Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Analysis
  • Published:

Additivity predicts the efficacy of most approved combination therapies for advanced cancer

Abstract

Most advanced cancers are treated with drug combinations. Rational design aims to identify synergistic combinations, but existing synergy metrics apply to preclinical, not clinical data. Here we propose a model of drug additivity for progression-free survival (PFS) to assess whether clinical efficacies of approved drug combinations are additive or synergistic. This model includes patient-to-patient variability in best single-drug response plus the weaker drug per patient. Among US Food and Drug Administration approvals of drug combinations for advanced cancers (1995–2020), 95% exhibited additive or less than additive effects on PFS times. Among positive or negative phase 3 trials published between 2014–2018, every combination that improved PFS was expected to succeed by additivity (100% sensitivity) and most failures were expected to fail (78% specificity). This study shows synergy is neither a necessary nor common property of clinically effective drug combinations. The predictable efficacy of approved combinations suggests that additivity can be a design principle for combination therapies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Concept of the HSA and additivity models.
Fig. 2: Clinical trial selection process.
Fig. 3: Most drug combinations approved for advanced cancers are as effective as expected by either the highest single agent or additivity model.
Fig. 4: Progression-free survival of combination therapies compared to predictions of HSA and additivity.
Fig. 5: Additivity is a more accurate model than HSA.
Fig. 6: Additivity can predict the success and failure of phase 3 trials.
Fig. 7: HSA and additivity models make similar predictions when monotherapy efficacies are highly variable.
Fig. 8: Some combinations with shared characteristics are associated with lesser efficacy than expected by additivity.

Similar content being viewed by others

Data availability

All clinical trials included in the study are listed in Supplementary Tables 1 and 3. References to the clinical trials can be found in Supplementary References. PDX drug response data were obtained from the supplementary tables of Gao et al.44. Reprocessed CTRPv2 cell line drug responses were downloaded from osf.io/sym6h/ (ref. 33). Source Data for Figs. 3 and 4, including digitally traced Kaplan–Meier survival curves, imputed patient event times and predicted PFS distributions under the HSA and additivity models can be retrieved from Figshare at https://doi.org/10.6084/m9.figshare.22229677. Source data are provided with this paper.

Code availability

Source code can be retrieved from github.com/palmerlabunc/clinical-additivity.

References

  1. Damon, L. E. & Cadman, E. C. Advances in rational combination chemotherapy. Cancer Invest. 4, 421–444 (1986).

    CAS  PubMed  Google Scholar 

  2. Verweij, J., Disis, M. L. & Cannistra, S. A. Phase I studies of drug combinations. J. Clin. Oncol. 28, 4545–4546 (2010).

    PubMed  Google Scholar 

  3. Paller, C. J. et al. Design of phase I combination trials: recommendations of the Clinical Trial Design Task Force of the NCI Investigational Drug Steering Committee. Clin. Cancer Res. 20, 4210–4217 (2014).

    PubMed  PubMed Central  Google Scholar 

  4. DeVita, V. T. & Schein, P. S. The use of drugs in combination for the treatment of cancer. N. Engl. J. Med. 288, 998–1006 (1973).

    CAS  PubMed  Google Scholar 

  5. Foucquier, J. & Guedj, M. Analysis of drug combinations: current methodological landscape. Pharmacol. Res. Perspect. 3, e00149 (2015).

    PubMed  PubMed Central  Google Scholar 

  6. Anon. Rationalizing combination therapies. Nat. Med. 23, 1113–1113 (2017).

  7. Boshuizen, J. & Peeper, D. S. Rational cancer treatment combinations: an urgent clinical need. Mol. Cell 78, 1002–1018 (2020).

    CAS  PubMed  Google Scholar 

  8. Ocana, A., Amir, E., Yeung, C., Seruga, B. & Tannock, I. F. How valid are claims for synergy in published clinical studies? Ann. Oncol. 23, 2161–2166 (2012).

