Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Dec 15.
Published in final edited form as: Cancer Res. 2014 Oct 17;74(24):7181–7184. doi: 10.1158/0008-5472.CAN-14-2329

Unique Molecular Landscapes in Cancer: Implications for Individualized, Curated Drug Combinations

Jennifer Wheler 1, J Jack Lee 2, Razelle Kurzrock 3
PMCID: PMC4292868  NIHMSID: NIHMS637171  PMID: 25326492

Abstract

With increasing lysophisticated technologies in molecular biology and ‘omic’ platforms to analyze patients’ tumors, more molecular diversity and complexity in cancer are being observed. Recently, we noted unique genomic profiles in a group of patients with metastatic breast cancer based on an analysis with next generation sequencing (NGS); amongst 57 consecutive patients, no two had the same molecular portfolio (1). Applied genomics therefore appears to represent a disruptive innovation in that it unveils a heterogeneity to metastatic cancer that may be ill suited to canonical clinical trials and practice paradigms. Upon recognizing that patients have unique tumor landscapes, it is possible that there may be a ‘mismatch’ between our traditional clinical trials system that selects patients based on common characteristics in order to evaluate a drug (drug-centric approach), and optimal treatment based on curated, individualized drug combinations for each patient (patient-centric approach).

Keywords: Clinical trials, Genomics, Personalized medicine


The canonical approach to cancer research and treatment is a drug-centric one. Specifically, for the most part, new therapeutics have been developed in large groups of patients who share a common histology based on the organ of origin of the cancer. The success of this strategy for patients with metastatic cancer has been modest. Recently, a more refined tactic has been instituted. Instead of grounding the classification system on the organ of origin or histology alone, new nosologies based on underlying molecular drivers or pathways, mostly as subsets of classic histologies, are being tested (24). The latter strategy combines a companion diagnostic that selects a subgroup of patients with a specific cancer by virtue of their harboring a specific biomarker which can be targeted by the agent under study. For example, a clinical trial may select patients with melanoma who have a BRAF mutation to enroll on a trial of a BRAF inhibitor (4, 5). This is a rational approach to drug development conceptually and has also resulted in impressive response rates in clinical testing (5, 6). Beyond this first generation advance of combining companion diagnostics and drugs within histologies, are even more novel studies, known as “basket” or “bucket” or “histology-agnostic” trials, that select patients by the presence of the biomarker (e.g. BRAF mutations), regardless of the organ of origin of the cancer, for treatment with cognate inhibitors (e.g., BRAF inhibitors).

Although genomic selection has shown remarkably high response rates in metastatic cancer (3, 4, 7), inevitably most patients relapse, often within months, and succumb to their disease. The foundation of these approaches (both the canonical histology-based approach and the new genomic one) is finding a commonality amongst patients in order to test a drug or combination of drugs. Ultimately, therefore, these are drug-centric strategies. Yet, the new reality unveiled by genomics –that each patient with metastatic disease has a complex and, often, unique set of genomic aberrations--suggests that ideal management of metastatic cancer may require a new model (8, 9). Several approaches to this problem have been suggested (10). The classic one entails finding common pathway themes, even amongst the diversity of cancer aberrations (11, 12). Indeed, several investigators have shown that, even though patients may have diverse genomic aberrations, the pathways activated may converge (11, 12). Finding crucial oncogenic hubs that are held in common between patients therefore might be critical. However, it is possible that despite the convergence of pathways, as full genomic sequencing is supplemented by transcriptomics, proteomics and more, amplified diversity may be identified. Furthermore, it is increasingly clear that, even when a single pathway is activated, the precise aberration may have crucial implications for therapy. For instance, different epidermal growth factor receptor (EGFR) abnormalities, while all activating the EGFR machinery, may respond to distinct therapeutic molecules. In general, the exon 19 aberrations such as L747_A750del that delete the four amino acids LREA, as well as the L834R exon 21 mutation, are sensitive to the reversible epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors, including erlotinib and gefitinib (13, 14). In contrast, in-frame insertions and/or duplications of 3 to 21 base pairs clustered between codons 767 and 774 in EGFR exon 20 are resistant to both reversible and irreversible TKIs; T790M exon 20 mutations are also resistant to some irreversible TKIs such as afatinib in the clinic (1517). The precise effects of these diverse aberrations on protein structure/function and, hence, therapeutic actionability may require a precise understanding of three-dimensional protein crystal structure and function.

With rare exceptions, patients with advanced disease will fail to achieved urable responses with even targeted drug regimens. Resistance emerges through a variety of mechanisms (2, 18, 19), as clones of tumor cells (either with de novo or acquired resistance) evolve. Rational combination approaches selected based on current bioinformatic and molecular analytic approaches, tailored for each individual based on their unique molecular profiles, and curated to prevent the development of resistance, represent a solution that warrants exploration. However, logistically, this approach represents several challenges, since it fails to conform to the historic framework created for clinical trials and practice. Most importantly, if the complexity revealed by genomics suggests that patients with metastatic tumors need customized therapy, it is a patient-centric, rather than drug-centric approach that might provide the highest yield. A patient-centric approach would require tailoring combinations of agents to the precise portfolio of abnormalities seen in an individual patient’s cancer. Indeed, as our computing and bioinformatics powers grow, it may be also feasible to take into consideration host genomic and other characteristics, and perform in silico prediction of which drugs would not only maximize response but also minimize toxicity for a particular patient.

Current approaches to combination therapy in oncology are based on lessons learned from the cytotoxic era. Classic chemotherapy has serious toxicity, and therefore each time two or more drugs were combined, they needed to be tested in the controlled environment of a clinical trial. However, applying this approach to targeted individualized combinations presents significant obstacles. For instance, if there are about 300 drugs available to oncologists (about 150 that are approved by the FDA (20) and another 150 that are in advanced investigational use or could be repurposed from other fields), the number of two drug combinations is 44,850; the number of three drug combinations 4,455,100. If we continue to evaluate drugs on a combination-by-combination basis – the drug-centric approach – would require over 1,000 years to test possible combinations based on currently available targeted agents. Not only is the timeline for evaluating these combinations untenable, but the cost of conducting the needed number of clinical trials to evaluate each possible combination of drugs would be astronomical. Therefore, the risk: benefit ratio of de novo tailored combinations need to be weighed against the risk:benefit ratio of continuing our current paradigms. Can new drug combinations be given safely without extensive testing? In this regard, a few other factors should be considered. First, non-oncology drugs are routinely combined, with an excellent safety record, without a priori testing in a clinical trial. For instance, the average cancer patient is already on a median of 8 drugs (21) (perhaps including an antidiabetic, antidepressant, antibiotic, etc.) when they are treated, and anti-neoplastics are added, without formal safety testing for the combinations. Our understanding of molecular pathways, CYP metabolic enzymes and so forth help inform our safe use of these combinations. These and additional parameters gleaned from assessing hundreds or thousands of drug combinations already performed could form the basis of guidelines for de novo new combinations. Although this approach may not be risk free, the risk:benefit ratio of moving forward would need to be weighed against that of continuing the current system of drug by drug, combination by combination testing. Finally, rules of thumb have been found for phase I drug testing (e.g., 1/10 of the lethal dose in 10% of animals (LD10) for mice for initial dosing) and have resulted in strong safety records (22, 23). The question that arises is could modern targeted agents therefore be safely combined in this way as well, that is with the use of a “rule of thumb,” since they are generally well tolerated, and considerable knowledge about their metabolism is available.

In conclusion, genomics has unveiled a new complexity to metastatic cancer. Despite emerging genomic-based clinical protocols, mainly focused on single companion diagnostics and drugs, we may still be trying to ‘retrofit’ new knowledge on an old clinical trials system (i.e., treating groups of patients by seeking common characteristics so that a drug or test can be validated). Suggested solutions have included finding convergence hubs (24, 25) and/or moving to earlier disease that might be less complex (26). However, though convergence is attractive, it is plausible that complexity is considerably greater than a small number of pathways. Indeed, within a single pathway, individual mutations may result in gene product conformational changes that are amenable to treatment by vastly different drugs. Further, our current genomic platforms may be assessing just the tip of the iceberg of malignant complexity, which could be significantly amplified when we interrogate tumors by not only genomics, but also transcriptomics, proteomics, epigenetics, and more.

A new patient-centric model for clinical research and treatment may need consideration. This model would involve testing the “strategy” of N-of-one molecular matching with customized combinations. Each patient would be treated with a consistent strategy, that is molecular matching, but with a differing combination of tailored drugs selected by that strategy. Further, rather than locking down the knowledge base, specific information on molecular profiles, treatments, and outcomes for each patient would be collected in a database, and constantly updated as new information is obtained in real time. Statistical methods such as the Bayesian hierarchical modelling, individual patient data meta-analysis, and various regression and classification methods for analyzing heterogeneity of treatment effect could be applied to identify effective treatment/marker pairs (2729). Consequently, the most currently updated information would be deployed to guide the treatment choice of subsequent patients. If the strategy proved successful and properly validated, it could then be extended to new patients and new combinations previously untested.

Genomics represent a disruptive technology that has revealed an underlying complexity to cancer that may not fit well with our current approach to clinical research and therapy. Our current models have yielded incremental steps forward, but most patients who develop metastatic cancer still die, treatments have considerable toxicities, and the costs of cancer care is substantial and soaring. Current omics technology suggests that patients with metastatic malignancies each or for the most part have unique tumors, and optimized therapy will require an individualized approach. This reality may require moving from a drug-centric to a patient-centric approach that validates an N-of-One strategy and then applies it to new patients (Table 1). To maximize benefit, a heuristic tactic for new individualized regimens will need to be created and rules established based on historic precedent, mathematical probabilities and known molecular pathways. Such a system represents a departure from old paradigms of clinical research and practice and is not risk free. However, the complexity of cancer may not be resolvable by adhering to traditional paradigms, and ultimately the risk:benefit ratio of the status quo versus new approaches will need to be ascertained.

Table 1.

Key Changes Needed to Implement Customized Combination Therapy

Current Model Patient-centric model with customized combinations Oversight areas that would be affected Comment
Diagnosis Patients are grouped together based on a histologic or molecular characteristic in order to be treated with a drug or combination of drugs Combinations of drugs are tailored to each patient’s molecular portfolio Internal review boards, guidelines for practice, payors, Food and Drug Administration (FDA)
Therapy Decisions/Molecular Tests Histologic diagnosis and/or companion diagnostics of single genes are used to determine treatment Panel of genes are interrogated in order to determine treatment CLIA, FDA
Molecular Tests Tests are locked down and cannot be changed after approval Tests are dynamic and incorporate new knowledge as it develops CLIA, FDA
Safety of Therapy Safety of new combinations of cancer drugs are determined by Phase I testing Safety of new combinations of drugs are determined by validated algorithms Internal review boards, guidelines for practice, payors, FDA Combinations of drugs without phase I are already routinely given to patients with multiple illnesses aside from cancer
Clinical Research The safety and efficacy of drugs or combinations are tested.

The drug or combination of drugs is the same for all patients on the trial
The safety and efficacy of a strategy is tested (strategy is molecular matching with customized combinations).
While the strategy remains consistent between patients, the drugs may be different for the patients on the trial
Internal review boards, guidelines for practice, payors, FDA Novel statistical methods will be needed
Drug approval Safety and efficacy in large randomized trials needed in histologically defined disease Safety data needed. Trials for efficacy may be small and approval may not be limited by histology FDA
Access to drugs Patients can access drugs after approval based on large randomized trials Patients will be able to access drugs if they meet requirement that strategy of matching has been used to choose the drugs FDA, payors

Acknowledgments

Financial Support: Funded in part by the Joan and Irwin Jacobs Fund and MyAnswerToCancer philanthropic fund.

