Additivity predicts the efficacy of most approved combination therapies for advanced cancer =========================================================================================== * Haeun Hwangbo * Sarah Patterson * Andy Dai * Deborah Plana * Adam C. Palmer ## ABSTRACT Most advanced cancers are treated with drug combinations. Rational design aims to identify synergistic drug interactions to produce superior treatments. However, metrics of drug interaction (i.e. synergy, additivity, antagonism) are applicable to pre-clinical experiments, and there has been no established method to quantify synergy versus additivity in clinical settings. Here, we propose and apply a model of drug additivity for progression-free survival data to assess if the clinical efficacies of approved drug combinations are more than, or equal to, the sum of their parts. Among FDA-approvals for advanced cancers between 1995-2020, we identified 37 combinations across 13 cancer types where monotherapies and combination therapy could be compared. 95% of combination therapies exhibited progression-free survival times that were additive, or less than additive. The predictable efficacy of many of the best drug combinations established over the past 25 years suggests that additivity can be used as a design principle for novel drug combinations and clinical trials. ## INTRODUCTION The most effective known treatments for many types of cancer involve combination therapy. Because of the vast number of possible combinations, prioritizing drug combinations that are most likely to succeed in the clinic is a critical need1–3. Historically, combinations were designed empirically based on single-agent activity and non-overlapping toxicity4. In recent decades, understanding of oncogenic mechanisms has favored the design of drug combinations based on molecular reasoning. Rational combination design often aims to identify synergistic drug interactions, whereby two or more drugs enhance each other’s efficacy to produce a more than additive anti-tumor effect5–7. Accordingly, the National Cancer Institute’s Clinical Trial Design Taskforce recommends that phase I trials of novel combination therapies have a rationale for pharmacological or biological interaction3. Many clinical trials of novel combinations are motivated by synergy in cell lines or mice, with the hope that synergy will also occur in humans (**Supplementary Table S1**). Most novel combinations are not ultimately approved, and pre-clinical synergy is not significantly associated with clinical success8. Some combination therapies are found to be superior to monotherapy (or *N*+1 drugs are superior to *N*), yet a positive clinical trial does not confirm synergistic interaction, as superior efficacy could result from additive, more than additive, or less than additive effects. This is not a semantic difference: it fundamentally concerns whether a mechanism of positive drug interaction is needed to develop clinically effective combination therapies. Yet, it is rarely investigated whether synergy is achieved in clinical settings; in fact, a calculation to distinguish additive from synergistic clinical efficacy has not been defined. In pre-clinical experiments, pharmacological interactions - antagonism, additivity, and synergism - are rigorously defined and quantifiable. Additivity refers to the expected effect of combining non-interacting drugs, and synergy or antagonism refers to stronger or weaker effects, respectively9. Additivity can be defined by the potency of a dose-response10, by a statistical calculation of effect size11, or by models that synthesize both metrics12,13. These pre-clinical definitions do not readily apply to patient data. Without a clinical definition of additivity, synergy has been often stated based on “*higher response rates than one might vaguely expect from the single agents… without realizing that the term should be a quantitative description of drug effect*”14. This deficiency has persisted since its recognition in 1986. Previous models of clinical trials have calculated how multiple drugs increase the chance of response to at least one drug, which is more conservative than additivity. Specifically, in 1961 Frei and colleagues described a law to estimate the response rate to combination therapy (Rab) from monotherapy response rates (Ra, Rb): Rab = 1 - (1-Ra)(1-Rb)15. This calculates the chance that at least one drug induces a response and is therefore a model of ‘*highest single agent*’ (HSA) that does not account for multiple drug effects ‘adding up’ in individual patients. We previously adapted Frei’s law to progression-free survival (PFS), to compute PFS distributions expected from an increased chance of single-drug response16. Since its publication, this model of HSA has been validated by many phase III trials of combination immunotherapy, with ten FDA approvals resulting from trials where observed PFS distributions are statistically indistinguishable from the model prediction17. We observed that some combination therapies surpass the HSA prediction, but this model does not distinguish between superiority arising from additivity or synergy16. Here we propose a model for clinical drug additivity and apply it to PFS results from all combination therapy trials in advanced cancers that led to FDA approval between 1995 and 2020, for which matched combination and monotherapy data are available. This model provides a null hypothesis against which to test for clinical synergy in patient populations. We find that the model of additivity accurately matches the clinical efficacy of most approved drug combinations; only 2 of 37 (5%) combination therapies analyzed were significantly more than additive (at P < 0.05). ‘Synergy’ is often misleadingly interchanged with effectiveness, but these findings of additivity do not dispute established evidence of clinical efficacy. They instead show that most approved drug combinations owe their efficacy to having effective ingredients. This work elucidates the mechanism of clinically effective drug combinations in oncology, providing a design principle for future pre-clinical drug development and clinical trial design. ## RESULTS ### Definition of clinical additivity We defined clinical drug additivity as the sum of PFS benefits in individual patients. This expands on our published HSA model16,17 which estimates a PFS distribution for a drug combination by simulating a cohort of virtual patients whose single drug responses are sampled from clinically observed single-drug PFS distributions and assigning to each virtual patient their maximum single drug response time. To model additivity, the PFS times of individual drugs (PFSA, PFSB) are added, with a correction to avoid double-counting the time to observe progression in the absence of effective therapy (PFSuntreated); thus, PFSAB = PFSA + PFSB – PFSuntreated (see Methods). This correction accounts for the fact that progression is observable at scheduled radiological scans18, and in ‘placebo only’ or ‘best supportive care’ arms in advanced cancers, the majority of patients exhibit disease progression by their first scheduled scan (**Supplementary Figure S1**). ### Study population We searched FDA approvals to obtain a comprehensive list of combination therapies approved for advanced cancers between 1995 and 2020 (**Figure 1**). 84 drug combinations met the inclusion criteria. Assessing additive versus non-additive efficacy requires Kaplan-Meier curves of PFS for the control (standard-of-care) arm, the experimental single-agent arm, and the combination arm. When all three arms were not present in the same trial, we searched for clinical trials that studied the missing arm in a comparable cohort with identical or similar dosage (**Supplementary Figure S2**). This yielded a total of 37 combination therapies across 13 cancer types, with data from 24,723 patients. ![Figure 1.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2022/10/22/2022.10.21.22281013/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2022/10/22/2022.10.21.22281013/F1) Figure 1. Pipeline of clinical trial selection process. (A) PRISMA diagram. n, number of combination therapies. ### Additivity explains the clinical efficacy of most combination therapies Each combination therapy’s PFS distribution was compared with the HSA model and the additivity model, as calculated from clinically observed monotherapy PFS (**Figure 2, 3, Supplementary Figure S3**). We consider a trial result to be consistent with a model if there is no statistically significant difference between observed and expected PFS by Cox Proportional Hazards (significance level of 0.05), which is the standard method for comparing survival distributions in trials. Note that when comparing observed and expected efficacy, a hazard ratio (HR) of 1 indicates a predictable magnitude of benefit; it does not mean ‘no benefit’. 2 out of 37 combinations (5%) were significantly superior to the additivity model, i.e., synergistic (**Figure 3A**). 25 of 37 combinations (68%) were statistically indistinguishable from the additivity model. Of these, 10 combinations were consistent with both HSA and additivity (**Figure 3B**); these similarities are discussed below. 9 combinations (24%) were significantly inferior to additivity but consistent with HSA, and one combination (3%) was inferior to HSA (**Figure 3C**). ![Figure 2.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2022/10/22/2022.10.21.22281013/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2022/10/22/2022.10.21.22281013/F2) Figure 2. Most drug combinations approved for advanced cancers are as effective as expected by either the Highest Single Agent (HSA) or additivity model. Observed and expected PFS distributions were compared by the Cox proportional hazard test, for models of HSA and additivity. Hazard ratio of 1 means observed and expected PFS are equal. Error bars indicate 95% confidence intervals. Clinical trials are from refs.19,20,45–81. BC, Breast Cancer; MM, Multiple Myeloma; CRC, Colorectal Cancer; CLL, Chronic Lymphocytic Leukemia; OC, Ovarian Cancer; TNBC, Triple-Negative Breast Cancer; ES-SCLC, Extensive-Stage Small Cell Lung Cancer; NSCLC, Non-Small Cell Lung Cancer; Bev., Bevacizumab; Atezo., Atezolizumab; 5-FU, 5-Fluorouracil; LV, Leucovorin; Dex., Dexamethasone; CPS, PD-L1 combined proportion score; TPS, PD-L1 tumor proportion score. ![Figure 3.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2022/10/22/2022.10.21.22281013/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2022/10/22/2022.10.21.22281013/F3) Figure 3. Progression-free survival (PFS) of combination therapies compared with predictions of HSA and additivity. (A) Combination therapies that are significantly more than additive; (B) statistically indistinguishable from additivity; and (C) significantly less than additive. Combinations in (C) are consistent with the HSA model except one marked by a green box. Panel numbers correspond to numbers in Figure 2. Overall, drug additivity was the most accurate model and 100% of these approved regimens could have been predicted to succeed by the additivity model. We assessed the goodness of fit using coefficient of determination (R2) between the expected and observed PFS curves (**Figure 4A, B**). R2 across all 37 combinations were 0.82 and 0.90 for HSA and additivity, respectively. We further assessed goodness of fit as the mean signed difference, which was on average +3.7% for HSA (observed effect was better than model) and -1.7% for additivity (observed effect worse than model) (**Figure 4C**). We next investigated whether HSA or additivity models could have predicted the success of these trials based only on single-agent data. For each trial, a hazard ratio was calculated for predicted combination therapy PFS versus the trial’s control arm (Cox Proportional Hazards model; ɑ = 0.05). All approved combinations were predicted to significantly improve PFS based on the additivity model (**Supplementary Figure S4**), whereas the HSA model was more conservative and only predicted the success of 73% of combinations. We conclude that the additivity model better explains the clinical efficacy of combination therapies and has the potential to prospectively estimate hazard ratios from single drug data to inform clinical trial design. ![Figure 4.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2022/10/22/2022.10.21.22281013/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2022/10/22/2022.10.21.22281013/F4) Figure 4. Additivity is a more accurate model than HSA but overestimates the activity of some drug combinations. (A) Observed versus expected PFS under models of HSA and additivity. Each line represents a different combination therapy’s PFS over time. (B) Histogram of goodness of fit (R2) between observed versus expected from HSA (left) and additivity (right). R2 below zero is shown at zero. Gray bars indicate combinations that are not consistent with the model. (C) Histogram of mean signed difference between observed and expected PFS distributions from HSA (left) and additivity (right). Positive values indicate observed PFS better than expected PFS. (D) Combination therapies were classified by inclusion of an immune checkpoint inhibitor, inclusion of an anti-angiogenesis agent, monotherapy approval of the experimental drug, and the net benefit of the combination (hazard ratio between combination arm and control arm above or below median of all trials analyzed, 0.61). Each group was tested for deviation from additivity (HRadditivity ≠ 1) by the Wilcoxon signed-rank test. The center of the boxplots indicates median and the upper and lower bounds of the boxes indicate first and third interquartile range. Whiskers extends to 1.5 interquartile range. NS, not significant. ### Synergistic combinations Two combination therapies exhibited significantly ‘more than additive’ PFS, being palbociclib plus letrozole in hormone receptor-positive / HER2-negative (HR+/HER2-) advanced breast cancer19 (P = 0.023), and daratumumab plus lenalidomide plus dexamethasone in relapsed or refractory multiple myeloma20 (P = 0.0001) (**Figure 3A**). Three other combinations of CDK4/6 inhibition plus endocrine therapy for HR+/HER2-advanced breast cancer, similar to palbociclib and letrozole, were consistent with additivity. Similarly, daratumumab plus lenalidomide plus dexamethasone was the only one among five daratumumab combinations for multiple myeloma that exhibited synergy. Data on daratumumab monotherapy has the substantial limitation of coming from patients with at least three prior lines of therapy21, whereas the combination therapy trial enrolled patients with a median of one previous line of therapy. Therefore, these data suggest two competing explanations: daratumumab is synergistic with lenalidomide plus dexamethasone (but not with bortezomib, carfilzomib, or pomalidomide plus dexamethasone), or daratumumab’s efficacy at the second line is underestimated by data from patients at a fourth or later line of treatment, which leads to a false positive finding of synergy. Finally, the observation that 5% of trials are more than additive at a significance level of 0.05 also implies a possibility for false positive signals. ### Similar predictions of additivity and HSA For several combinations, observed PFS was consistent with both the HSA and additivity models because they made similar predictions. This scenario arises when one or both therapies have highly variable PFS times, such that patients are unlikely to have a similar magnitude of benefit from two different drugs. When patients experience similar magnitudes of drug effect (e.g., 5 + 5 = 10), the prediction of additivity is much greater than HSA (10 vs. 5), but with dissimilar effect sizes (e.g., 10 + 1 = 11), additivity and HSA models are similar (11 vs. 10). This phenomenon can be demonstrated by simulation (**Supplementary Figure S5A**) and is empirically observable as a negative correlation (*r* = –0.55; *P* = 0.0005) between the variability of the monotherapy PFS distributions (![Graphic][1]) and the difference between HSA and additivity models (**Supplementary Figure S5B**). ### Analysis by shared characteristics We next asked if there are features of combination therapies associated with greater or lesser efficacy than expected by additivity (**Figure 4D**). Whereas the confidence intervals of individual trials may make it challenging to detect differences from additivity, here we leveraged the statistical power of all combination therapies of a group. Immune checkpoint inhibitors (ICIs) and anti-angiogenesis agents are thought to synergize with each other and other therapies22–28. Therefore, we compared combinations that do or do not contain an ICI or an anti-angiogenesis therapy. We also divided combinations into those where the new drug does or does not have monotherapy approval in the same disease, motivated by the observation that synergy is most prevalent among weaker therapies in pre-clinical experiments29,30. Finally, we divided combinations by their magnitude of improvement compared to the control arm (HR above or below 0.61, which was the median HR across all trials) to test if synergy is associated with overall greater or lesser clinical efficacy. Among these eight groups, six were indistinguishable from additivity, including the most clinically successful combination therapies (greatest benefit over control arm). Two groups were significantly less than additive, being combinations with a new drug that lacked monotherapy approval, and combinations with a smaller net benefit versus the control arm (Wilcoxon signed-rank test, nominal *P* = 0.008 and *P* = 0.014, respectively). ### Limitations Our analysis has limitations in common with previous articles using the HSA model16,17, the most important of which concern the availability of monotherapy data. 45 approved drug combinations could not be analyzed because there is no published data on the efficacy of one or more constituent drugs at matching doses (**Supplementary Table 2**). Of 37 combinations with available monotherapy data, in 17 cases monotherapy data was from patients who had experienced more prior therapy. Because therapies are less effective in more heavily pre-treated cancers, use of these data may overestimate the appearance of synergy. Therefore, the observed scarcity of synergy is more robust when considering limitations in available data. In seven combination therapies, one agent was used at a dose that was 10% to 25% lower than its monotherapy dosage. It is possible that positive interactions compensated for these dose changes. We have analyzed PFS and not overall survival (OS) because OS, while vital to clinical and regulatory decisions, is affected by subsequent therapies and post-progression survival, which confound the analysis of drug interactions. All data sources and limitations are described in **Supplementary Table 1**. ## DISCUSSION Across 25 years of FDA approvals of combination therapies for advanced cancer, dose-matched monotherapy data shows that 95% of combination therapies exhibited PFS distributions that were equal to the sum of their parts, or less. These findings *do not suggest* these are not effective combination therapies; additivity means that they are as effective as expected. Drug combinations often generate non-additive cellular phenotypes. Our findings do not refute such experimental observations but imply that humans rarely experience ‘more-than-additive’ durations of tumor control. The key conclusions of this study are that the clinical efficacies of most approved drug combinations are predictable from the efficacy of their constituents, and that drug-drug interactions are therefore not necessary to make clinically effective combination therapies. We caution against interpreting these findings as an adverse judgment of combination therapies; such a perception could arise from the common but erroneous belief that synergistic drug combinations are more effective than additive drug combinations6. The effect of a synergistic combination can be written as A + B + I where A and B are individual drug effects and I is the extra effect due to synergistic interaction, whereas the effect of an additive combination of different drugs can be written as C + D. If all single drugs were equally effective (A=B=C=D) then the synergistic combination would be most effective. However, at clinically relevant doses in humans, some drugs are more effective at treating cancers than others. Due to this simple fact, observing synergy between therapies A and B provides no information about the relative clinical efficacy of combinations AB versus CD (I>0 does not imply that A+B+I > C+D). Thus our observation of additivity by approved combination therapies in no way makes them disappointing or inferior to hypothetical synergistic combinations. Rather, these findings are useful because they suggest that combined drug efficacy can be prospectively estimated by additivity. A mechanistic rationale is widely seen as necessary to justify trials of combination therapy2,3,6,7. In specific contexts, synergistic drug interactions can be therapeutically valuable and remain worthy goals, such as to inhibit redundant or ‘bypass’ oncogenic signals31,32. As a general matter though, the prevalence of additivity among the best combination therapies established in the past quarter of a century refutes the dogma that drug combinations need to be justified by a mechanism of drug-drug interaction. The perception that synergy is required for efficacy pressures pre-clinical research to choose model systems, drug doses, and modes of analysis that maximize the potential to observe synergy in cell culture or mice, which chiefly arises among weak drugs or at sub-inhibitory doses29,30. Synergy metrics quantify the difference between observed and expected additive effects, which is appropriate to detect interactions. However, prioritizing drug combinations for interaction has the unintended effect of penalizing combinations of highly effective agents, because a large additive effect is subtracted. Indeed, the largest datasets on drug combinations in cancer cells show that synergy most frequently occurs among weak drugs30,33. These results support efficacy-based design of combination therapies as a complement to mechanism-based design. Put simply, a combination effect of ‘1+1 = 3’ demonstrates an interesting interaction but ‘10+10 = 20’ is a larger effect. The accuracy of the additivity model and scarcity of synergy suggests that tumor heterogeneity, not drug interaction, is the major origin of benefit of approved combinations of cancer therapies. The model of clinical additivity can be understood as describing both inter-patient and intra-tumor heterogeneity. Inter-patient variability was simulated by sampling single-drug PFS times from clinically observed distributions, which generates a spectrum of different combination responses across patients. Adding single-drug PFS times is the plain meaning of addition, but also, it corresponds to the Bliss independence model which describes the ability of drug combinations to overcome intra-tumor heterogeneity34 (e.g., 90% + 90% tumor cell kill = 99% cell kill; **Supplementary Figure S6**). As described previously16,35, tumor heterogeneity may explain the seeming inconsistency between pre-clinical synergies and clinical additivity. *In vitro* experiments show that synergy depends on dosage and occurs variably across heterogeneous panels of cell lines29,30. Synergy may therefore arise at certain concentrations in a fraction of patients, as it does in cell cultures, but without significantly affecting survival in diverse patient populations. Phase III trials are the decisive final step in establishing superior cancer treatments, and unfortunately few have positive results. The consistency between 25 years of practice-changing trial results and the additivity model suggests that it could be useful for the prospective design of phase III trials of combination therapies. By estimating survival distributions, the additivity model can predict the likelihood of success of novel drug combinations in different cancer types and inform trial designs and statistical analyses. Thus, the model of drug additivity has the potential to improve the rate of success of phase III trials and accelerate progress in cancer treatment. ## Methods ### Data Collection We searched all drug combinations FDA-approved for advanced cancers between 1995 and 2020, from FDA Oncology / Hematologic Malignancies Approval Notifications ([https://www.fda.gov/drugs/resources-information-approved-drugs/oncology-cancer-hematologic-malignancies-approval-notifications](https://www.fda.gov/drugs/resources-information-approved-drugs/oncology-cancer-hematologic-malignancies-approval-notifications)) and [centerwatch.com](http://centerwatch.com) ([https://www.centerwatch.com/directories/1067-fda-approved-drugs](https://www.centerwatch.com/directories/1067-fda-approved-drugs)) (**Figure 1**). We sought combinations where the approval was based on a trial of ‘standard of care’ versus ‘standard of care plus new agent’. We required Kaplan-Meier survival curves of PFS for the combination arm, the experimental single-agent arm, and the control (standard of care) arm. When all three arms were not available from the same trial, we searched clinical trial publications of all phases that included the missing arm based on the following criteria: (1) patients had the same disease, (2) dosage difference was less than 1.5-fold, and (3) the trial arm contained more than 30 patients. We collected PFS data from 39 clinical trials of FDA-approved combination therapies. Two combinations had a difference of more than three lines of therapies or had a mismatch in baseline patient characteristics that can potentially affect drug responses. These were analyzed in supplement and were removed from subsequent analyses. Biomarker-positive subpopulations (e.g., PD-L1 expression for immune checkpoint inhibitors) were also analyzed in supplement. Digitization of Kaplan-Meier plots was as previously described16,17. ### Clinical definition of drug additivity For PFS as a metric of clinical efficacy, the plain meaning of ‘additivity’ is to add PFS times. This corresponds to a common pre-clinical definition of additivity. Specifically, among the two dominant definitions of drug additivity, Loewe’s model requires dose-response measurements and cannot apply to clinical data, but the Bliss independence model can be adapted to clinical data. The Bliss model defines non-interacting drug pairs as conferring independent probabilities of cell death. If tumor progression arises from the exponential growth of cancer cells that were not killed by drug treatment, this corresponds to addition of PFS times of individual drugs (**Supplementary Figure S6**)36,37. Like the Bliss and Loewe models, this ‘null model’ of additivity does not anticipate that real mechanisms are as simple as the null model, but defines the efficacy expected in the absence of synergistic drug-drug interactions. In clinical practice, quantifying the PFS benefit of a drug must consider that progression takes time, even in the absence of effective therapy, and is observable at scheduled radiological scans. Therefore to ‘add’ the PFS benefits of drugs beyond the first scan, the equation for clinical additivity is PFSAB = PFSA + PFSB – PFSuntreated. In a variety of aggressive cancers, trials with a placebo-only or best supportive care arm show that most patients exhibit progression at their first scheduled, suggesting this time as a proxy for PFSuntreated (**Supplementary Figure S1**). When the first scheduled scan time differed between the monotherapy arms, we subtracted the larger of the two to avoid overestimating the activity of the drugs. ### Simulation of HSA and Additivity Model We previously published a method to compute the PFS distribution of a combination therapy from the PFS distributions of its constituents16. The underlying theory was originally named ‘independent drug action’ in 196111. For clarity, we refer to this model as ‘Highest Single Agent’ (HSA) because each patient’s PFS time is the longest of the two PFS times of constituent drugs. Constituents can be monotherapies or a combination of fewer drugs than the full combination. Briefly, this method uses PFS distributions of individual drugs (PFSA, PFSB) to generate a joint distribution, with correlation as supported by experimental data16,17. Drug screens in cell lines and patient-derived xenografts (PDX) were used to estimate the correlation between drug responses38–40. We calculated the Spearman correlation between drug pairs using area under the curve (AUC) for cell lines and best average response for PDXs as drug response metrics **(Supplementary Figure S7**). If a drug did not exist in the databases, it was substituted with another compound with the same mechanism of action (e.g., topotecan for irinotecan or oxaliplatin for cisplatin). We used all cancer types, where the drug was active (AUC < 0.8) in at least 10% of the cell lines if the correlation were similar within a cancer type and across all cancer types. We used cancer type-specific correlation otherwise. Average pairwise correlation (ρ=0.3 for drugs with different modalities and ρ=0.52 for cytotoxic chemotherapy combinations) of all active drugs in CTRPv241 was used when individual drug data was unavailable. For these combinations, the range of observed correlations covering 95% of all active drug pairs [-0.01, 0.64] corresponded to an average uncertainty of ± 2.29% and ± 2.67% in PFS for additivity and HSA models respectively. For the HSA model, each of 5,000 datapoints from the joint distribution is assigned a combination therapy PFS equal to the longest PFS time (PFSAB = max(PFSA, PFSB))16. The additivity model is the same except combination PFS is the sum of PFS times, with a correction for scan interval (PFSAB = PFSA + PFSB – PFSuntreated), as defined above. Predictions were made over the timespan reaching the shortest follow-up time between the trial arms if both arms did not drop as low as 5% PFS. Predictions were otherwise limited to the end of follow-up of the combination arm. The generation of partially correlated distributions introduces small stochastic differences, and therefore for each model we conducted 100 simulations, and selected that which produced the median result at 50% PFS. Simulated and clinically observed PFS distributions were compared using Cox’s proportional hazards model42. For observed combination therapy arms, individual patient data were imputed from published Kaplan-Meier curves at at-risk tables43,44. For modeled PFS distributions, we created 500 patient events by dividing PFS curves into equal increments; the same process was required for trials that did not include an at-risk table. To assess the quality of fit, 5000 datapoints with equal time intervals were used to calculate the coefficient of determination (R2) and mean signed difference between predicted and observed PFS distributions. Complete source data and software are in **Supplementary Data**. ## Supporting information Supplementary Figures [[supplements/281013_file02.pdf]](pending:yes) Supplementary Tables [[supplements/281013_file03.xlsx]](pending:yes) ## Data Availability All data produced in the present work are contained in the manuscript. ## DISCLOSURES A.C.P. has received consulting fees from Merck and research funding from Prelude Therapeutics. ## ACKNOWLEDGEMENTS We thank the investigators and patients who participated in the clinical trials analyzed in this work. H.H. is supported by the Korean Government Scholarship for Study Overseas, S.P. is supported by NIGMS grant T32-GM135095, and D.P. is supported by NIGMS grant T32-GM007753 and F30-CA260780. * Received October 21, 2022. * Revision received October 21, 2022. * Accepted October 22, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NoDerivs 4.0 International), CC BY-ND 4.0, as described at [http://creativecommons.org/licenses/by-nd/4.0/](http://creativecommons.org/licenses/by-nd/4.0/) ## REFERENCES 1. 1.Damon, L. E. & Cadman, E. C. Advances in rational combination chemotherapy. Cancer Invest. 4, 421–444 (1986). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=3542142&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 2. 2.Verweij, J., Disis, M. L. & Cannistra, S. A. Phase I studies of drug combinations. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 28, 4545–4546 (2010). 3. 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). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjIwLzE2LzQyMTAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMC8yMi8yMDIyLjEwLjIxLjIyMjgxMDEzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 4. 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). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJM197305102881905&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=4348752&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1973P520000005&link_type=ISI) 5. 5.Foucquier, J. & Guedj, M. Analysis of drug combinations: current methodological landscape. Pharmacol. Res. Perspect. 3, e00149 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/prp2.149&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26171228&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 6. 6.Rationalizing combination therapies. Nat. Med. 23, 1113–1113 (2017). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 7. 7.Boshuizen, J. & Peeper, D. S. Rational Cancer Treatment Combinations: An Urgent Clinical Need. Mol. Cell 78, 1002–1018 (2020). 8. 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). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/annonc/mdr608&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22314859&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 9. 9.Berenbaum, M. C. What is synergy? Pharmacol Rev 41, 93–141 (1989). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2692037&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1989CF38200001&link_type=ISI) 10. 10.Loewe, S. & Muischnek, H. Über Kombinationswirkungen. Naunyn-Schmiedebergs Arch. Für Exp. Pathol. Pharmakol. 114, 313–326 (1926). 11. 11.Bliss, C. I. The Toxicity of Poisons Applied Jointly. Ann. Appl. Biol. 26, 585–615 (1939). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1744-7348.1939.tb06990.x&link_type=DOI) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000188234200010&link_type=ISI) 12. 12.Meyer, C. T. et al. Quantifying Drug Combination Synergy along Potency and Efficacy Axes. Cell Syst. 8, 97-108.e16 (2019). 13. 13.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). 14. 14.Wittes, R. E. & Goldin, A. Unresolved issues in combination chemotherapy. Cancer Treat. Rep. 70, 105–125 (1986). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=3510730&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1986AYY5500012&link_type=ISI) 15. 15.Frei, E. et al. Studies of Sequential and Combination Antimetabolite Therapy in Acute Leukemia: 6-Mercaptopurine and Methotrexate. Blood 18, 431–454 (1961). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czo4OiIxOC80LzQzMSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzEwLzIyLzIwMjIuMTAuMjEuMjIyODEwMTMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 16. 16.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.e13 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2017.11.009&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29245013&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 17. 17.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). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6ODoiMjgvMi8zNjgiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMC8yMi8yMDIyLjEwLjIxLjIyMjgxMDEzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 18. 18.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). 19. 19.Finn, R. S. et al. Palbociclib and Letrozole in Advanced Breast Cancer. N. Engl. J. Med. 375, 1925–1936 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1607303&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27959613&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 20. 20.Dimopoulos, M. A. et al. Daratumumab, Lenalidomide, and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 375, 1319–1331 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1607751&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27705267&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 21. 21.Lonial, S. et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. The Lancet 387, 1551–1560 (2016). 22. 22.Cooper, Z. A., Reuben, A., Amaria, R. N. & Wargo, J. A. Evidence of synergy with combined BRAF-targeted therapy and immune checkpoint blockade for metastatic melanoma. OncoImmunology 3, e954956 (2014). 23. 23.Melero, I. et al. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat. Rev. Cancer 15, 457–472 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrc3973&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26205340&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 24. 24.Blagosklonny, M. V. How avastin potentiates chemotherapeutic drugs: Action and reaction in antiangiogenic therapy. Cancer Biol. Ther. 4, 1307–1310 (2005). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4161/cbt.4.12.2315&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16322683&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000236044400010&link_type=ISI) 25. 25.He, M. et al. Immune Checkpoint Inhibitor-Based Strategies for Synergistic Cancer Therapy. Adv. Healthc. Mater. 10, 2002104 (2021). 26. 26.Galluzzi, L., Humeau, J., Buqué, A., Zitvogel, L. & Kroemer, G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat. Rev. Clin. Oncol. 17, 725–741 (2020). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 27. 27.Gerber, H.-P. & Ferrara, N. Pharmacology and Pharmacodynamics of Bevacizumab as Monotherapy or in Combination with Cytotoxic Therapy in Preclinical Studies. Cancer Res. 65, 671–680 (2005). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjg6IjY1LzMvNjcxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 28. 28.Yi, M. et al. Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol. Cancer 18, 60 (2019). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 29. 29.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). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-019-09799-2&link_type=DOI) 30. 30.Jaaks, P. et al. Effective drug combinations in breast, colon and pancreatic cancer cells. Nature 603, 166–173 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-022-04437-2&link_type=DOI) 31. 31.Prahallad, A. et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483, 100–103 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature10868&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22281684&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000300877900053&link_type=ISI) 32. 32.Vellano, C. P. et al. Androgen receptor blockade promotes response to BRAF/MEK-targeted therapy. Nature 606, 797–803 (2022). 33. 33.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). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-019-09799-2&link_type=DOI) 34. 34.Pomeroy, A. E., Schmidt, E. V., Sorger, P. K. & Palmer, A. C. Drug independence and the curability of cancer by combination chemotherapy. Trends Cancer S2405-8033(22)00137–6 (2022) doi:10.1016/j.trecan.2022.06.009. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.trecan.2022.06.009&link_type=DOI) 35. 35.Plana, D., Palmer, A. C. & Sorger, P. K. Independent Drug Action in Combination Therapy: Implications for Precision Oncology. Cancer Discov. 12, 606–624 (2022). 36. 36.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). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=14117037&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 37. 