    CAS  PubMed  Google Scholar 

  9. Wittes, R. E. & Goldin, A. Unresolved issues in combination chemotherapy. Cancer Treat. Rep. 70, 105–125 (1986).

    CAS  PubMed  Google Scholar 

  10. Berenbaum, M. C. What is synergy? Pharmacol. Rev. 41, 93–141 (1989).

    CAS  PubMed  Google Scholar 

  11. Frei, W. Versuche über Kombination von Desinfektionsmitteln. Z. Fuer Hyg. Infekt. 75, 433 (1913).

    Google Scholar 

  12. Loewe, S. & Muischnek, H. Über Kombinationswirkungen. Naunyn-Schmiedebergs Arch. Für Exp. Pathol. Pharmakol. 114, 313–326 (1926).

    CAS  Google Scholar 

  13. Bliss, C. I. The toxicity of poisons applied jointly. Ann. Appl. Biol. 26, 585–615 (1939).

    CAS  Google Scholar 

  14. Meyer, C. T. et al. Quantifying drug combination synergy along potency and efficacy axes. Cell Syst. 8, 97–108 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Wooten, D. J., Meyer, C. T., Lubbock, A. L. R., Quaranta, V. & Lopez, C. F. MuSyC is a consensus framework that unifies multi-drug synergy metrics for combinatorial drug discovery. Nat. Commun. 12, 4607 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Frei, E. et al. Studies of sequential and combination antimetabolite therapy in acute leukemia: 6-mercaptopurine and methotrexate. Blood 18, 431–454 (1961).

    Google Scholar 

  17. Pomeroy, A. E., Schmidt, E. V., Sorger, P. K. & Palmer, A. C. Drug independence and the curability of cancer by combination chemotherapy. Trends Cancer 8, 915–929 (2022).

  18. Palmer, A. C. & Sorger, P. K. Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy. Cell 171, 1678–1691 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Palmer, A. C., Izar, B., Hwangbo, H. & Sorger, P. K. Predictable clinical benefits without evidence of synergy in trials of combination therapies with immune-checkpoint inhibitors. Clin. Cancer Res. 28, 368–377 (2022).

    PubMed  PubMed Central  Google Scholar 

  20. Plana, D., Palmer, A. C. & Sorger, P. K. Independent drug action in combination therapy: implications for precision oncology. Cancer Discov. 12, 606–624 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Laplace, P.-S. Théorie analytique des probabilités. (Courcier, 1812).

  22. Plana, D., Fell, G., Alexander, B. M., Palmer, A. C. & Sorger, P. K. Cancer patient survival can be parametrized to improve trial precision and reveal time-dependent therapeutic effects. Nat. Commun. 13, 873 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Carvajal, R. D. et al. Selumetinib in combination with dacarbazine in patients with metastatic uveal melanoma: a phase III, multicenter, randomized trial (SUMIT). J. Clin. Oncol. 36, 1232–1239 (2018).

    CAS  PubMed  Google Scholar 

  24. Tournigand, C. et al. Bevacizumab with or without erlotinib as maintenance therapy in patients with metastatic colorectal cancer (GERCOR DREAM; OPTIMOX3): a randomised, open-label, phase 3 trial. Lancet Oncol. 16, 1493–1505 (2015).

    CAS  PubMed  Google Scholar 

  25. Finn, R. S. et al. Palbociclib and letrozole in advanced breast cancer. N. Engl. J. Med. 375, 1925–1936 (2016).

    CAS  PubMed  Google Scholar 

  26. Dimopoulos, M. A. et al. Daratumumab, lenalidomide, and dexamethasone for multiple myeloma. N. Engl. J. Med. 375, 1319–1331 (2016).

    CAS  PubMed  Google Scholar 

  27. Plesner, T. et al. Phase 1/2 study of daratumumab, lenalidomide, and dexamethasone for relapsed multiple myeloma. Blood 128, 1821–1828 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Finn, R. S. et al. PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res. 11, R77 (2009).

    PubMed  PubMed Central  Google Scholar 

  29. van der Veer, M. S. et al. Towards effective immunotherapy of myeloma: enhanced elimination of myeloma cells by combination of lenalidomide with the human CD38 monoclonal antibody daratumumab. Haematologica 96, 284–290 (2011).

    PubMed  Google Scholar 

  30. Llombart-Cussac, A. et al. Fulvestrant-palbociclib vs letrozole-palbociclib as initial therapy for endocrine-sensitive, hormone receptor-positive, ERBB2-negative advanced breast cancer: a randomized clinical trial. JAMA Oncol. 7, 1791–1799 (2021).

    PubMed  Google Scholar 

  31. Lonial, S. et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet 387, 1551–1560 (2016).

    CAS  PubMed  Google Scholar 

  32. Hwang, T. J. et al. Failure of investigational drugs in late-stage clinical development and publication of trial results. JAMA Intern. Med. 176, 1826–1833 (2016).

    PubMed  Google Scholar 

  33. Ling, A. & Huang, R. S. Computationally predicting clinical drug combination efficacy with cancer cell line screens and independent drug action. Nat. Commun. 11, 5848 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Jaaks, P. et al. Effective drug combinations in breast, colon and pancreatic cancer cells. Nature 603, 166–173 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Prahallad, A. et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483, 100–103 (2012).

    CAS  PubMed  Google Scholar 

  36. Vellano, C. P. et al. Androgen receptor blockade promotes response to BRAF/MEK-targeted therapy. Nature 606, 797–803 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. AstraZeneca-Sanger Drug Combination DREAM Consortium et al. Community assessment to advance computational prediction of cancer drug combinations in a pharmacogenomic screen. Nat. Commun. 10, 2674 (2019).

    Google Scholar 

  38. Palmer, A. C., Chidley, C. & Sorger, P. K. A curative combination cancer therapy achieves high fractional cell killing through low cross-resistance and drug additivity. eLife 8, e50036 (2019).

  39. Skipper, H. E., Schabel, F. M. & Wilcox, W. S. Experimental evaluation of potential anticancer agents. XIII. On the criteria and kinetics associated with curability of experimental leukemia. Cancer Chemother. Rep. 35, 1–111 (1964).

    CAS  PubMed  Google Scholar 

  40. Koch, R. et al. Biomarker-driven strategy for MCL1 inhibition in T-cell lymphomas. Blood 133, 566–575 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Jonker, D. J. et al. Cetuximab for the treatment of colorectal cancer. N. Engl. J. Med. 357, 2040–2048 (2007).

    CAS  PubMed  Google Scholar 

  42. Li, J. et al. Regorafenib plus best supportive care versus placebo plus best supportive care in Asian patients with previously treated metastatic colorectal cancer (CONCUR): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 16, 619–629 (2015).

    CAS  PubMed  Google Scholar 

  43. Van Cutsem, E. et al. Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. J. Clin. Oncol. 25, 1658–1664 (2007).

    PubMed  Google Scholar 

  44. Fuchs, C. S. et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 383, 31–39 (2014).

    CAS  PubMed  Google Scholar 

  45. Kang, Y.-K. et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 390, 2461–2471 (2017).

    CAS  PubMed  Google Scholar 

  46. Ranson, M. Randomized trial of paclitaxel plus supportive care versus supportive care for patients with advanced non-small-cell lung cancer. J. Natl. Cancer Inst. 92, 1074–1080 (2000).

    CAS  PubMed  Google Scholar 

  47. Shepherd, F. A. et al. Erlotinib in previously treated non–small-cell lung cancer. N. Engl. J. Med. 353, 123–132 (2005).

  48. Jassem, J. et al. Phase III trial of pemetrexed plus best supportive care compared with best supportive care in previously treated patients with advanced malignant pleural mesothelioma. J. Clin. Oncol. 26, 1698–1704 (2008).

    CAS  PubMed  Google Scholar 

  49. Motzer, R. J. et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. 372, 8 (2008).

  50. Seashore-Ludlow, B. et al. Harnessing connectivity in a large-scale small-molecule sensitivity dataset. Cancer Discov. 5, 1210–1223 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Gao, H. et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat. Med. 21, 1318–1325 (2015).