References

  • 1.Wheler JJ, Parker BA, Lee JJ, Atkins JT, Janku F, Tsimberidou AM, et al. Unique molecular signatures as a hallmark of patients with metastatic breast cancer: Implications for current treatment paradigms. Oncotarget. 2014 doi: 10.18632/oncotarget.1946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012;367:1694–703. doi: 10.1056/NEJMoa1210093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Shaw AT, Kim DW, Nakagawa K, Seto T, Crino L, Ahn MJ, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013;368:2385–94. doi: 10.1056/NEJMoa1214886. [DOI] [PubMed] [Google Scholar]
  • 4.Sosman JA, Kim KB, Schuchter L, Gonzalez R, Pavlick AC, Weber JS, et al. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med. 2012;366:707–14. doi: 10.1056/NEJMoa1112302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Falchook GS, Long GV, Kurzrock R, Kim KB, Arkenau TH, Brown MP, et al. Dabrafenib in patients with melanoma, untreated brain metastases, and other solid tumours: a phase 1 dose-escalation trial. Lancet. 2012;379:1893–901. doi: 10.1016/S0140-6736(12)60398-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–16. doi: 10.1056/NEJMoa1103782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Tsimberidou AM, Iskander NG, Hong DS, Wheler JJ, Falchook GS, Fu S, et al. Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative. Clin Cancer Res. 2012;18:6373–83. doi: 10.1158/1078-0432.CCR-12-1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–92. doi: 10.1056/NEJMoa1113205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502:333–9. doi: 10.1038/nature12634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Stewart DJ, Batist G. Redefining Cancer: A new paradigm for better and faster treatment innovation. J Popul Ther Clin Pharmacol. 2014;21:e56–e65. [PubMed] [Google Scholar]
  • 11.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 12.Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med. 2004;10:789–99. doi: 10.1038/nm1087. [DOI] [PubMed] [Google Scholar]
  • 13.He M, Capelletti M, Nafa K, Yun CH, Arcila ME, Miller VA, et al. EGFR exon 19 insertions: a new family of sensitizing EGFR mutations in lung adenocarcinoma. Clin Cancer Res. 2012;18:1790–7. doi: 10.1158/1078-0432.CCR-11-2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Shan Y, Eastwood MP, Zhang X, Kim ET, Arkhipov A, Dror RO, et al. Oncogenic mutations counteract intrinsic disorder in the EGFR kinase and promote receptor dimerization. Cell. 2012;149:860–70. doi: 10.1016/j.cell.2012.02.063. [DOI] [PubMed] [Google Scholar]
  • 15.Arcila ME, Nafa K, Chaft JE, Rekhtman N, Lau C, Reva BA, et al. EGFR exon 20 insertion mutations in lung adenocarcinomas: prevalence, molecular heterogeneity, and clinicopathologic characteristics. Mol Cancer Ther. 2013;12:220–9. doi: 10.1158/1535-7163.MCT-12-0620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Arcila ME, Oxnard GR, Nafa K, Riely GJ, Solomon SB, Zakowski MF, et al. Rebiopsy of lung cancer patients with acquired resistance to EGFR inhibitors and enhanced detection of the T790M mutation using a locked nucleic acid-based assay. Clin Cancer Res. 2011;17:1169–80. doi: 10.1158/1078-0432.CCR-10-2277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kim Y, Ko J, Cui Z, Abolhoda A, Ahn JS, Ou SH, et al. The EGFR T790M mutation in acquired resistance to an irreversible second-generation EGFR inhibitor. Mol Cancer Ther. 2012;11:784–91. doi: 10.1158/1535-7163.MCT-11-0750. [DOI] [PubMed] [Google Scholar]
  • 18.Bean J, Brennan C, Shih JY, Riely G, Viale A, Wang L, et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci U S A. 2007;104:20932–7. doi: 10.1073/pnas.0710370104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005;2:e73. doi: 10.1371/journal.pmed.0020073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.FDA. FDA Drug Approvals and Databases. 2013 Oct 18; [cited 2013; Available from: http://www.fda.gov/drugs/InformationOnDrugs/default.htm.
  • 21.Borad MJ, Curtis KK, Babiker HM, Benjamin M, Tibes R, Ramanathan RK, et al. The impact of concomitant medication use on patient eligibility for phase I cancer clinical trials. J Cancer. 2012;3:345–53. doi: 10.7150/jca.4714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Horstmann E, McCabe MS, Grochow L, Yamamoto S, Rubinstein L, Budd T, et al. Risks and benefits of phase 1 oncology trials, 1991 through 2002. N Engl J Med. 2005;352:895–904. doi: 10.1056/NEJMsa042220. [DOI] [PubMed] [Google Scholar]
  • 23.Kurzrock R, Benjamin RS. Risks and benefits of phase 1 oncology trials, revisited. N Engl J Med. 2005;352:930–2. doi: 10.1056/NEJMe058007. [DOI] [PubMed] [Google Scholar]
  • 24.Saini KS, Loi S, de Azambuja E, Metzger-Filho O, Saini ML, Ignatiadis M, et al. Targeting the PI3K/AKT/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer. Cancer Treat Rev. 2013;39:935–46. doi: 10.1016/j.ctrv.2013.03.009. [DOI] [PubMed] [Google Scholar]
  • 25.Axelrod M, Gordon VL, Conaway M, Tarcsafalvi A, Neitzke DJ, Gioeli D, et al. Combinatorial drug screening identifies compensatory pathway interactions and adaptive resistance mechanisms. Oncotarget. 2013;4:622–35. doi: 10.18632/oncotarget.938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Westin JR, Kurzrock R. It’s about time: lessons for solid tumors from chronic myelogenous leukemia therapy. Mol Cancer Ther. 2012;11:2549–55. doi: 10.1158/1535-7163.MCT-12-0473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Duan N, Kravitz RL, Schmid CH. Single-patient (n-of-1) trials: a pragmatic clinical decision methodology for patient-centered comparative effectiveness research. J Clin Epidemiol. 2013;66:S21–8. doi: 10.1016/j.jclinepi.2013.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Willke RJ, Zheng Z, Subedi P, Althin R, Mullins CD. From concepts, theory, and evidence of heterogeneity of treatment effects to methodological approaches: a primer. BMC Med Res Methodol. 2012;12:185. doi: 10.1186/1471-2288-12-185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zucker DR, Ruthazer R, Schmid CH. Individual (N-of-1) trials can be combined to give population comparative treatment effect estimates: methodologic considerations. J Clin Epidemiol. 2010;63:1312–23. doi: 10.1016/j.jclinepi.2010.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES