Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jan 1.
Published in final edited form as: Surg Oncol Clin N Am. 2019 Oct 29;29(1):35–49. doi: 10.1016/j.soc.2019.08.003

Genomics and the History of Precision Oncology

Deborah B Doroshow 1, James H Doroshow 2
PMCID: PMC6878897  NIHMSID: NIHMS1538295  PMID: 31757312

Introduction

The use of molecular characterization of an individual patient’s tumor in routine oncologic practice began only 20 years ago. In that short time, it has enhanced the specificity and efficacy of cancer therapy.1 Molecular selection criteria, both genomic and protein-based, have now been used to support the first histology-independent FDA approvals of anticancer agents.27 This paradigmatic shift in oncologic practice has been accompanied by discontinuation of nonspecific cytotoxic anticancer agent development.8

The use of precision medicine principles in cancer therapy9 depends upon measurement of biologic characteristics in a tumor that suggest the potential value of a specific molecularly-targeted treatment. This shift away from nonspecific mechanisms of tumor cell killing has occurred because of improvements in biomarker discovery and validation, and the availability of instrumentation capable of previously-inconceivable levels of diagnostic throughput. Precision oncology has also advanced because of innovations in clinical trial design.1012

While precision oncology has changed both the landscape of treatment options for patients with cancer and the fabric of clinical and translational research, cytotoxic agents remain the backbone of therapy for the majority of cancers, and targeted agents rarely provide durable responses. Moreover, innovative clinical trial designs to evaluate novel targets remain in the early stages of development, engendering numerous operational challenges11 and modest clinical benefit to date.12,13 This paper examines the history of precision oncology, including milestone developments in therapeutics, translational science, and clinical trial design over the last 20 years.

Several case studies illustrate the benefits and limitations of the “one gene, one drug, one disease” paradigm. While clinical responses may be common and dramatic, they are rarely lasting. Moreover, this model does not apply to the majority of neoplastic diseases. These case studies also illustrate the ways in which targeted therapies have unexpected effects, and frequently transform the natural history of disease. The identification and targeting of molecular aberrations requires sufficiently efficient and accurate technologies, many of which have become more sensitive and comprehensive over the last ten years. Finally, master protocols have been developed to provide a coordinated framework for evaluation of multiple therapeutic approaches in one or more molecularly-defined tumor types, with the goal of improving the efficiency of the cancer clinical trials process. Such studies can be designed to provide sufficient information to support an application for new drug approval by the FDA, or, more frequently, to identify biomarker-selected drugs that can be more effectively predicted to be successful in the setting of a subsequent, definitive randomized study.

The “Magic Bullet” Paradigm in Precision Oncology, 2000-2010

Most retellings of the beginnings of precision oncology focus on the development of a series of drugs, each of which was intended to treat a single tumor type with a single molecular aberration. Each were heralded by physicians and by the lay public as near-miracles of science. While these developments were critical to the development of precision oncology, they were often unpredictable and uniformly never sudden. We discuss them here not as a means of uncritically listing a series of successes, but as a window into the promises and limitations of precision oncology more broadly.

In the late 1980s, Dennis Slamon and his colleagues at the University of California, Los Angeles demonstrated that one quarter of breast cancers could be characterized by amplification or overexpression of human epidermal growth factor-2 (HER2), a tyrosine kinase receptor which activates multiple signal transduction pathways to regulate cell growth.14,15 Moreover, patients harboring such tumors had a poorer prognosis than those who did not.14,15 Simultaneously, multiple investigators determined that HER2 overexpression induced tumorigenesis, making the protein a desirable therapeutic target.1618 A humanized HER2 antibody, engineered at Genentech in 1992,19 was quickly introduced into multiple phase I and phase II clinical trials, the latter of which focused on enrolling only patients with increased HER2 expression by immunohistochemistry.20 However, the definition of increased HER2 expression was inconsistent; one trial enrolled patients with >25% membrane staining, a second enrolled those whose tumors had “light to strong” IHC staining, and a third used the now-standard 2+/3+ nomenclature to refer to weak or complete tumor cell membrane staining in >10% of cells.20

Despite modest activity in a placebo-controlled phase 3 study (using the latter definition of HER2 positivity) trastuzumab (Herceptin®) was approved by the FDA in 1998.21 Trastuzumab was hailed by the popular press as “ushering in a new era of cancer treatment that attempts to target the very flawed genetic mechanisms that cause the disease.”22 The Washington Post described how one patient’s impending death was dramatically reversed by Herceptin, which shrunk her liver metastases “to mere dots” and showed “how 20 years of basic cancer research are beginning to pay off for patients.”23 Yet the popular press also reminded readers that while trastuzumab appeared to be a wonder drug, it was not a cure.22

While trastuzumab did not dramatically improve outcomes as a single agent, it has become a critical, life-prolonging adjunct to chemotherapy in the metastatic,24 neoadjuvant,25 and adjuvant26 settings. The identification of HER2 as a valuable therapeutic target spurred additional work resulting in the development of trastuzumab emtansine, a novel antibody-drug conjugate targeting HER2,27 and pertuzumab, a monoclonal antibody binding a different epitope of HER2.28 Perhaps most intriguingly, trastuzumab has changed the natural history of HER2-positive breast cancer. Whereas Slamon and colleagues initially identified HER2 overexpression as a poor prognostic marker, patients who are treated with HER2-directed therapies may no longer be at a survival disadvantage. Now, HER2 overexpression has become a predictive biomarker, indicative of a subset of patients likely to respond to HER2-directed therapy.

Three years after trastuzumab was approved by the FDA, another landmark drug was introduced: imatinib for the treatment of chronic myeloid leukemia (CML). The press similarly described the drug as a ‘magic bullet’, proclaiming that “the dream of a pill that can treat cancer with almost no side effects became a reality” and referring to targeted drugs as “smart bombs” that did not induce the collateral damage seen with cytotoxic chemotherapy.29 Patients who had participated in clinical trials spread the news in online chat rooms before trial results had even been published.30 This enthusiasm was understandable. The phase I study of imatinib, a small molecule inhibitor of the BCR-ABL tyrosine kinase, had identified no maximally tolerated dose (MTD), and 53 of 54 patients achieved a complete hematologic response;31 based on these results as well as those of three phase II studies, imatinib was granted accelerated approval by the FDA in 2001. The 72 days required for FDA review was the fastest agency approval in the history of anticancer agent development.32

Despite the perception of a sudden breakthrough, the story of imatinib illustrates the lengthy research timeframe involved in identifying a molecular target and crafting a drug to engage it. While CML had been described as a disease entity in Germany and Scotland in 1845,33 the majority of work done to identify the BCR-ABL fusion as the causative aberration in CML was performed between 1960 and 1990.34 In 1960, Peter Nowell and David Hungerford described a “minute chromosome” present in the peripheral blood of 7 patients with what was then known as chronic granulocytic leukemia.35 In the early 1970s, Caspersson et al. and O’Riordan et al. identified the abnormally small chromosome as number 22 using novel quinacrine mustard fluorescence techniques,36,37 and Janet Rowley described its balanced translocation with the long arm of chromosome 9.38 In the 1970s and 1980s, work with retroviruses was critical to the identification of multiple putative oncogenes; one of these was c-abl, the human cellular homologue of the Abelson murine leukemia virus, which was translocated from 9q to the breakpoint cluster region (BCR) of 22q- in patients with CML.34 In the mid-1980s, Davis et al.39 and Ben Neriah et al.40 discovered that the resulting chimeric mRNA transcript was itself a tyrosine kinase, and murine work published by Daley et al.41 and Heisterkamp et al.42 in 1990 identified the BCR-ABL translocation as necessary and sufficient to induce CML.

Thus, by 1990, the BCR-ABL oncogene had been established as universally causative in CML; hence, the resulting tyrosine kinase was an attractive therapeutic target. Yet, general skepticism remained regarding the feasibility of inhibiting tyrosine kinases, with regard to the specificity, toxicity, and efficacy of doing so in heterogeneous cancers.34 High throughput screens of chemical libraries identified the 2-phenylaminopyrimidines as promising inhibitors of BCR-ABL in the early 1990s, and by 1996 Brian Druker and colleagues had published in vitro and in vivo data demonstrating that one such agent - STI571, or imatinib - potently inhibited the BCR-ABL kinase and killed CML cells.34 Five years later, STI571 had been approved by the FDA. While the development of imatinib had itself been brisk, the identification of BCR-ABL as a therapeutic target was an effort spanning more than three decades, an element of the imatinib story that is easy to overlook.34

The history of imatinib has been unusual in other ways, one of which is the durability of the responses produced by the drug. Resistance to therapy does develop and has spurred the introduction of second- and third-line agents. Still, patients with CML who are treated with BCR-ABL tyrosine kinase inhibitors can expect to live near-normal lifespans.43 The introduction of these drugs, five of which are now FDA-approved, has altered the natural history of CML to the extent that the field is now exploring the potential of therapy discontinuation.44

While the importance of identifying a molecular target prior to developing a relevant therapy may appear obvious, in many prominent cases a target has been identified and refined during the course of drug development. For the epidermal growth factor receptor (EGFR), similar developments occurred in the development of EGFR-targeted agents in both lung and colorectal cancer. Initial work on EGFR, which was known to play an important role in modulating proliferative cell signaling, was based on its overexpression in multiple tumor types, including non-small cell lung cancer (NSCLC).45 However, phase I trials of the EGFR tyrosine kinase inhibitor (TKI) gefitinib produced few clinical responses.4648 Two phase II studies of gefitinib in NSCLC were slightly more promising, with response rates of 9-19%.49,50 However, a retrospective analysis of tumor specimens from these two trials found no relationship between EGFR expression (as determined by immunohistochemistry) and clinical response.51 Based on the results of these trials, gefitinib was granted accelerated approval by the FDA in 2003.52 However, review by the FDA also noted the negative, unpublished results of two phase III studies that failed to show clinical benefit of gefitinib in combination with chemotherapy in patients with advanced NSCLC.5254

Several groups of investigators sought to characterize responders further by analyzing tumor specimens from patients entered on phase II studies as well as those treated during expanded access programs. They identified several subgroups of patients with higher response rates: Japanese patients (compared to non-Japanese patients),50 women,49,55 never smokers, 56,57 and patients with adenocarcinoma histologies.5557 Simultaneously, several groups of investigators sequenced the EGFR gene in lung cancer specimens from patients who had been treated with gefitinib on clinical trials. They identified mutations in the EGFR tyrosine kinase domain in almost all tumors from patients who had responded, which were not present in nonresponders, establishing mutated EGFR - not overexpressed EGFR - as the molecular target for gefitinib.5759

While EGFR-targeted therapies have become a success story in lung cancer, their evolving history is more representative of targeted therapies than that of imatinib, in that resistance to treatment is inevitable. Over the last ten years, studies have focused on the development of second and third generation EGFR inhibitors which specifically target mechanisms of resistance to first generation inhibitors such as the T790M mutation in exon 21 of EGFR.6064 Currently, the effort to characterize mechanisms of resistance to third generation inhibitors is ongoing with the aim of developing therapies that target these alterations or prevent their emergence.65 Patients starting first-line osimertinib, a third-generation EGFR TKI, may respond for up to 22 months.66 However, upon progression they are faced with the options of cytotoxic chemotherapy and immune checkpoint inhibitors, the latter of which have reduced efficacy in EGFR mutant NSCLC.6769

While resistance to targeted therapies may be due to so-called “on-target” molecular alterations, such as additional EGFR mutations, it often results from compensatory mechanisms, especially when a target is one member of a signaling pathway. This type of resistance proved an early stumbling block in the development of cetuximab, a competitive inhibitor of the extracellular domain of EGFR. Similar to gefinitib, cetuximab was studied in colon cancer based on the premise that EGFR was overexpressed in the majority of colorectal cancers, as well as promising preclinical data in colorectal cancer models.70 The first phase III study of cetuximab, which studied it alone and in combination with irinotecan, utilized this rationale and required evidence of immunohistochemical expression of EGFR to enroll.71 Seeking to better characterize cetuximab responders and nonresponders only a few years later, two French groups screened the tumors of clinical trial participants for mutations in KRAS, which was involved in EGFR downstream signaling. Not only did mutated KRAS predict for resistance to cetuximab, zero patients who responded to cetuximab had tumors with a KRAS mutation,7274 a finding confirmed in a larger retrospective study performed by an group from Australia and Canada.75 Extended RAS testing is now recommended to demonstrate that a tumor is truly wildtype prior to treating a patient with cetuximab in order to predict primary resistance.76

It is now clear that downstream resistance accounts for a substantial proportion of acquired resistance to targeted therapies, a lesson learned in the development of BRAF inhibitors for patients with metastatic melanoma. Although the BRAF inhibitors vemurafenib and dabrafenib both improved progression free survival (PFS) in patients with untreated BRAFV600E mutated advanced melanoma compared to chemotherapy in two phase 3 studies, responses were short-lived, with a median PFS of just over 5 months for both drugs.77,78 Additional pharmacodynamic studies performed on patient tumor specimens from these and other trials found that acquired resistance to BRAF inhibitors was frequently associated with up-regulation of signaling in the downstream MAPK pathway.7981 As a result, combinations of BRAF and MEK inhibitors were studied in multiple large phase 3 trials and were found to improve both PFS and overall survival (OS) compared to the use of BRAF inhibitors alone;8285 these combinations now comprise the standard of care therapy for patients with BRAF-mutated melanoma. These cases clearly demonstrate both the promise and the limitations of the “magic bullet” model of precision oncology, and have more recently led to efforts to identify promising combinations of targeted therapies.86

The Promise and Limitations of “Tumor Profiling”

To detect molecular alterations that can be targeted, reliable, and efficient technology is required. The early work on trastuzumab was limited to immunohistochemical staining, whereas fluorescent in situ hybridization (FISH) is now a routine component of HER2 testing for tumor samples that demonstrate equivocal (2+) IHC staining; this combined approach has changed the definition of “HER2 positive” tumors.87 In the 2000s and early 2010s, molecular alterations could be detected using either immunohistochemical evaluations of protein expression or PCR-based evaluations of mutational “hotspots,” which could miss uncommon alterations. In 2019, a patient’s tumor may undergo high throughput massively parallel DNA and RNA sequencing (often referred to as next generation sequencing, or NGS) over a matter of two or three weeks to identify potential therapeutic targets.88,89 These analyses, which may examine several hundred genes or even comprise whole exome or whole genome sequencing, are regularly performed on the tumors of patients treated at tertiary cancer centers and in the community. The latter was made more accessible due to the recent announcement from the Centers for Medicare and Medicaid Services that Medicare will cover next-generation tumor profiling for patients with advanced cancer.90

Genomic and proteomic analyses now have the capacity to detect a variety of aberrations beyond point mutations, including insertion and deletion mutations (indels), copy number alterations, chromosomal rearrangements and gene fusions, DNA methylation patterns, transcript levels, and levels of protein expression.89 Analysis of a patient’s tumor is often paired with evaluation of matched normal cells, most often from a buccal swab or peripheral blood, to distinguish somatic aberrations found only in a tumor from germline abnormalities.91 More recently, examinations of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) have been investigated as a means of dynamically and non-invasively assessing tumor burden as well as evaluating the changing genomic landscape of a tumor throughout a patient’s treatment, with an eye to better understanding mechanisms of drug resistance.9294

These advances have enabled the detection of low frequency alterations and have fostered the development of many new targeted therapies. However, complex challenges remain. Tumor profiling reports may list the molecular aberrations identified in a patient’s tumor, but only a minority at best may be “targetable” with approved or experimental agents. Such reports may neglect to describe the allelic frequency of such aberrations or distinguish between driver mutations - those which induce tumorigenesis - and passenger mutations, which are not themselves pathogenic.89,95,96 Moreover, tumor heterogeneity may limit the applicability of these findings.97,98 As in the case of BRAF inhibition in metastatic melanoma, efforts to target tumor molecular aberrations have been hampered by the almost universal development of on- or off-target resistance.95 This has led to a recent focus on evaluating combinations of agents to delay the emergence of such resistance, for example by inhibiting a signaling pathway at more than one level.82

Novel Methods for Evaluating Targeted Therapies Using “Master Protocols”

While technologies like NGS have facilitated the detection of targetable molecular alterations, novel clinical trial designs - many of which are still in the early phases of development - have become critical to testing targeted agents. In the early 2010s, investigations of “exceptional responders” to targeted therapies were common.99 In other instances, a patient’s response to a therapy that had been “matched” to a specific tumor alteration was compared to that patient’s response to a previous standard of care agent.100 Many academic cancer centers developed molecular tumor boards in which a group of experts reviewed the molecular alterations in a patient’s tumor and recommended a matched FDA-approved therapy, an off-label standard therapy, or a clinical trial.6,88 Such tumor boards are increasingly common in both the academic and community settings.

In efforts to evaluate the concept of precision oncology more broadly, several studies have attempted to assess whether “matched” therapy provides more clinical benefit than standard-of-care therapy. Multiple single-institution, observational studies have shown that it is feasible to match patients to both standard and investigational therapies, and that doing so may improve clinical outcomes. However, only a minority of patients could be assigned a matched therapy, and none of the trials were randomized.101105 SHIVA, the first randomized trial of precision oncology as an approach, randomized patients with multiple tumor histologies to receive either one of 11 molecularly targeted agents based on the presence or absence of aberrations in the hormone receptor, PI3K/AKT/mTOR, and RAF/MEK pathways or physician’s choice of standard therapy.106 While no difference in PFS was observed, the study used only a limited range of targeted therapies and did not account for differing levels of evidence regarding the relevance of each patient’s pathway aberration.

Master protocols, which permit the testing of patients with multiple tumor histologies and/or tumor molecular aberrations, are both powerful and complex frameworks used to test a variety of hypotheses simultaneously. By grouping tumors by molecular alteration, they move oncology toward a less histology-based and more molecularly-based diagnostic and clinical framework.13 Classically, master protocols have been described as falling into one of two categories: basket studies, which seek to treat patients across multiple histologies whose tumors share the same alteration, and umbrella studies, which assign patients with one tumor type to one of several therapies based on tumor profiling data.13 While such studies may be used for FDA registration of a new agent, they are more often signal-finding trials intended to identify potentially interesting therapies worthy of further study in certain patient populations.

Basket studies play a critical role in promoting a histology-agnostic approach to treating cancer. Two phase II studies of pembrolizumab, an anti-programmed death 1 (PD-1) antibody, in patients with mismatch repair (MMR)-deficient tumors demonstrated its efficacy across all MMR-deficient solid tumors107,108, and led to the first tissue agnostic approval of a drug by the FDA in 2017.109 The following year, a phase I/II basket study of larotrectinib in tumors with TRK-fusions led to the second such FDA approval in 2018.110 Basket trials are not always unmitigated successes, however. A study of vemurafenib in patients with advanced BRAFV600 mutated malignancies (exclusive of melanoma) found an overall response rate (ORR) of 42% of patients with NSCLC and 29% of those with anaplastic thyroid cancer,111 leading to FDA approvals for dabrafenib in patients with V600E-mutated NSCLC112 and anaplastic thyroid cancer.113 However, few or no responses were observed in multiple other tumor types examined. Currently, multiple clinical trials are ongoing to evaluate the use of inhibitors of DNA damage repair (DDR) in tumors with a variety of DDR-deficient mutations; preliminary results are encouraging114 but still evolving.115

Umbrella trials have, to date, primarily served as exploratory signal-finding studies. They often operate using adaptive designs, wherein new arms may be added based on new evidence or removed based on lack of response, and patients may be assigned to a therapy based on an algorithm that utilizes evolving data to account for that patient’s likelihood of response.11 For example, the BATTLE studies assigned patients with advanced NSCLC to treatment arms based on molecular profiling of their tumors using real-time analyses of on-trial biopsies.116,117

The terms “basket trial” and “umbrella trial” are useful heuristics but may not adequately describe all large platform precision oncology trials.118 A basket trial looking at patients whose tumors are DDR-deficient, for example, examines a group of functionally similar molecular alterations. The ongoing NCI-MATCH (Molecular Analysis for Therapy Choice) study uses on-study biopsies to assign patients with any histology to a broad range of therapies based on their tumor molecular alterations.119 While neither is a strict umbrella or basket trial, these hybrid platform studies enable the evaluation of multiple histologies and multiple mutations or other alterations.12

Master protocols present numerous challenges. They are time-consuming, require significant coordination among multiple stakeholders, and can be costly.13 Due to their complexity, master protocols provide a difficult format for sponsors hoping to achieve registration and create significant work for regulatory officials and institutional review boards faced with numerous amendments.11 They are rarely randomized, making it difficult to draw conclusions about the efficacy of an agent.118 Varying statistical designs may limit the ability of investigators to draw definitive conclusions. For example, some basket studies are designed as a series of Simon two-stage studies, treating each arm as a separate trial for statistical purposes and serving as a signal-finding study, whereas others allow aggregation of data from similar arms, which permits investigators to deem a therapy effective earlier.120

Despite these challenges, master protocols offer many opportunities for both patients and investigators. They enable patients with rare cancers to participate more readily in clinical trials and may lead to new therapeutic options. They efficiently group patients with multiple tumor types, are adaptable, and enable large collaborations.11,13 They also can provide access to laboratories performing validated assessments of specific, treatment-defining molecular alterations in patients’ tumors. Once established, the infrastructure for these trials can speed the screening of new therapeutic agents across a wide range of both common and understudied malignancies. However, platform trials are still in a relatively early stage of development.

Summary

In conclusion, rapid improvements in a variety of molecular characterization technologies over the past two decades have directly supported the development of new systemic cancer therapies that can be selected to target specific pharmacological vulnerabilities in select patient populations across a wide range of human cancers. Molecular matching of drugs to specific targets for individual patients has substantively improved treatment for many hematological malignancies and solid tumors. Although this approach has become widespread only over the past 5-10 years, it has provided clinical benefits for many patients whose malignancies heretofore lacked effective therapy. Furthermore, in light of continuing improvements in our understanding of tumor biology and the tumor microenvironment, as well as remarkably efficient chemical biology and immunologic approaches now available for the development of therapeutics, further improvements in our ability to optimize cancer treatment based on the characteristics of an individual patient’s tumor—the definition of precision oncology—are highly likely.

Key points:

  • The use of molecular tumor characteristics to select systemic therapy for individual patients has grown dramatically over the past 20 years

  • The identification of tumoral DNA abnormalities using rapid gene sequencing techniques has underpinned the discovery of treatments for certain patients independent of disease histology

  • Clinical trial designs using the ‘Master Protocol’ concept have facilitated the simultaneous evaluation of multiple new therapies based on matching drugs to specific genomic abnormalities

Synopsis.

Progress toward the implementation of a molecular characterization paradigm in cancer drug development over the past 20 years, reviewed in this paper, has markedly enhanced our capability to select patients who are more likely to benefit from cancer therapy. Dramatic improvements in genomic and related diagnostic testing platforms have simultaneously permitted evaluation of the efficacy of treatment assignment based on pre-defined biologic features of an individual patient’s tumor or germline using master protocols that may include many malignancies as well as their molecularly-characterized subsets. With this approach, a wide range of new targeted and immunologic treatment approaches have been defined for groups of patients who, heretofore, lacked effective therapeutic options.

Acknowledgments

Disclosure statement: Deborah Doroshow does consulting for Boehringer Ingelheim, Ipsen. James Doroshow has nothing to disclose. The article is supported by ZIA BC 011078; Phase 0/1 Clinical Trials from the National Cancer Institute (JHD); ASCO Young Investigator Award (DBD)

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Contributor Information

Deborah B. Doroshow, Department of Medicine and Cancer Center, Icahn School of Medicine at Mount Sinai, New York, NY, 1 Gustave Levy Place, New York, NY 10029.

James H. Doroshow, Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD.