    CAS  PubMed  Google Scholar 

  52. Rees, M. G. et al. Correlating chemical sensitivity and basal gene expression reveals mechanism of action. Nat. Chem. Biol. 12, 109–116 (2016).

    CAS  PubMed  Google Scholar 

  53. Cox, D. R. Regression models and life-tables. J. R. Stat. Soc. 32, 187–220 (1972).

    Google Scholar 

  54. Guyot, P., Ades, A., Ouwens, M. J. & Welton, N. J. Enhanced secondary analysis of survival data: reconstructing the data from published Kaplan–Meier survival curves. BMC Med. Res. Methodol. 12, 9 (2012).

    PubMed  PubMed Central  Google Scholar 

  55. Alexander, B. M., Schoenfeld, J. D. & Trippa, L. Hazards of hazard ratios — deviations from model assumptions in immunotherapy. N. Engl. J. Med. 378, 1158–1159 (2018).

    PubMed  Google Scholar 

  56. Vogel, C. L. et al. First-line herceptin monotherapy in metastatic breast cancer. Oncology 61, 37–42 (2001).

    CAS  PubMed  Google Scholar 

  57. Miao, E., Seetharamu, N., Sullivan, K., Eng, S. & Lee, C.-S. Impact of tyrosine kinase inhibitor starting dose on outcomes in patients with non-small cell lung cancer. J. Pharm. Pract. 34, 11–16 (2021).

    PubMed  Google Scholar 

  58. Cortese, M. J. et al. A multi-center analysis of the impact of DA-EPOCH-R dose-adjustment on clinical outcomes of patients with double/triple-hit lymphoma. Leuk. Lymphoma 64, 107–118 (2023).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the investigators and patients who participated in the clinical trials analyzed in this work. We thank E.V. Schmidt, C. Chen, D.M. Weinstock, S. Hughes, N.E. Sharpless and P.K. Sorger for discussions. We thank I. Zhou for assistance in data collection. H.H. is supported by the Korean Government Scholarship for Study Overseas, S.C.P. is supported by NIGMS grant T32-GM135095 and D.P. is supported by NIGMS grant T32-GM007753 and F30-CA260780. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. The authors received no specific funding for this work.

Author information

Authors and Affiliations

Authors

Contributions

H.H., D.P. and A.C.P. conceived the study. H.H. implemented the method and performed data analysis. H.H., S.C.P. and A.D. collected clinical trial data. H.H. and A.C.P. wrote the manuscript. All authors reviewed the manuscript and approved the final version.

Corresponding author

Correspondence to Adam C. Palmer.

Ethics declarations

Competing interests

A.C.P. has received consulting fees from AstraZeneca, Kymera and Merck, and research funding from Prelude Therapeutics. A.C.P. declares that these interests had no role in study design, analysis or decision to publish. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor:Alexia-Ileana Zaromytidou.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Addition of PFS times is consistent with the Bliss Independence model.

Our model of clinical drug additivity is formally defined as the addition of PFS times. This null hypothesis does not depend on assumptions about underlying tumor growth kinetics and cytotoxic mechanisms. However, mathematically, the addition of PFS times conveniently corresponds to the Bliss independence model under simplifying assumptions. Bliss independence is a common null hypothesis used to classify drug synergy or antagonism in cell-based experiments, which states that P(a + b) = P(a)P(b), where P(x) is the fraction of cells surviving toxin x. This corresponds to the addition of cytotoxic effects on a logarithmic scale. (A) When drug A kills 90% of cancer cells and drug B kills 99% of cancer cells, it will take PFSA and PFSB respectively to observe disease progression, assuming exponential growth of the surviving cancer cell population. If it takes PFSunt for an untreated tumor to progress, drug A and drug B extend PFS by tA = PFSA-PFSunt and tB = PFSB-PFSunt respectively. (B) When drug A and B are additive, A + B will produce 99.9% kill by Bliss Independence. PFS will be extended by tA + tB beyond that of an untreated patient. A mathematical derivation of the concordance between Bliss Independence and addition of PFS times, under simplifying assumptions can be found in Supplementary Notes 1.

Extended Data Fig. 2 Frei’s model of independent drug action is a model of highest single agent.

Bliss’ model and Frei’s model are applications of the addition rule of probability at different biological scales, resulting in different biological interpretations. Bliss’ model was developed to analyze fraction of organisms killed by multiple toxins. Bliss’ model has been applied in cancer research to analyze fraction of tumor cells killed by multiple therapies. In the context of a population of cancer cells, Bliss’ model implies that more cancer cells will be killed by drug A + B than either drug A or B alone. This results in a patient’s response being better than Highest Single Agent. Frei applied the same fundamental principle of probability to remission rates in a population of patients treated with multiple therapies. In the context of a population of patients, Frei’s model implies that more patients will respond to drug A + B then either drug A or B alone. However, this does not require or imply that an individual patient’s response to drug A + B is better than drug A or B alone. The improved response rate described by Frei’s model occurs even if each individual patient’s response is equal to that of the Highest Single Agent.

Extended Data Fig. 3 When patients receive no active therapy for advanced cancer, disease progression is commonly observed at the first scheduled scan.

Progression-Free Survival distributions are shown for patients receiving ‘Best Supportive Care’ (BSC) for (A) advanced colorectal cancer41; (B) BSC plus placebo for metastatic colorectal cancer42; (C) BSC for metastatic colorectal cancer43; (D) BSC plus placebo for advanced gastric or gastro-esophageal junction (GEJ) cancer44; (E) placebo for advanced GEJ cancer45; (F) BSC for advanced non-small-cell lung cancer (NSCLC)46; (G) Placebo for stage IIIB or IV NSCLC47, (H) BSC for advanced malignant pleural mesothelioma48; (I) BSC plus placebo for metastatic renal cell carcinoma49. Red vertical lines indicate the time of first tumor evaluation by radiological scans.

Source data

Extended Data Fig. 4 PFS of combination therapies and their constituent therapies observed in clinical trials compared with predictions of HSA and additivity.

Combinations from the main analysis, two additional combinations that did not strictly satisfy the inclusion criteria, and biomarker subgroups are included. All combination naming follows ‘experimental drug plus control drugs’ format. The clinical trial publications of the combination therapy are cited below the combination names. Panel numbers from Fig. 3 are annotated. BC, breast cancer; Dara., daratumumab; CRC, colorectal cancer; 5FU, 5-fluorouracil; CLL, chronic lymphocytic leukemia; MM, multiple myeloma; Dex., dexamethasone; Bev., bevacizumab; Pembro., pembrolizumab; Atezo., atezolizumab; Chemo., chemotherapy; ITT, intention to treat population; CPS, PD-L1 combined proportion score; LV, leucovorin; TPS, PD-L1 tumor proportion score.

Extended Data Fig. 5 Addition of PFS is a robust clinical definition of drug additivity.

(A) Combinations with reduced doses are mostly inadequate to assess drug interactions. The ideal set of measurements to assess effect addition is when two drugs are combined at full dose (left). The combination therapies analyzed here were administered at least 75% of the monotherapy doses. To test Loewe’s dose-additivity model, at least one measurement of combination activity should be along the equipotent line (right). In the combination therapies we have declined to analyze, the available data cannot determine the shape of the isobole, and therefore cannot test whether a combination is Loewe-synergistic. A typical case that we have declined to analyze is when one drug in the combination is administered at a lower dose than was used in its monotherapy studies. If a combination is administered with one agent at lower dose, and its efficacy is consistent with drug additivity assuming full dose, then two competing hypotheses could explain the observation: 1) The single-agent’s efficacy is not diminished with lowered dose, and the effect of the combination is additive. 2) The combination is synergistic (by Loewe’s model, that is, potency is enhanced). These two explanations are indistinguishable without data on the dose-response relationship in humans. Clinically administered doses are often set at the maximum tolerated dose identified in phase I trials, and for many cancer therapies, clinical studies have observed that lowering a drug’s dose as much as two-fold does not diminish its clinical efficacy56,57,58. For this reason, hypothesis 1 is a possibility unless one has data on single-agent efficacy at the relevant dose. (B) Multiplying Hazard Ratios results in unrealistic predictions with survival times that are far longer than arithmetic addition. Exponential survival functions were simulated with rate parameters of λ = 1, 0.2, and 0.25, for placebo, drug A, and drug B, respectively. The effects of an ‘additive’ combination were simulated by the addition of PFS times (the model proposed in this study, using correlation ρ = 0.3 and scan interval 1.5 months), or by multiplying the hazard ratios of single drugs relative to placebo, which results in a rate of λ = 0.05. Median PFS time from multiplying hazard ratios is four times as large as the most active single drug (14 months vs. 3.5 months), making it much more than the addition of single-drug PFS times. (C) Additivity is less affected by the correlation of monotherapy responses compared to HSA. Combination effects under the HSA model (top left) and the additivity model (top right) vary modestly by the level of correlation (rho) between the monotherapy drug responses. Here, examples of monotherapy PFS distributions were constructed from lognormal survival functions, with an experimental drug (μ = 1, σ = 1, natural log base) and control drug (μ = 1.2, σ = 1, natural log base). Either the HSA model (bottom left) or the additivity model (bottom right) was compared to the control drug by the Cox proportional hazard model. 500 samples were used for each simulated arms to calculate the hazard ratio. Under the HSA model, combination efficacy was modestly improved by lower correlation, but under the additivity model, correlation has negligible influence. Centers of the error bars indicate hazard ratio between the model and the control arm. Error bars indicate 95% confidence intervals. (D) The additivity model predicts near zero benefit when combining placebo or best supportive care (that is, wholly ineffective anti-cancer) drugs. Since wholly ineffective drugs will have the same effect (that is, no therapeutic benefit) on patients, the correlation in response should be ρ = 1. We have also included hypothetical cases where the correlation is lower to demonstrate that the level of correlation has a small impact on the predictions in this scenario.

Source data

Extended Data Fig. 6 Correlations between drug responses from preclinical models were used to compute expected PFS distributions.

(A) Distributions of pairwise Spearman correlations between anti-cancer agents from CTRPv2. (Mean of all drug pairs, 0.30; targeted therapies, 0.28; cytotoxic chemotherapy – targeted therapy pairs, 0.31; cytotoxic chemotherapies, 0.52) (B) Correlation between colorectal cancer PDXs’ best average response from 5-fluorouracil (5FU) and cetuximab. Spearman correlations were measured in pan-cancer cell lines for (C) docetaxel and 5FU (substitution for capecitabine), (D) lapatinib and 5FU (substitution for capecitabine), (E) topotecan (substitution for irinotecan) and 5FU, (F) oxaliplatin (substitution for cisplatin) and gemcitabine, and (G) oxaliplatin (substitution for cisplatin) and methotrexate (substitution for pemetrexed). (H) Correlation between trametinib and dabrafenib in melanoma cell lines. Sample sizes (n) are numbers of unique cancer cell cultures (not replicates of one culture) in panels C-H, and number of unique PDXs (not replicates of one PDX) in panel B.

Source data

Supplementary information

Supplementary Information

Supplementary Note 1 and Supplementary References

Reporting Summary

Supplementary Table

Supplementary Tables 1–3

Source data

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 7

Statistical Source Data.

Source Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 3

Digitized Kaplan–Meier curve. Original data sources are listed in reference. Data are also on Figshare.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 6

Statistical Source Data. Original data source is from CTRPv2 and Gao et al.51.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hwangbo, H., Patterson, S.C., Dai, A. et al. Additivity predicts the efficacy of most approved combination therapies for advanced cancer. Nat Cancer 4, 1693–1704 (2023). https://doi.org/10.1038/s43018-023-00667-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00667-z

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer