Skip to main content
ESMO Open logoLink to ESMO Open
. 2020 Oct 29;5(5):e000871. doi: 10.1136/esmoopen-2020-000871

Evaluation of the contribution of randomised cancer clinical trials evaluating agents without documented single-agent activity

Jared Foster 1,, Boris Freidlin 1, E L Korn 1, Malcolm Smith 2
PMCID: PMC7597487  PMID: 33122353

Abstract

Background

With the development of targeted agents, the approach to combination cancer therapy has evolved to focus on identifying ways in which pathway inhibition by one agent may enhance the activity of other agents. In theory, this implies that under this new paradigm, agents are no longer required to show single-agent activity, as the pathway inhibited by the targeted agent may only have a therapeutic effect when given with other agents. This raises the question of the extent to which anticancer agents without single-agent activity can contribute to effective combination regimens.

Patients and methods

We reviewed outcomes of randomised phase 2 combination trials sponsored by the National Cancer Institute Cancer Therapy Evaluation Program that were activated in 2008 to 2017 and noted the single-agent activity of the experimental agents.

Results

Fifty-three trials were identified, and 50 had available results: 7 (14%), 15 (30%) and 28 (56%) had experimental agents with single-agent activity classified as active, inactive and indeterminate, respectively. Thirteen per cent (95% CI=1.7% to 40.5%) of trials evaluating inactive agents and 11.6% (95% CI=3.9% to 25.1%) of trials evaluating agents without known single-agent activity (pooled inactive and indeterminate) were positive, compared with 42.9% (95% CI=9.9% to 81.6%) for agents with single-agent activity.

Conclusions

Incorporating agents without documented single-agent activity into treatment regimens is unlikely to produce meaningful improvements in activity unless there is compelling biological rationale. This finding has important implications for the prioritisation of anticancer agents for combination testing, and for the allocation of clinical trial resources.

Keywords: drug development, combination therapy, clinical trials


Key questions.

What is already known about this subject?

  • With the development of targeted agents, the approach to combination therapy has evolved to focus on identifying ways in which pathway inhibition by an agent may enhance the activity of other components of a treatment regimen. Agents without single-agent activity would previously have been discarded, but this new paradigm removed the requirement for single-agent activity, as the pathway inhibited by the targeted agent may only have a therapeutic effect when given with other agents. Removing this requirement for single-agent activity has dramatically expanded the number of potential combinations evaluable in cancer patients, as agents can now be selected for evaluation in a combination regimen based on a plausible mechanism of action, and on some level of preclinical supportive data that the mechanism is relevant.

What does this study add?

  • We examined the outcomes of randomised phase 2 trials sponsored by the NCI Cancer Therapy Evaluation Program (CTEP) to better understand the extent to which anticancer agents without single agent activity can contribute to effective combination therapy regimens. The results of this review may help inform future prioritisation of anticancer agents for combination testing, and the allocation of clinical trial resources.

How might this impact on clinical practice?

  • Our results suggest that this approach has a very low yield in the absence of compelling biological rationale, and that the level of evidence currently used to select agents lacking single-agent activity for combination testing is inadequate.

Introduction

Determining which combinations of anticancer agents to evaluate in specific patient populations was reasonably straightforward through most of the second half of the 20th century, and was based on rules from the 1950s developed for treating children with acute lymphoblastic leukaemia, with supportive evidence coming from studies of mouse models of leukaemia.1 2 The basic principle was that combinations of active agents were likely to be more effective than the same agents used alone, and that sufficient numbers of active agents used in combination could potentially result in curative therapy. Potential explanations of the benefit of treatment regimens that combine active agents include increased anticancer cell kill, circumvention of development of resistance to the single agents that make up the regimen, greater efficacy against heterogeneous tumours with different resistance mechanisms and synergistic interactions between agents used in the regimen.3 Furthermore, combinations of active agents can also address between-patient tumour heterogeneity when different patients are sensitive to different agents of the combination.

With the development of targeted agents that potently inhibit signalling pathways, the approach to combination therapy has evolved to focus on identifying ways in which pathway inhibition by an agent may enhance the activity of other components of a treatment regimen. Whereas previously agents without single-agent activity would have been discarded, this new paradigm removed the requirement for single-agent activity, as the pathway inhibited by the targeted agent may only have a therapeutic effect when given with other agents. Without a requirement for single-agent activity, the number of potential combinations evaluable in cancer patients has expanded dramatically, as agents can now be selected for evaluation in a combination regimen based on a plausible mechanism of action, and on some level of preclinical supportive data that the mechanism is relevant. In most settings, isolating the contribution of the new agent to the combination requires a randomised trial. Typically, an ‘add-on’ design is used, where patients are randomised between two arms, with the control arm receiving the standard regimen and the combination arm receiving the new agent in addition to the standard regimen.

The question of the extent to which anticancer agents without single-agent activity can contribute to effective combination therapy regimens is an empirical one. To investigate this question, we identified randomised phase 2 trials sponsored by the National Cancer Institute (NCI) Cancer Therapy Evaluation Program (CTEP) and categorised the trials into those for which the experimental agent had single-agent activity, minimal or no single-agent activity or unknown single-agent activity. The outcomes of these phase 2 trials were then examined.

Methods

The CTEP trials database was searched for randomised comparative phase II trials involving more than one agent (including radiation) that were activated on or after 1 January 2008 and were permanently closed to accrual by 31 December 2017. This search was limited to add-on trials. Because the focus was on clinical trials evaluating agents tested in combination with systemic therapies, trials in which the experimental regimen involved the addition of surgery or radiation to the control-arm regimen were excluded, as were trials in which the experimental agent was being evaluated as a radiosensitizer. Because the result of adding anti-angiogenic agents targeting the vascular endothelial growth factor (VEGF) pathway to chemotherapy regimens has been well-delineated for multiple cancer types as modestly prolonging time to event in the advanced disease setting but not in the adjuvant setting,4 trials adding agents with VEGF targets were excluded. Trials involving immunotherapy agents were also excluded given the distinctive mechanism of action of these agents and the possibility that the rules for identifying beneficial combinations involving the addition of immunotherapy agents may differ from those of agents acting directly on cancer cells. In addition, trials in which patients were required to have previously progressed on the experimental agent (or another similar agent) were excluded. Trials which achieved less than 30% of the target accrual (unless this was because an interim stopping bound was crossed) were also excluded. For trials with more than two arms and multiple additive comparisons, each additive comparison was treated as a separate trial. To find trial publications, Internet searches were performed using the CTEP trial ID, NCI’s National Clinical Trials Network (NCTN) number, trial title and a combination of the principal investigator’s surname, agents in the trial and cancer histology.

For each trial, all published/publicly available single-agent trial data (including data that were not available at the time the trial protocol was written) were used to classify the level of single-agent response activity of the experimental agent for the specific histology of interest. As summarised in table 1, experimental agents were classified as active for a particular histology if the corresponding observed response rate (pooled across all relevant trials) was greater than 5%, and those for which the observed response rate was at most 5% were classified as inactive. Experimental agents received the classification ‘indeterminate’ activity if at most one response was observed among 13 or fewer patients, or exactly one response was observed among 14 to 19 patients. Single-agent activity assessments were completed prior to viewing the results of the trials in this review. After it was discovered that some experimental agents that met criteria for single-agent activity were used in combination at dose intensities 50% less than their single-agent dose per unit time, these agents were reclassified as having indeterminate single-agent activity.

Table 1.

Single-agent activity classification guidelines

Number of evaluable patients Conclusion about activity based on number of responses
Active Inactive Not enough data
n<14 >2 -- <1
n>14 and n<20 >2 0 1
n>20 >5% <5% --

Once single-agent activity was assessed for the experimental agent in each trial, the trial publications were reviewed, and results were classified as ‘positive’, ‘negative’ or ‘unknown’ based on the following criteria. If at least 80% of the target accrual was achieved (and the trial was not stopped early due to futility or efficacy), the trial was classified as positive or negative based on the published primary analysis (assuming this was the per-protocol primary analysis). Likewise, if a trial was stopped early as a result of a preplanned interim efficacy or futility analysis, the results were classified as positive or negative, respectively. If a trial achieved less than 80% of the target accrual (and an early stopping rule was not triggered), we used the published primary analysis (as well as the point estimate and SE, when necessary) to (separately) test the protocol-defined null and alternative hypotheses using one-sided errors set at the protocol-defined type I and type II errors, respectively. If the observed result was significantly better than the null, the trial was classified as positive. If the observed result was significantly worse than the alternative (and did not differ significantly from the null), the trial was classified as negative. Finally, the trial was classified as unknown if the observed result was not found to differ significantly from either the null or alternative.

The sample proportion of positive trials (and the corresponding 95% CI) was computed using only trials that were classified as positive or negative. These estimates were obtained separately for the subgroups of trials whose experimental agents were classified as active, inactive and indeterminate. In addition, though power was limited, the proportion of positive trials was compared among the trial subgroups using Fisher’s exact test with one-sided type I error of 0.05: the active experimental agent subgroup was compared with the inactive-agent subgroup alone and to the pooled inactive and indeterminate-activity agent subgroups.

Results

Sixty-seven eligible protocols representing 73 randomised trials were identified (figure 1). Three trials were excluded because results were not published (or otherwise publicly available), and an additional 20 trials were excluded because they enrolled <30% of their planned accrual for reasons unrelated to efficacy/futility (including 13 trials with zero accrual), leaving 50 trials analysed in this review. Among these 50 trials, 7 (14%), 15 (30%) and 28 (56%) had experimental agents that were classified as active, inactive and indeterminate activity, respectively.

Figure 1.

Figure 1

Flow diagram of trial selection. (‘Active/inactive/indeterminate’ in the last row refers to the single-agent activity of the experimental agent.) CTEP, Cancer Therapy Evaluation Program; VEGF, vascular endothelial growth factor.

Five of 43 (11.6%) randomised trials evaluating experimental agents not known to have single-agent activity were considered positive based on their protocol-defined criteria for success: 2 of 15 (13.3%) trials of inactive experimental agents and 3 of 28 (10.7%) trials with indeterminate activity. These two subgroups of trials are considered together because a common reason for not evaluating single-agent activity is the belief that the experimental agent in question is likely to be ineffective as a single-agent and needs to be studied in combination. Details for the five trials evaluating experimental agents with either no (8457 and S1406) or indeterminate (E2511, GOG-0186I and N064B) single-agent activity that met protocol-defined criteria for ‘positive’ are provided in table 2. Additional details of these five trials are provided below:

Table 2.

Clinical trials of experimental agents without confirmed single-agent activity that met protocol-defined criteria for success

Clinical Trial # NCT # Tumour type Exptl agent Single-agent activity (N/IND) Stnd arm agent(s) N (exptl) N (stnd) Primary endpoint Exptl outcome Stnd outcome P
value
Exptl ORR Stnd ORR Reference #
8457 NCT01142401 ER(+) breast cancer Bortezomib No Fulvestrant 59 59 PFS Median 2.7 months Median 2.7 months 0.06 NA NA 5
S1406 NCT02164916 BRAF mutant colorectal cancer Vemurafenib No Cetuximab
+ irinotecan
49 50 PFS Median 4.4 months Median 2.0 months <0.001 0.16 0.04 6
E2511 NCT01642251 Extensive-stage small cell lung cancer Veliparib IND Cisplatin
+ etoposide
64 64 PFS Median 6.1 months Median 5.5 months 0.023 0.72 0.66 7
GOG-0186I NCT01305213 Ovarian Fosbretabulin Tromethamine IND Bevacizumab 53 51 PFS Median 7.3 months Median 4.8 months 0.05 0.357 0.282 8
N064B NCT00550836 Pancreatic cancer Panitumumab IND Erlotinib
+ gemcitabine
46 46 OS Median 8.3 months Median 4.2 months 0.18 0.065 0.087 10

ER, oestrogen receptor; Exptl, experimental; IND, indeterminate; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; Stnd, standard.

  • Trial 8457 evaluated the addition of bortezomib to fulvestrant for women with oestrogen receptor (ER)-positive metastatic breast cancer resistant to aromatase inhibitors.5 There was no difference in median progression-free survival (PFS) between the experimental and control arms (2.7 months each), but the HR for PFS significantly favoured the combination arm (HR=0.73 with p=0.06, one-sided log-rank test) as the result of a modest separation of the curves at later time points. Although the study met protocol-defined criteria for ‘positive’, the impact of the addition of bortezomib appeared small and there has been no subsequent definitive clinical trial of bortezomib pursued for the study’s patient population.

  • The S1406 study evaluated the addition of vemurafenib to irinotecan and cetuximab for patients with BRAF-mutant colorectal cancer.6 For patients receiving vemurafenib, the median PFS was more than doubled (4.4 vs 2.0 months, HR=0.42 with p<0.001, one-sided log-rank test) and the objective response rate was higher (16% vs 4%). This study is discussed further below.

  • Trial E2511 evaluated the addition of veliparib to cisplatin and etoposide for newly diagnosed patients with extensive-stage small cell lung cancer.7 The two arms showed a small difference in median PFS favouring the veliparib arm (6.1 vs 5.5 months, stratified HR=0.63 with p=0.01, one-sided stratified log-rank test).

  • Trial GOG-0186I evaluated fosbretabulin (combretastatin A4-phosphate) in combination with bevacizumab for patients with ovarian cancer, and extended median PFS from 4.8 months for single-agent bevacizumab to 7.3 months for the combination (HR=0.69 with p=0.05).8 However, a subsequent phase 2 to phase 3 trial evaluating fosbretabulin in combination with bevacizumab and chemotherapy failed when the company terminated the study due to the lack of a meaningful improvement in PFS.9

  • Trial N064B evaluated the addition of panitumumab to erlotinib and gemcitabine for patients with advanced pancreatic cancer.10 Median overall survival was significantly improved (8.3 vs 4.2 months, HR=0.82 with p=0.18), but with an extension in median PFS of only 1.6 months (3.6 months vs 2.0 months) and increased toxicity among patients receiving dual epidermal growth factor receptor (EGFR) blockade. The authors concluded that further studies of EGFR inhibitors administered concurrently with cytotoxic agents were unlikely to result in a meaningful improvement in the outcome of patients with metastatic pancreatic cancer and were therefore not recommended.

There were seven randomised trials with experimental agents that had single-agent activity (table 3). Among these were three positive trials, E1412, E2211 and E240811–13 (see online supplemental table 1 for details). The single-agent response rates for the agents studied ranged from approximately 10% for rapalogs for ovarian cancer to 50% for bortezomib for follicular lymphoma.14 15 The single-agent response activity for capecitabine was high (75%), but was based on only four patients.16

Table 3.

Clinical trials of experimental agents with confirmed single-agent activity

Clinical Trial # NCT # Tumour type Exptl agent Stnd arm agent(s) N (exptl) N (stnd) Primary endpoint Exptl outcome Stnd outcome P
value
Positive study Y/N (per protocol) Exptl # eval for response Exptl # responders Exptl ORR Stnd # eval for Response Stnd # responders Stnd ORR Reference
#
E1412 NCT01856192 Diffuse large B cell lymphoma Lenalidomide Rituximab
+ chemotherapy
145 135 PFS Median not reached NA 0.03 Y NA NA 0.97 NA NA 0.92 11
E2211 NCT01824875 Pancreatic neuroendocrine tumours Capecitabine  Temozolomide  72 72 PFS Median 22.7 months Median 14.4 months 0.01 Y NA NA 0.33 NA NA 0.28 12
E2408 NCT01216683 Follicular lymphoma Bortezomib Rituximab
+ bendamustine
85 137 CR 74% 58% 0.016 Y NA NA 0.74 NA NA 0.58 13
E2408 NCT01216683 Follicular lymphoma Lenalidomide Rituximab 112 60 1 year DFS 45% 72% 0.99 N NA NA NA NA NA NA 41
9012 NCT01576172 Metastatic castration-resistant prostate cancer Veliparib Abiraterone
+ prednisone
79 74 PSA response 72% 63% 0.27 N 76 55 0.724 72 46 0.639 42
CALGB-50904 NCT01286272 Follicular lymphoma Bortezomib Ofatumumab
+ bendamustine
62 66 CR 60% 62% 0.68 N 62 37 0.597 66 41 0.621 43
GOG-0186G NCT00886691 Ovarian Everolimus Bevacizumab 75 75 PFS Median 5.9 months Median 4.5 months 0.39 N 63 14 0.222 58 7 0.121 44

CR, Complete Response; DFS, Disease Free Survival; Exptl, experimental; ORR, objective response rate; PFS, progression-free survival; PSA, Prostate-Specific Antigen; Stnd, standard.

Supplementary data

esmoopen-2020-000871supp001.pdf (112.7KB, pdf)

The percentage of positive trials among trials evaluating inactive agents was 13.3% (95% CI=1.7% to 40.5%), and that for trials evaluating agents without known single-agent activity (pooled inactive and indeterminate activity single agents) was 11.6% (CI=3.9% to 25.1%) (table 4). This is contrasted with 42.9% (95% CI=9.9% to 81.6%) positive trials among trials evaluating active agents. The comparison of positive trial rates between active and inactive agents did not meet criteria for statistical significance (p=0.16), nor did the comparison of such rates between active and no-known-activity agents (p=0.07), but, as noted earlier, there is limited power for these comparisons.

Table 4.

Trial outcomes by single-agent activity level

Single-agent activity Trial result Proportion of positive trials
(95% CI)
P value*
Negative Positive
Active 4 3 0.43 (0.10 to 0.82)
Inactive 13 2 0.13 (0.02 to 0.40) 0.16
Indeterminate 25 3 0.11 (0.02 to 0.28)
Inactive/indeterminate 38 5 0.12 (0.04 to 0.25) 0.07

*One-sided Fisher’s exact test comparing proportion of positive trials to that among active agents.

Discussion

Our key finding is that incorporating agents without documented single-agent activity into treatment regimens is unlikely to produce meaningful improvements in clinical activity. Only 5 of 43 (11.6%) trials evaluating experimental agents without documented single-agent activity were considered positive based on their protocol-defined criteria for success. When considering the percentage of ‘positive’ studies among the 43 trials using agents without documented single-agent activity, it is relevant to note that the average type 1 error rate among these 43 trials was 0.11. Hence a finding of 10% to 15% positive trials aligns with the expected rate for false-positive results. Furthermore, it appears that only one of these five positive trials, S1406, identified potential true therapeutic advances. This means a single trial among 43 trials of agents without documented single-agent activity represents a potential true treatment advance.

The addition of an agent without single-agent activity to another treatment can be hypothesised to increase efficacy through several mechanisms (eg, synergy, prevention of resistance, overcoming existing resistance). As noted above, the most clear-cut example of a positive clinical trial for an agent without single-agent activity is S1406, which evaluated the addition of vemurafenib to irinotecan and cetuximab for BRAF-mutant metastatic colorectal cancer. The study was convincingly positive, with a HR for PFS of 0.042 (p<0.001). The inactivity of BRAF inhibitors for BRAF-mutated colorectal cancer illustrates the importance of cellular context in defining the activity of some targeted agents.17 Multiple research teams identified the role of EGFR-pathway signalling in the resistance of colorectal cancer to vemurafenib,18–20 and hence the mechanistic rationale for concurrent BRAF-EGFR inhibition for overcoming an existing resistance mechanism was compelling: a known genomic alteration (BRAF) associated with sensitivity to a kinase inhibitor in other diseases plus a clearly defined resistance pathway in colorectal cancer resulting from EGFR-pathway signalling. A subsequent phase 3 trial in colorectal cancer confirmed clinical benefit for combining BRAF inhibition (using encorafenib and binimetinib) with EGFR inhibition with cetuximab.21 This example provides proof of concept that it is possible for an agent without single-agent activity to meaningfully contribute when used in combination if the biological rationale for the combination is truly understood and if excessive toxicity doesn’t intervene.

A complementary line of research that informs the question of the role of anticancer agents lacking single-agent activity is the evaluation of the contribution of components of known effective regimens. Palmer and Sorger evaluated Food and Drug Administration (FDA)-approved drug combinations and found that patient-to-patient variability and independent drug action were able to explain the superiority of many FDA-approved drug combinations in the absence of drug synergy or additivity.22 In a second report from these authors examining the contributions of individual agents to a curative regimen (R-CHOP), they found that effectiveness of the regimen was adequately explained by assuming independently active drugs with non-overlapping mechanisms of resistance, and that there was no need to assume synergistic interactions between the regimen’s components. Both of these reports are consistent with our finding that agents lacking single-agent activity are unlikely to add in a meaningful way to standard treatment regimens.

There are agents such as bevacizumab that slow tumour growth through an anti-angiogenic mechanism, and they were not included in this analysis because the activity of this class of agents when added to standard-of-care regimens has been evaluated in phase 3 trials for multiple tumour types and has been described.4 As single agents, objective response rates tend to be low, with notable exceptions for cancers such as renal cell carcinoma, ovarian cancer and alveolar soft part sarcoma. For patients with advanced disease, this class of agents can prolong progression-free survival by a modest amount for multiple tumour types, and in some cases overall survival may be extended as well. However, this class of agents is not effective when added to standard regimens in the adjuvant (curative) setting. The cyclin-dependent kinase (CDK)4/6 inhibitors may be similar to the anti-angiogenic agents in primarily working through slowing tumour growth, although they work through a different mechanism of action. These agents have been shown to prolong PFS when added to endocrine therapy for patients with advanced ER(+), human epidermal growth factor receptor 2 (HER2)(-) breast cancer, but early results suggest that their role in potentially curative treatments (ie, adjuvant therapy) may be limited.23 Their single-agent activity was not extensively studied, but for abemaciclib objective responses were observed.24

Kimmelman and colleagues applied a complementary approach to analyse the benefit, burden and impact of combination trials, and their findings are comparable to those that we report.25 They examined 323 published post-approval trials exploring combinations involving anticancer agents first licensed 2005 to 2007 inclusive (termed index agents). Over 70% of the trials evaluated a combination for an indication for which the index agent was not approved by FDA. Among the combinations evaluated, the only ones that resulted in either subsequent FDA approvals or a National Comprehensive Cancer Network guideline recommendation were those for which the index agent had single-agent activity for the relevant patient population. Importantly, the authors noted that for those trials randomising between a combination arm and a comparator, patients receiving the combination experienced comparable overall survival rates but higher rates of grade 3 to grade 4 severe adverse events and deaths.

Our results also complement those of Gyawali and Prasad who examined 18 FDA-approved combination therapies that incorporated a drug with negligible single-agent activity.26 Seven of the 18 combinations included a VEGF-targeted agent. Median prolongation in PFS (or time to progression) was only 2.3 months, and only 9 of 18 approved combinations were associated with improved overall survival (median increase 1.6 months). These analyses indicate that, even in the ‘best case’ scenario of a combination that achieves regulatory approval, clinical benefit is generally minimal.

The challenge of enhanced toxicity associated with combinations, such that dose reductions of component agents is required, was an issue for combinations involving the poly (ADP-ribose) polymerase (PARP) inhibitor veliparib with cytotoxic chemotherapy agents/regimens (8788, 9026, E2511 and S1513). While veliparib is tolerated at 400 mg two times per day (total daily dose of 800 mg) on a continuous dosing schedule as a single agent and shows activity in breast cancer gene (BRCA)-mutant patients at this dose,27–29 the clinical trials that we evaluated that combined veliparib with cytotoxic agents (8788, 9026, E2511 and S1513) used veliparib at a dose per unit time reduced by 75% to more than 90% compared with its single-agent dose per unit time.7 30–32 The issue of enhanced toxicity also applies to novel-novel combinations, as exemplified by studies in which phosphoinositide 3-kinase (PI3K) pathway and mitogen-activated protein (MAP) kinase pathway inhibitors are combined. Identifying tolerable doses for these combinations has been difficult due to high levels of toxicity at doses below the recommended doses of the agents used alone,33–36 and clinical development of these combinations has not progressed despite promising mechanistic and preclinical rationale.

The paradigm of using documentation of pathway inhibition rather than objective response rate as a threshold for testing agents in combination is similar to the approach proposed in 2007 for cancer vaccines and related biologics.37 This proposal from the Cancer Vaccine Clinical Trial Working Group was based on concerns that conventional phase 2 trials with objective response endpoints were not adequate for capturing the clinical potential of immunotherapy treatments, and that clinical trials designed to identify relevant biological activities of the vaccine/agent were needed to better credential candidate therapeutics for further evaluation. The paper was written in the early years of clinical development of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) targeted therapies,38 and the initial reports of objective responses to programmed cell death protein 1 (PD1)-targeted antibodies39 and to T-cell engaging antibodies appeared shortly thereafter.40 These subsequent reports re-established objective response rate as the gold-standard for an agent’s potential as a clinically effective immunotherapeutic agent.

In closing, the concept that targeted agents lacking single-agent activity may be able to potentiate in clinically meaningful ways the activity of other agents by inhibiting relevant signalling pathways has become widespread among cancer researchers. Our results suggest that applying this concept in clinical trials has a low yield in the absence of compelling biological rationale, and that the level of evidence currently used to select agents lacking single-agent activity for combination testing is inadequate. This finding has important implications for the prioritisation of anticancer agents for combination testing and for the allocation of clinical trial resources towards more promising lines of clinical investigation.

Footnotes

Contributors: JF: conception and design of this review; acquisition, analysis and interpretation of data; drafting, revising and final approval of the manuscript. BF: conception and design of this review; acquisition and interpretation of data; drafting, revising and final approval of the manuscript. ELK: conception and design of this review; acquisition and interpretation of data; drafting, revising and final approval of the manuscript. MS: conception and design of this review; interpretation of data; drafting, revising and final approval of the manuscript. JF, BF, ELK and MS agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Funding: The authors have not declared a specific grant for this research from any funding agency in the public, commercial or not-for-profit sectors.

Competing interests: None declared.

Patient consent for publication: Not required.

Provenance and peer review: Not commissioned; externally peer reviewed.

Data availability statement: All data relevant to the study are included in the article or uploaded as supplementary information. Data analysed in this manuscript are available in the online supplemental table.

Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.

References

  • 1.Skipper HE, Schabel FM, Wilcox WS. Experimental evaluation of potential anticancer agents. XIII. on the criteria and kinetics associated with "curability" of experimental leukemia. Cancer Chemother Rep 1964;35:1–111. [PubMed] [Google Scholar]
  • 2.Frei E, Karon M, Levin RH, et al. The effectiveness of combinations of antileukemic agents in inducing and maintaining remission in children with acute leukemia. Blood 1965;26:642–56. 10.1182/blood.V26.5.642.642 [DOI] [PubMed] [Google Scholar]
  • 3.Greco WR, Faessel H, Levasseur L. The search for cytotoxic synergy between anticancer agents: a case of Dorothy and the ruby slippers? J Natl Cancer Inst 1996;88:699–700. 10.1093/jnci/88.11.699 [DOI] [PubMed] [Google Scholar]
  • 4.Smith MA. Lessons learned from adult clinical experience to inform evaluations of VEGF pathway inhibitors in children with cancer. Pediatr Blood Cancer 2014;61:1497–505. 10.1002/pbc.25036 [DOI] [PubMed] [Google Scholar]
  • 5.Adelson K, Ramaswamy B, Sparano JA, et al. Randomized phase II trial of fulvestrant alone or in combination with bortezomib in hormone receptor-positive metastatic breast cancer resistant to aromatase inhibitors: a new York cancer Consortium trial. NPJ Breast Cancer 2016;2:16037. 10.1038/npjbcancer.2016.37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kopetz S, McDonough SL, Morris VK, et al. Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF -mutant metastatic colorectal cancer (SWOG 1406). JCO 2017;35:520 10.1200/JCO.2017.35.4_suppl.520 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Owonikoko TK, Dahlberg SE, Sica GL, et al. Randomized phase II trial of cisplatin and etoposide in combination with veliparib or placebo for extensive-stage small-cell lung cancer: ECOG-ACRIN 2511 study. J Clin Oncol 2019;37:222–9. 10.1200/JCO.18.00264 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Monk BJ, Sill MW, Walker JL, et al. Randomized phase II evaluation of bevacizumab versus bevacizumab plus Fosbretabulin in recurrent ovarian, tubal, or peritoneal carcinoma: an NRG Oncology/Gynecologic Oncology Group study. J Clin Oncol 2016;34:2279–86. 10.1200/JCO.2015.65.8153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mateon Therapeutics Mateon therapeutics. mateon therapeutics announces termination of focus study in ovarian cancer and restructuring to prioritize OXi4503 for AML, 2017. Available: http://investor.mateon.com/static-files/3d1b50e1-fbb4-42e1-b4af-c0e3a10f7952
  • 10.Halfdanarson TR, Foster NR, Kim GP, et al. A phase II randomized trial of panitumumab, erlotinib, and gemcitabine versus erlotinib and gemcitabine in patients with untreated, metastatic pancreatic adenocarcinoma: North central cancer treatment Group trial N064B (Alliance). Oncologist 2019;24:589–e160. 10.1634/theoncologist.2018-0878 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Nowakowski GS, Hong F, Scott DW, et al. Addition of lenalidomide to R-CHOP (R2CHOP) improves outcomes in newly diagnosed diffuse large B-cell lymphoma (DLBCL): first report of ECOG-ACRIN1412 a randomized phase 2 us intergroup study of R2CHOP vs R-CHOP. Hematol Oncol 2019;37:37–8. 10.1002/hon.6_2629 [DOI] [Google Scholar]
  • 12.Kunz PL, Catalano PJ, Nimeiri H, et al. A randomized study of temozolomide or temozolomide and capecitabine in patients with advanced pancreatic neuroendocrine tumors: a trial of the ECOG-ACRIN cancer research Group (E2211). JCO 2018;36:4004 10.1200/JCO.2018.36.15_suppl.4004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Evens AM, Hong FX, Habermann TM, et al. Effect of bortezomib on complete remission (CR) rate when added to bendamustine-rituximab (Br) in previously untreated high-risk (HR) follicular lymphoma (fl): a randomized phase II trial of the ECOG-ACRIN cancer research Group (E2408). J clin oncol 2016;34. [Google Scholar]
  • 14.O'Connor OA, Portlock C, Moskowitz C, et al. Time to treatment response in patients with follicular lymphoma treated with bortezomib is longer compared with other histologic subtypes. Clin Cancer Res 2010;16:719–26. 10.1158/1078-0432.CCR-08-2647 [DOI] [PubMed] [Google Scholar]
  • 15.Behbakht K, Sill MW, Darcy KM, et al. Phase II trial of the mTOR inhibitor, temsirolimus and evaluation of circulating tumor cells and tumor biomarkers in persistent and recurrent epithelial ovarian and primary peritoneal malignancies: a gynecologic Oncology Group study. Gynecol Oncol 2011;123:19–26. 10.1016/j.ygyno.2011.06.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bongiovanni A, Riva N, Calpona S, et al. Metronomic capecitabine in gastroenteropancreatic neuroendrocrine tumors: a suitable regimen and review of the literature. Onco Targets Ther 2014;7:1919–26. 10.2147/OTT.S68573 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kopetz S, Desai J, Chan E, et al. Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. J Clin Oncol 2015;33:4032–8. 10.1200/JCO.2015.63.2497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Klinger B, Sieber A, Fritsche-Guenther R, et al. Network quantification of EGFR signaling unveils potential for targeted combination therapy. Mol Syst Biol 2013;9:673. 10.1038/msb.2013.29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Corcoran RB, Ebi H, Turke AB, et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to Raf inhibition with vemurafenib. Cancer Discov 2012;2:227–35. 10.1158/2159-8290.CD-11-0341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Prahallad A, Sun C, Huang S, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012;483:100–3. 10.1038/nature10868 [DOI] [PubMed] [Google Scholar]
  • 21.Kopetz S, Grothey A, Yaeger R, et al. Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N Engl J Med 2019;381:1632–43. 10.1056/NEJMoa1908075 [DOI] [PubMed] [Google Scholar]
  • 22.Palmer AC, Sorger PK. Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy. Cell 2017;171:e13:1678–91. 10.1016/j.cell.2017.11.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pfizer Inc Pfizer provides update on phase 3 Pallas trial of IBRANCE® (palbociclib) plus endocrine therapy in HR+, HER2- early breast cancer, 2020. Available: https://wwwpfizercom/news/press-release/press-release-detail/pfizer-provides-update-phase-3-pallas-trial-ibrancer
  • 24.Dickler MN, Tolaney SM, Rugo HS, et al. MONARCH 1, a phase II study of Abemaciclib, a CDK4 and CDK6 Inhibitor, as a single agent, in patients with refractory HR+/HER2- metastatic breast cancer. Clin Cancer Res 2017;23:5218–24. 10.1158/1078-0432.CCR-17-0754 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Carlisle BG, Doussau A, Kimmelman J, Benefit KJ. Benefit, burden, and impact for a cohort of post-approval cancer combination trials. Clin Trials 2020;17:18–29. 10.1177/1740774519873883 [DOI] [PubMed] [Google Scholar]
  • 26.Gyawali B, Prasad V. Drugs that lack single-agent activity: are they worth pursuing in combination? Nat Rev Clin Oncol 2017;14:193–4. 10.1038/nrclinonc.2017.27 [DOI] [PubMed] [Google Scholar]
  • 27.Coleman RL, Sill MW, Bell-McGuinn K, et al. A phase II evaluation of the potent, highly selective PARP inhibitor veliparib in the treatment of persistent or recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer in patients who carry a germline BRCA1 or BRCA2 mutation - An NRG Oncology/Gynecologic Oncology Group study. Gynecol Oncol 2015;137:386–91. 10.1016/j.ygyno.2015.03.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Pahuja S, Beumer JH, Appleman LJ, et al. Outcome of BRCA 1/2-mutated (BRCA+) and triple-negative, BRCA wild type (BRCA-wt) breast cancer patients in a phase I study of single-agent veliparib (V). JCO 2014;32:135 10.1200/jco.2014.32.26_suppl.135 [DOI] [Google Scholar]
  • 29.Lowery MA, Kelsen DP, Capanu M, et al. Phase II trial of veliparib in patients with previously treated BRCA-mutated pancreas ductal adenocarcinoma. Eur J Cancer 2018;89:19–26. 10.1016/j.ejca.2017.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kummar S, Wade JL, Oza AM, et al. Randomized phase II trial of cyclophosphamide and the oral poly (ADP-ribose) polymerase inhibitor veliparib in patients with recurrent, advanced triple-negative breast cancer. Invest New Drugs 2016;34:355–63. 10.1007/s10637-016-0335-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pietanza MC, Waqar SN, Krug LM, et al. Randomized, double-blind, phase II study of temozolomide in combination with either veliparib or placebo in patients with Relapsed-Sensitive or refractory small-cell lung cancer. J Clin Oncol 2018;36:2386–94. 10.1200/JCO.2018.77.7672 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chiorean EG, Guthrie KA, Philip PA, et al. Randomized phase II study of second-line modified FOLFIRI with PARP inhibitor ABT-888 (veliparib) (NSC-737664) versus FOLFIRI in metastatic pancreatic cancer (mPC): SWOG S1513. JCO 2019;37:4014 10.1200/JCO.2019.37.15_suppl.4014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Westin SN, Sill MW, Coleman RL, et al. Safety lead-in of the MEK inhibitor trametinib in combination with GSK2141795, an Akt inhibitor, in patients with recurrent endometrial cancer: an NRG Oncology/GOG study. Gynecol Oncol 2019;155:420–8. 10.1016/j.ygyno.2019.09.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kurzrock R, Patnaik A, Rosenstein L, et al. Phase I dose-escalation of the oral MEK1/2 inhibitor GSK1120212 (GSK212) dosed in combination with the oral Akt inhibitor GSK2141795 (GSK795). J Clin Oncol 2011;29:3085 10.1200/jco.2011.29.15_suppl.308521383288 [DOI] [Google Scholar]
  • 35.Juric D, Soria J-C, Sharma S, et al. A phase 1B dose-escalation study of BYL719 plus binimetinib (MEK162) in patients with selected advanced solid tumors. JCO 2014;32:9051 10.1200/jco.2014.32.15_suppl.9051 [DOI] [Google Scholar]
  • 36.Tolcher AW, Patnaik A, Papadopoulos KP, et al. Phase I study of the MEK inhibitor trametinib in combination with the Akt inhibitor afuresertib in patients with solid tumors and multiple myeloma. Cancer Chemother Pharmacol 2015;75:183–9. 10.1007/s00280-014-2615-5 [DOI] [PubMed] [Google Scholar]
  • 37.Hoos A, Parmiani G, Hege K, et al. A clinical development paradigm for cancer vaccines and related biologics. J Immunother 2007;30:1–15. 10.1097/01.cji.0000211341.88835.ae [DOI] [PubMed] [Google Scholar]
  • 38.Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nat Immunol 2002;3:611–8. 10.1038/ni0702-611 [DOI] [PubMed] [Google Scholar]
  • 39.Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010;28:3167–75. 10.1200/JCO.2009.26.7609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bargou R, Leo E, Zugmaier G, et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 2008;321:974–7. 10.1126/science.1158545 [DOI] [PubMed] [Google Scholar]
  • 41.Evens AM, Hong FX, Habermann TM, et al. A 3-Arm randomized phase II trial with Bendamustine/Rituximab therapy in untreated high risk (HR) follicular lymphoma (fl): bortezomib induction or novel IMiD (R) continuation (bionic) study from the ECOG-ACRIN cancer research Group. Blood 2017;130:482. [Google Scholar]
  • 42.Hussain M, Daignault-Newton S, Twardowski PW, et al. Targeting androgen receptor and DNA repair in metastatic castration-resistant prostate cancer: results from NCI 9012. J Clin Oncol 2018;36:991–9. 10.1200/JCO.2017.75.7310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Blum KA, Polley M-Y, Jung S-H, et al. Randomized trial of ofatumumab and bendamustine versus ofatumumab, bendamustine, and bortezomib in previously untreated patients with high-risk follicular lymphoma: CALGB 50904 (Alliance). Cancer 2019;125:3378–89. 10.1002/cncr.32289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Tew WP, Sill MW, Walker JL, et al. Randomized phase II trial of bevacizumab plus everolimus versus bevacizumab alone for recurrent or persistent ovarian, fallopian tube or peritoneal carcinoma: an NRG oncology/gynecologic Oncology Group study. Gynecol Oncol 2018;151:257–63. 10.1016/j.ygyno.2018.08.027 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary data

esmoopen-2020-000871supp001.pdf (112.7KB, pdf)

RESOURCES