtubule formation and breakdown leading to problems during cell division and triggering of apoptosis. DHA‐ and nab‐paclitaxel are modified forms of the drug

PD1 (programmed cell death protein‐1)

PD1 is a receptor located on the surface of the T‐cells that down regulates the immune system (an immune checkpoint). The inhibition of this receptor with monoclonal antibodies, such as nivolumab and pembrolizumab, 'unleashes' immune response to fight against malignant cells

PF‐3512676

An synthetic oligonucleotide that acts as a Toll‐like receptor‐9 (TLR‐9) agonist. It is used as an immunomodulatory agent alone, or in combination with chemotherapy, to boost anti‐tumour effects by enhancing B‐cell proliferation and antigen‐specific antibody production and cytokine secretion

Polychemotherapy

A combination of multiple chemotherapeutic agents

Procarbazine

An alkylating agent used as an antineoplastic chemotherapy drug in various tumours such as glioblastoma multiforme and Hodgkin's lymphoma

Programmed death‐1 (PD‐1)

PD‐1 is an inhibitory receptor located on the surface of the T‐cells that down regulates the immune system when bound by its ligands (PD‐L1 and PD‐L2, often found on cancer cells). The inhibition of this receptor with monoclonal antibodies, such as pembrolizumab and nivolumab, releases the brake on immune cells thus allowing them to freely fight malignant cells

Ramucirumab

A human monoclonal antibody that targets the vascular endothelial growth factor receptor 2 (VEGFR2) to block VEGF binding and thus inhibit angiogenesis. It is approved for use in advanced gastric adenocarcinoma and metastatic non‐small cell lung carcinoma

RAS‐RAF‐MEK‐ERK pathway

This is also known as 'MAPK/ERK pathway', which is a chain of proteins in the cell that communicates a signal from a receptor on the surface of the cell to the nucleus of the cell (where DNA is located). When one of the proteins in the pathway is mutated, it can be stuck in the 'on' or 'off' position, which is a necessary step in the development of many cancers, including melanoma. Drugs, such as BRAF and MEK inhibitors, can reverse this switch

Small‐molecule inhibitors

Low molecular weight drugs targeting molecules mutated or overexpressed in tumours; examples include BRAF inhibitors (which block the BRAF protein) or MEK inhibitors (which block the MEK protein)

Sorafenib

An inhibitor of various tyrosine protein kinases including RAF

Selumetinib

An inhibitor of the MAPK kinase (MEK) downstream of BRAF

T‐cell

A white blood cell type, which plays a key role in immunity

Tasisulam

A small‐molecule agent that induces apoptosis through the intrinsic mitochondrial pathway

Tamoxifen

A cytostatic hormonal therapeutic agent used mainly as a treatment for oestrogen receptor positive breast cancer. Tamoxifen acts through competing with oestrogen for its receptor thus reducing oestrogen‐related effects in breast tissue such as DNA synthesis and cell proliferation

Temozolomide

An oral alkylating agent that can be used in chemotherapy regimens for various cancers such as glioblastoma multiforme

Trametinib

An inhibitor of MAPK kinase (MEK) 1 and 2 approved for use in people with V600E‐mutated metastatic melanoma

Vemurafenib

A small‐molecule inhibitor of mutated BRAF, an oncogene involved in cell survival or proliferation

Vincristine

An anti‐mitotic agent that binds tubulin thus preventing cell proliferation and triggering apoptosis

Vindesine

An anti‐mitotic agent that acts by targeting microtubules and preventing cell division thus useful as a chemotherapy drug in various cancers

Vitespen

A tumour‐derived heat shock protein that is used as an adjuvant in cancer immunotherapy

Figures and Tables -
Table 1. Glossary of terms used
Table 2. Reasons for excluding 39 studies from meta‐analysis

Study ID

Reason for exclusion from meta‐analysis

Hamid 2014

Single study investigating tasisulam

Kefford 2010

Single study investigating bosentan

Hofmann 2011

Single study comparing dacarbazine and best supportive care

Schadendorf 2006

Single study investigating dendritic cells therapy

Agarwala 2002

Single study investigating histamine with interleukin‐2

Bajetta 1985

Different polychemotherapy regimens not compared in other studies

Beretta 1976

Different polychemotherapy regimens not compared in other studies

Cocconi 1992

Different polychemotherapy regimens not compared in other studies

Dummer 2006

Different PEG‐interferon schedules tested

Flaherty 2001

Inpatient and outpatient interleukin‐2‐based regimens not compared in other studies

Glaspy 2009

Different lenalidomide schedules not compared in other studies

Jelic 2002

Different polychemotherapy regimens not compared in other studies

Keilholz 1997

Study comparing biochemotherapy versus biotherapy

Legha 1996

Study comparing alternating and sequential biochemotherapy and chemotherapy

Miller 1989

Single study investigating Indomethacine with interferon

Moon 1975

Different single‐agent chemotherapy regimens not compared in other studies

Presant 1982

Different polychemotherapy regimens not compared in other studies

Richtig 2004

Different temozolomide and interferon schedules tested

Wittes 1978

Different polychemotherapy regimens not compared in other studies

Vuoristo 2005

Different interferon‐based regimens not compared in other studies

Punt 2006

Different biochemotherapy regimens not compared in other studies

Reichle 2007

Single study investigating chemotherapy and COX‐2 inhibitor

Sparano 1993

Single study comparing interleukin‐2 with versus without interferon‐alpha

Wolchok 2010

Different ipilimumab schedules tested

Avril 2004

Single study comparing fotemustine and dacarbazine

O'Day 2011

Single study testing Intetumumab

Ranson 2007

Single study testing lomeguatrib

Hersh 2015

Single study testing nab‐paclitaxel

Bedikian 2006

Single study testing oblimersen

Bedikian 2011

Single study testing DHA‐paclitaxel

Weber 2009

Single study testing PF‐3512676

Carvajal 2014

Single study testing ramucirumab

Balch 1984

Single study testing dacarbazine and C parvum after surgery

Eigentler 2008

Single study testing vindesine after surgery

Lawson 2015

Single study testing GM‐CSF and a polypeptide vaccination after surgery

Eisen 2010

Single study testing lenalidomide

Middleton 2015

Single study testing veliparib

Testori 2008

Single study testing vetaspen

Figures and Tables -
Table 2. Reasons for excluding 39 studies from meta‐analysis
Table 3. Studies included in meta‐analysis

Comparison

Experimental (class of) drug

Study ID

Polychemotherapy versus single agent chemotherapy

Polychemotherapy

Bellett 1976

Carter 1975

Chapman 1999

Chauvergne 1982

Chiarion Sileni 2001

Costanza 1977

Luikart 1984

Ringborg 1989

Zimpfer‐Rechner 2003

Bafaloukos 2005

Glover 2003

Costanza 1972

Kogoniia 1981

Lopez 1984

Biochemotherapy versus chemotherapy

Interferon‐alpha

Bajetta 1994

Bajetta 2006

Dorval 1999

Falkson 1991

Falkson 1995

Gorbonova 2000

Kaufmann 2005

Thomson 1993

Vorobiof 1994

Young 2001

Kirkwood 1990

Daponte 2013

Falkson 1998

Danson 2003

Maio 2010

Interleukin‐2

Keilholz 2005

Sertoli 1999

Hauschild 2001

Interleukin‐2 plus interferon‐alpha

Atkins 2008

Atzpodien 2002

Eton 2002

Johnston 1998

Middleton 2007

Ridolfi 2002

Rosenberg 1999

Immune checkpoint inhibitors versus chemotherapy (or other immune checkpoint inhibitors)

Anti‐CTLA4 monoclonal antibodies

Hodi 2010

Hodi 2014

Ribas 2013

Robert 2011

Anti‐PD1 monoclonal antibodies

Ribas 2015

Robert 2015a

Weber 2015

Robert 2015b

Anti‐CTLA4 plus anti‐PD1 monoclonal antibodies

Larkin 2015

Postow 2015

Small‐molecule targeted drugs versus chemotherapy (or other small‐molecule targeted drugs)

BRAF inhibitors

Hauschild 2012

McArthur 2014

MEK inhibitors

Flaherty 2012b

Gupta 2014

Robert 2013

BRAF plus MEK inhibitors

Flaherty 2012a

Larkin 2014

Long 2015

Robert 2015

Chemotherapy with versus without other agents

Bacille Calmette‐Guérin (BCG)

Costanzi 1982

Mastrangelo 1979

Newlands 1976

Ramseur 1978

Verschraegen 1993

Veronesi 1984

Corynebacterium parvum

Clunie 1980

Gough 1978

Presant 1979

Robidoux 1982

Thatcher 1986

Kokoschka 1978

Tamoxifen

Agarwala 1999

Cocconi 1992

Rusthoven 1996

Anti‐angiogenic drugs

Cui 2013

Kim 2012

Sorafenib

Flaherty 2013

Hauschild 2009

McDermott 2008

Elesclomol

O'Day 2009

O'Day 2013

Single agent chemotherapy versus other single agent chemotherapy

Temozolomide

Chiarion‐Sileni 2011

Middleton 2000

Patel 2011

Hodi 2010a; Hodi 2014; Maio 2010; Schwartzentruber 2011a were included in a meta‐analysis of immunostimulating agents.

Figures and Tables -
Table 3. Studies included in meta‐analysis
Comparison 1. Polychemotherapy versus single agent chemotherapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Overall survival Show forest plot

6

594

Hazard Ratio (IV, Random, 95% CI)

0.99 [0.85, 1.16]

1.2 Progression‐free survival Show forest plot

5

398

Hazard Ratio (IV, Random, 95% CI)

1.07 [0.91, 1.25]

1.3 Tumour response Show forest plot

14

1885

Risk Ratio (M‐H, Random, 95% CI)

1.27 [1.02, 1.58]

1.4 Toxicity (≥ G3) Show forest plot

3

514

Risk Ratio (M‐H, Fixed, 95% CI)

1.97 [1.44, 2.71]

Figures and Tables -
Comparison 1. Polychemotherapy versus single agent chemotherapy
Comparison 2. Chemotherapy ± tamoxifen

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Overall survival Show forest plot

4

643

Hazard Ratio (IV, Random, 95% CI)

1.03 [0.80, 1.33]

2.2 Progression‐free survival Show forest plot

2

475

Hazard Ratio (IV, Random, 95% CI)

1.06 [0.93, 1.22]

2.3 Tumour response Show forest plot

4

643

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.94, 1.89]

2.4 Toxicity (≥ G3) Show forest plot

1

271

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.38, 1.28]

Figures and Tables -
Comparison 2. Chemotherapy ± tamoxifen
Comparison 3. Temozolomide versus dacarbazine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

3.1 Overall survival Show forest plot

3

1313

Hazard Ratio (IV, Random, 95% CI)

0.98 [0.85, 1.12]

3.2 Progression‐free survival Show forest plot

3

1313

Hazard Ratio (IV, Random, 95% CI)

0.87 [0.74, 1.03]

3.3 Tumour response Show forest plot

3

1313

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.85, 1.73]

3.4 Toxicity (≥ G3) Show forest plot

2

1164

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.98, 1.35]

Figures and Tables -
Comparison 3. Temozolomide versus dacarbazine
Comparison 4. Chemotherapy ± interferon‐alpha

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

4.1 Overall survival Show forest plot

11

1785

Hazard Ratio (IV, Random, 95% CI)

0.87 [0.73, 1.04]

4.2 Progression‐free survival Show forest plot

6

1272

Hazard Ratio (IV, Random, 95% CI)

0.87 [0.74, 1.01]

4.3 Tumour response Show forest plot

15

2419

Risk Ratio (M‐H, Random, 95% CI)

1.36 [1.12, 1.66]

4.4 Toxicity (≥ G3) Show forest plot

3

791

Risk Ratio (M‐H, Random, 95% CI)

1.72 [0.37, 7.95]

Figures and Tables -
Comparison 4. Chemotherapy ± interferon‐alpha
Comparison 5. Chemotherapy ± interleukin‐2

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

5.1 Overall survival Show forest plot

2

644

Hazard Ratio (IV, Random, 95% CI)

0.95 [0.82, 1.11]

5.2 Progression‐free survival Show forest plot

1

363

Hazard Ratio (IV, Random, 95% CI)

0.87 [0.70, 1.08]

5.3 Tumour response Show forest plot

3

735

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.64, 1.13]

Figures and Tables -
Comparison 5. Chemotherapy ± interleukin‐2
Comparison 6. Chemotherapy ± interferon‐alpha and interleukin‐2

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

6.1 Overall survival Show forest plot

7

1307

Hazard Ratio (IV, Random, 95% CI)

0.94 [0.84, 1.06]

6.2 Progression‐free survival Show forest plot

6

964

Hazard Ratio (IV, Random, 95% CI)

0.90 [0.83, 0.99]

6.3 Tumour response Show forest plot

7

1307

Risk Ratio (M‐H, Random, 95% CI)

1.36 [1.11, 1.67]

6.4 Toxicity (≥ G3) Show forest plot

2

657

Risk Ratio (M‐H, Random, 95% CI)

1.35 [1.14, 1.61]

Figures and Tables -
Comparison 6. Chemotherapy ± interferon‐alpha and interleukin‐2
Comparison 7. Chemotherapy ± interferon‐alpha and interleukin‐2 (first line)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

7.1 Overall survival Show forest plot

5

1118

Hazard Ratio (IV, Random, 95% CI)

0.96 [0.83, 1.10]

7.2 Progression‐free survival Show forest plot

4

775

Hazard Ratio (IV, Random, 95% CI)

0.86 [0.76, 0.99]

7.3 Tumour response Show forest plot

5

1118

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.15, 1.83]

7.4 Toxicity (≥ G3) Show forest plot

1

241

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.12, 1.87]

Figures and Tables -
Comparison 7. Chemotherapy ± interferon‐alpha and interleukin‐2 (first line)
Comparison 8. Chemotherapy ± Bacille Calmette‐Guérin (BCG)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

8.1 Overall survival Show forest plot

2

154

Hazard Ratio (IV, Random, 95% CI)

0.87 [0.61, 1.25]

8.2 Tumour response Show forest plot

6

770

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.65, 1.12]

Figures and Tables -
Comparison 8. Chemotherapy ± Bacille Calmette‐Guérin (BCG)
Comparison 9. Chemotherapy ± Corynebacterium parvum

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

9.1 Overall survival Show forest plot

4

242

Hazard Ratio (IV, Random, 95% CI)

0.95 [0.74, 1.22]

9.2 Tumour response Show forest plot

7

537

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.77, 1.38]

Figures and Tables -
Comparison 9. Chemotherapy ± Corynebacterium parvum
Comparison 10. Anti‐CTLA4 monoclonal antibodies (first line)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

10.1 Overall survival Show forest plot

2

1157

Hazard Ratio (IV, Random, 95% CI)

0.81 [0.65, 1.01]

10.2 Progression‐free survival Show forest plot

1

502

Hazard Ratio (IV, Random, 95% CI)

0.76 [0.63, 0.92]

10.3 Tumour response Show forest plot

2

1157

Risk Ratio (M‐H, Random, 95% CI)

1.28 [0.92, 1.77]

10.4 Toxicity (≥ G3) Show forest plot

2

1142

Risk Ratio (M‐H, Random, 95% CI)

1.69 [1.19, 2.42]

Figures and Tables -
Comparison 10. Anti‐CTLA4 monoclonal antibodies (first line)
Comparison 11. Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

11.1 Overall survival Show forest plot

2

784

Hazard Ratio (IV, Random, 95% CI)

0.83 [0.52, 1.33]

11.2 Progression‐free survival Show forest plot

2

785

Hazard Ratio (IV, Random, 95% CI)

1.06 [0.75, 1.51]

11.3 Tumour response Show forest plot

2

785

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.38, 1.47]

11.4 Toxicity (≥ G3) Show forest plot

2

785

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.69, 1.11]

Figures and Tables -
Comparison 11. Anti‐CTLA4 monoclonal antibodies ± other immunostimulating agents (second line)
Comparison 12. Anti‐PD1 monoclonal antibodies versus chemotherapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

12.1 Overall survival Show forest plot

1

418

Hazard Ratio (IV, Random, 95% CI)

0.42 [0.37, 0.48]

12.2 Progression‐free survival Show forest plot

2

957

Hazard Ratio (IV, Random, 95% CI)

0.49 [0.39, 0.61]

12.3 Tumour response Show forest plot

3

1367

Risk Ratio (M‐H, Random, 95% CI)

3.42 [2.38, 4.92]

12.4 Toxicity (≥ G3) Show forest plot

3

1360

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.31, 0.97]

Figures and Tables -
Comparison 12. Anti‐PD1 monoclonal antibodies versus chemotherapy
Comparison 13. Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

13.1 Overall survival Show forest plot

1

834

Hazard Ratio (IV, Random, 95% CI)

0.63 [0.60, 0.66]

13.2 Progression‐free survival Show forest plot

2

1465

Hazard Ratio (IV, Random, 95% CI)

0.54 [0.50, 0.60]

13.3 Tumour response Show forest plot

2

1465

Risk Ratio (M‐H, Random, 95% CI)

2.47 [2.01, 3.04]

13.4 Toxicity (≥ G3) Show forest plot

2

1435

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.54, 0.91]

Figures and Tables -
Comparison 13. Anti‐PD1 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies
Comparison 14. Anti‐PD1 monoclonal antibodies and anti‐CTLA4 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

14.1 Progression‐free survival Show forest plot

2

738

Hazard Ratio (IV, Random, 95% CI)

0.40 [0.35, 0.46]

14.2 Tumour response Show forest plot

2

738

Risk Ratio (M‐H, Random, 95% CI)

3.50 [2.07, 5.92]

14.3 Toxicity (≥ G3) Show forest plot

2

764

Risk Ratio (M‐H, Random, 95% CI)

1.57 [0.85, 2.92]

Figures and Tables -
Comparison 14. Anti‐PD1 monoclonal antibodies and anti‐CTLA4 monoclonal antibodies versus anti‐CTLA4 monoclonal antibodies alone
Comparison 15. Chemotherapy ± sorafenib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

15.1 Overall survival Show forest plot

3

1194

Hazard Ratio (IV, Random, 95% CI)

1.00 [0.88, 1.14]

15.2 Progression‐free survival Show forest plot

3

1194

Hazard Ratio (IV, Random, 95% CI)

0.89 [0.73, 1.09]

15.3 Tumour response Show forest plot

3

1194

Risk Ratio (M‐H, Random, 95% CI)

1.17 [0.91, 1.50]

15.4 Toxicity (≥ G3) Show forest plot

3

1194

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.93, 1.26]

Figures and Tables -
Comparison 15. Chemotherapy ± sorafenib
Comparison 16. Chemotherapy ± elesclomol

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

16.1 Overall survival Show forest plot

1

651

Hazard Ratio (IV, Random, 95% CI)

1.10 [0.92, 1.32]

16.2 Progression‐free survival Show forest plot

2

732

Hazard Ratio (IV, Random, 95% CI)

0.75 [0.50, 1.13]

16.3 Tumour response Show forest plot

2

732

Risk Ratio (M‐H, Random, 95% CI)

1.86 [0.98, 3.50]

16.4 Toxicity (≥ G3) Show forest plot

1

651

Risk Ratio (M‐H, Random, 95% CI)

1.22 [1.00, 1.50]

Figures and Tables -
Comparison 16. Chemotherapy ± elesclomol
Comparison 17. Chemotherapy ± anti‐angiogenic drugs

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

17.1 Overall survival Show forest plot

2

324

Hazard Ratio (IV, Random, 95% CI)

0.60 [0.45, 0.81]

17.2 Progression‐free survival Show forest plot

2

324

Hazard Ratio (IV, Random, 95% CI)

0.69 [0.52, 0.92]

17.3 Tumour response Show forest plot

2

324

Risk Ratio (M‐H, Random, 95% CI)

1.71 [0.96, 3.03]

17.4 Toxicity (≥ G3) Show forest plot

2

324

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.09, 5.32]

Figures and Tables -
Comparison 17. Chemotherapy ± anti‐angiogenic drugs
Comparison 18. Single agent BRAF inhibitor

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

18.1 Overall survival Show forest plot

2

925

Hazard Ratio (IV, Random, 95% CI)

0.40 [0.28, 0.57]

18.2 Progression‐free survival Show forest plot

2

925

Hazard Ratio (IV, Random, 95% CI)

0.27 [0.21, 0.34]

18.3 Tumour response Show forest plot

2

925

Risk Ratio (M‐H, Random, 95% CI)

6.78 [4.84, 9.49]

18.4 Toxicity (≥ G3) Show forest plot

2

925

Risk Ratio (M‐H, Random, 95% CI)

1.27 [0.48, 3.33]

Figures and Tables -
Comparison 18. Single agent BRAF inhibitor
Comparison 19. Single agent MEK inhibitor

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

19.1 Overall survival Show forest plot

3

496

Hazard Ratio (IV, Random, 95% CI)

0.85 [0.58, 1.25]

19.2 Progression‐free survival Show forest plot

3

496

Hazard Ratio (IV, Random, 95% CI)

0.58 [0.42, 0.80]

19.3 Tumour response Show forest plot

3

496

Risk Ratio (M‐H, Random, 95% CI)

2.01 [1.35, 2.99]

19.4 Toxicity (≥ G3) Show forest plot

1

91

Risk Ratio (M‐H, Fixed, 95% CI)

1.61 [1.08, 2.41]

Figures and Tables -
Comparison 19. Single agent MEK inhibitor
Comparison 20. Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

20.1 Overall survival Show forest plot

4

1784

Hazard Ratio (IV, Random, 95% CI)

0.70 [0.59, 0.82]

20.2 Progression‐free survival Show forest plot

4

1784

Hazard Ratio (IV, Random, 95% CI)

0.56 [0.44, 0.71]

20.3 Tumour response Show forest plot

4

1784

Risk Ratio (M‐H, Random, 95% CI)

1.32 [1.20, 1.46]

20.4 Toxicity (≥ G3) Show forest plot

4

1774

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.85, 1.20]

Figures and Tables -
Comparison 20. Combination of BRAF and MEK inhibitors versus single agent BRAF inhibitor
Comparison 21. Immunostimulating agents

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

21.1 Overall survival Show forest plot

4

1458

Hazard Ratio (IV, Random, 95% CI)

0.82 [0.67, 0.99]

21.2 Progression‐free survival Show forest plot

4

1458

Hazard Ratio (IV, Random, 95% CI)

0.92 [0.74, 1.14]

21.3 Tumour response Show forest plot

4

1451

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.60, 2.50]

21.4 Toxicity (≥ G3) Show forest plot

4

1458

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.77, 1.08]

Figures and Tables -
Comparison 21. Immunostimulating agents