37.Koch, R. et al. Biomarker-driven strategy for MCL1 inhibition in T-cell lymphomas. Blood 133, 566–575 (2019). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czo5OiIxMzMvNi81NjYiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMC8yMi8yMDIyLjEwLjIxLjIyMjgxMDEzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 38. 38.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). 39. 39.Seashore-Ludlow, B. et al. Harnessing Connectivity in a Large-Scale Small-Molecule Sensitivity Dataset. Cancer Discov. 5, 1210–1223 (2015). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiY2FuZGlzYyI7czo1OiJyZXNpZCI7czo5OiI1LzExLzEyMTAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMC8yMi8yMDIyLjEwLjIxLjIyMjgxMDEzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 40. 40.Gao, H. et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat. Med. 21, 1318–1325 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.3954&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26479923&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 41. 41.Rees, M. G. et al. Correlating chemical sensitivity and basal gene expression reveals mechanism of action. Nat Chem Biol 12, 109–16 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nchembio.1986&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26656090&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 42. 42.Cox, D. R. Regression Models and Life-Tables. J. R. Stat. Soc. 32, 187–220 (1972). 43. 43.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). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1471-2288-12-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22297116&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 44. 44.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](http://medrxiv.org/lookup/external-ref?access_num=29562148&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 45. 45.Sledge, G. W. et al. MONARCH 2: Abemaciclib in Combination With Fulvestrant in Women With HR+/HER2− Advanced Breast Cancer Who Had Progressed While Receiving Endocrine Therapy. J. Clin. Oncol. 35, 2875–2884 (2017). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 46. 46.Johnston, S. et al. MONARCH 3 final PFS: a randomized study of abemaciclib as initial therapy for advanced breast cancer. Npj Breast Cancer 5, 5 (2019). 47. 47.Thomas, E. et al. Phase II Clinical Trial of Ixabepilone (BMS-247550), an Epothilone B Analog, in Patients With Taxane-Resistant Metastatic Breast Cancer. J. Clin. Oncol. 25, 3399–3406 (2007). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyNS8yMy8zMzk5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 48. 48.Stein, S. Lapatinib in combination with capecitabine in the management of ErbB2-positive (HER2-positive) advanced breast cancer. Biol. Targets Ther. 61 (2008) doi:10.2147/BTT.S1713. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2147/BTT.S1713&link_type=DOI) 49. 49.Van Cutsem, E. et al. Fluorouracil, Leucovorin, and Irinotecan Plus Cetuximab Treatment and RAS Mutations in Colorectal Cancer. J. Clin. Oncol. 33, 692–700 (2015). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjg6IjMzLzcvNjkyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 50. 50.Wilke, H. et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 15, 1224–1235 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1470-2045(14)70420-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25240821&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 51. 51.Vermorken, J. B. et al. Open-Label, Uncontrolled, Multicenter Phase II Study to Evaluate the Efficacy and Toxicity of Cetuximab As a Single Agent in Patients With Recurrent and/or Metastatic Squamous Cell Carcinoma of the Head and Neck Who Failed to Respond to Platinum-Based Therapy. J. Clin. Oncol. 25, 2171–2177 (2007). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyNS8xNi8yMTcxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 52. 52.Goede, V. et al. Obinutuzumab plus Chlorambucil in Patients with CLL and Coexisting Conditions. N. Engl. J. Med. 370, 1101–1110 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1313984&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24401022&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000333054000006&link_type=ISI) 53. 53.Long, G. V. et al. Dabrafenib plus trametinib versus dabrafenib monotherapy in patients with metastatic BRAF V600E/K-mutant melanoma: long-term survival and safety analysis of a phase 3 study. Ann. Oncol. 28, 1631–1639 (2017). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 54. 54.Stewart, A. K. et al. Carfilzomib, Lenalidomide, and Dexamethasone for Relapsed Multiple Myeloma. N. Engl. J. Med. 372, 142–152 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1411321&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25482145&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 55. 55.Palumbo, A. et al. Daratumumab, Bortezomib, and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 375, 754–766 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1606038&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27557302&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 56. 56.Facon, T. et al. Daratumumab plus Lenalidomide and Dexamethasone for Untreated Myeloma. N. Engl. J. Med. 380, 2104–2115 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1817249&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31141632&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 57. 57.Weber, D. M. et al. Lenalidomide plus Dexamethasone for Relapsed Multiple Myeloma in North America. N. Engl. J. Med. 357, 2133–2142 (2007). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa070596&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18032763&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000251076500005&link_type=ISI) 58. 58.Aghajanian, C. et al. OCEANS: A Randomized, Double-Blind, Placebo-Controlled Phase III Trial of Chemotherapy With or Without Bevacizumab in Patients With Platinum-Sensitive Recurrent Epithelial Ovarian, Primary Peritoneal, or Fallopian Tube Cancer. J. Clin. Oncol. 30, 2039–2045 (2012). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIzMC8xNy8yMDM5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 59. 59.Coleman, R. L. et al. Bevacizumab and paclitaxel–carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum-sensitive ovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG-0213): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 18, 779–791 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1470-2045(17)30279-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 60. 60.Pujade-Lauraine, E. et al. Bevacizumab Combined With Chemotherapy for Platinum-Resistant Recurrent Ovarian Cancer: The AURELIA Open-Label Randomized Phase III Trial. J. Clin. Oncol. 32, 1302–1308 (2014). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIzMi8xMy8xMzAyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 61. 61.Cristofanilli, M. et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol. 17, 425–439 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1470-2045(15)00613-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26947331&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 62. 62.Cortes, J. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. 396, 12 (2020). 63. 63.Tewari, K. S. et al. Improved Survival with Bevacizumab in Advanced Cervical Cancer. N. Engl. J. Med. 370, 734–743 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1309748&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24552320&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 64. 64.Giantonio, B. J. et al. Bevacizumab in Combination With Oxaliplatin, Fluorouracil, and Leucovorin (FOLFOX4) for Previously Treated Metastatic Colorectal Cancer: Results From the Eastern Cooperative Oncology Group Study E3200. J. Clin. Oncol. 25, 1539–1544 (2007). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyNS8xMi8xNTM5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 65. 65.Cunningham, D. et al. Cetuximab Monotherapy and Cetuximab plus Irinotecan in Irinotecan-Refractory Metastatic Colorectal Cancer. N. Engl. J. Med. 351, 337–345 (2004). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa033025&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15269313&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000222801200007&link_type=ISI) 66. 66.Horn, L. et al. First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer. N. Engl. J. Med. 379, 2220–2229 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1809064&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 67. 67.Gandhi, L. et al. Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell Lung Cancer. N. Engl. J. Med. 378, 2078–2092 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1801005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29658856&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 68. 68.Larkin, J. et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 381, 1535–1546 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1910836&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31562797&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 69. 69.Dimopoulos, M. et al. Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for patients with relapsed or refractory multiple myeloma (CANDOR): results from a randomised, multicentre, open-label, phase 3 study. The Lancet 396, 186–197 (2020). 70. 70.Dimopoulos, M. A. et al. Daratumumab plus pomalidomide and dexamethasone versus pomalidomide and dexamethasone alone in previously treated multiple myeloma (APOLLO): an open-label, randomised, phase 3 trial. Lancet Oncol. 22, 801–812 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/s1470-2045(21)00128-5&link_type=DOI) 71. 71.Wang-Gillam, A. et al. NAPOLI-1 phase 3 study of liposomal irinotecan in metastatic pancreatic cancer: Final overall survival analysis and characteristics of long-term survivors. Eur. J. Cancer 108, 78–87 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ejca.2018.12.007&link_type=DOI) 72. 72.O’Shaughnessy, J. et al. Superior Survival With Capecitabine Plus Docetaxel Combination Therapy in Anthracycline-Pretreated Patients With Advanced Breast Cancer: Phase III Trial Results. J. Clin. Oncol. 20, 2812–2823 (2002). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyMC8xMi8yODEyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 73. 73.Baselga, J. et al. Everolimus in Postmenopausal Hormone-Receptor–Positive Advanced Breast Cancer. N. Engl. J. Med. 366, 520–529 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1109653&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22149876&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000299991300008&link_type=ISI) 74. 74.Slamon, D. J. et al. Use of Chemotherapy plus a Monoclonal Antibody against HER2 for Metastatic Breast Cancer That Overexpresses HER2. N. Engl. J. Med. 344, 783–792 (2001). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJM200103153441101&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11248153&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000167440400001&link_type=ISI) 75. 75.Van Cutsem, E. et al. Addition of Aflibercept to Fluorouracil, Leucovorin, and Irinotecan Improves Survival in a Phase III Randomized Trial in Patients With Metastatic Colorectal Cancer Previously Treated With an Oxaliplatin-Based Regimen. J. Clin. Oncol. 30, 3499–3506 (2012). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIzMC8yOC8zNDk5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 76. 76.Socinski, M. A. et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 378, 2288–2301 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1716948&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 77. 77.West, H. et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 20, 924–937 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1470-2045(19)30167-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) 78. 78.Sandler, A. B. et al. Phase III Trial of Gemcitabine Plus Cisplatin Versus Cisplatin Alone in Patients With Locally Advanced or Metastatic Non–Small-Cell Lung Cancer. J. Clin. Oncol. 18, 122–122 (2000). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjg6IjE4LzEvMTIyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 79. 79.Burger, R. A., Fleming, G. F., Mannel, R. S., Greer, B. E. & Liang, S. X. Incorporation of Bevacizumab in the Primary Treatment of Ovarian Cancer. N. Engl. J. Med. 11 (2011). 80. 80.Crawford, E. D. et al. A controlled trial of leuprolide with and without flutamide in prostatic carcinoma. N. Engl. J. Med. 321, 419–424 (1989). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJM198908173210702&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2503724&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F10%2F22%2F2022.10.21.22281013.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1989AK85200002&link_type=ISI) 81. 81.Vogelzang, N. J. et al. Phase III Study of Pemetrexed in Combination With Cisplatin Versus Cisplatin Alone in Patients With Malignant Pleural Mesothelioma. J. Clin. Oncol. 21, 2636–2644 (2003). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyMS8xNC8yNjM2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTAvMjIvMjAyMi4xMC4yMS4yMjI4MTAxMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) [1]: /embed/inline-graphic-1.gif