References

  • 1.Doroshow JH. Precision medicine in oncology In: DeVita VT, Rosenberg SA, eds. Cancer: Principles and Practice of Oncology. 11th ed. Philadelphia: Wolters Kluwer; 2019:186–196. [Google Scholar]
  • 2.Sparano JA, Gray RJ, Makower DF, et al. Adjuvant chemotherapy guided by a 21-Gene expression assay in breast cancer. N Engl J Med. 2018;379:111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Shaw AT, Felip E, Bauer TM, et al. Lorlatinib in non-small-cell lung cancer with ALK or ROS1 rearrangement: an international, multicentre, open-label, single-arm first-in-man phase 1 trial. Lancet Oncol. 2017;18:1590–1599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359:1757–1765. [DOI] [PubMed] [Google Scholar]
  • 5.Khorashad JS, Kelley TW, Szankasi P, et al. BCR-ABL1 compound mutations in tyrosine kinase inhibitor-resistant CML: frequency and clonal relationships. Blood. 2013;121:489–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Board Editorial. Making precision oncology the standard of care. Lancet Oncology. 2017;18:835. [DOI] [PubMed] [Google Scholar]
  • 7.Drilon A, Laetsch TW, Kummar S, et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N Engl J Med. 2018;378:731–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.DeVita VT, Chu E. A history of cancer chemotherapy. Cancer Res. 2008;68:8643–8653. [DOI] [PubMed] [Google Scholar]
  • 9.Desmond-Hellmann S, Sawyers CL, Cox DR, Fraser-Liggett C. Toward Precision Medicine: Building a Knowledge Networkfor Biomedical Research and a New Taxonomy of Disease. Washington D.C.: National Academies Press; 2011. [PubMed] [Google Scholar]
  • 10.Kummar S, Williams PM, Lih C- J, et al. Application of molecular profiling in clinical trials for advanced metastatic cancers. JNatl Cancer Inst. 2015;107(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cecchini M, Rubin EH, Blumenthal GM, et al. Challenges with Novel Clinical Trial Designs: Master Protocols. Clin Cancer Res. January 2019:clincanres.3544.2018. [DOI] [PubMed] [Google Scholar]
  • 12.Eckhardt SG, Lieu C. Is Precision Medicine an Oxymoron? JAMA Oncol. November 2018. [DOI] [PubMed] [Google Scholar]
  • 13.Woodcock J, LaVange LM. Master Protocols to Study Multiple Therapies, Multiple Diseases, or Both. N Engl J Med. 2017;377:62–70. [DOI] [PubMed] [Google Scholar]
  • 14.Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235:177–182. [DOI] [PubMed] [Google Scholar]
  • 15.Slamon DJ, Godolphin W, Jones LA, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707–712. [DOI] [PubMed] [Google Scholar]
  • 16.Hudziak RM, Schlessinger J, Ullrich A. Increased expression of the putative growth factor receptor p185HER2 causes transformation and tumorigenesis of NIH 3T3 cells. Proc Natl AcadSci USA. 1987;84:7159–7163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Di Fiore PP, Pierce JH, Kraus MH, Segatto O, King CR, Aaronson SA. erbB-2 is a potent oncogene when overexpressed in NIH/3T3 cells. Science. 1987;237:178–182. [DOI] [PubMed] [Google Scholar]
  • 18.Guy CT, Webster MA, Schaller M, Parsons TJ, Cardiff RD, Muller WJ. Expression of the neu protooncogene in the mammary epithelium of transgenic mice induces metastatic disease. Proc Natl Acad Sci USA. 1992;89:10578–10582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Carter P, Presta L, Gorman CM, et al. Humanization of an anti-p185HER2 antibody for human cancer therapy. Proc Natl Acad Sci USA. 1992;89:4285–4289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Baselga J Clinical trials of Herceptin® (trastuzumab). Eur J Cancer. 2001;37:18–24. [PubMed] [Google Scholar]
  • 21.Cobleigh MA, Vogel CL, Tripathy D, et al. Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol. 1999;17:2639–2648. [DOI] [PubMed] [Google Scholar]
  • 22.Roan S Weapon in the War on Cancer. Los Angeles Times. October 26, 1998:S1. [Google Scholar]
  • 23.Neergaard L FDA Looks At Therapy For Cancer; Drug Attacks Gene Linked to Disease. Washington Post. August 31, 1998:A10. [Google Scholar]
  • 24.Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. NEngl J Med. 2001;344:783–792. [DOI] [PubMed] [Google Scholar]
  • 25.Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353:1673–1684. [DOI] [PubMed] [Google Scholar]
  • 26.Smith I, Procter M, Gelber RD, et al. 2-year follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: a randomised controlled trial. Lancet. 2007;369:29–36. [DOI] [PubMed] [Google Scholar]
  • 27.Doroshow DB, LoRusso PM. Trastuzumab emtansine: determining its role in management of HER2+ breast cancer. Future Oncol. 2018;14:589–602. [DOI] [PubMed] [Google Scholar]
  • 28.Gerratana L, Bonotto M, Bozza C, et al. Pertuzumab and breast cancer: another piece in the anti-HER2 puzzle. Expert Opin Biol Ther. 2017;17:365–374. [DOI] [PubMed] [Google Scholar]
  • 29.Peres J FDA OKs breakthrough leukemia pill. Chicago Tribune. May 11, 2001:1.26. [Google Scholar]
  • 30.Druker BJ. Perspectives on the development of imatinib and the future of cancer research. Nat Med. 2009;15:1149–1152. [DOI] [PubMed] [Google Scholar]
  • 31.Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344:1031–1037. [DOI] [PubMed] [Google Scholar]
  • 32.Cohen MH, Williams G, Johnson JR, et al. Approval summary for Imatinib Mesylate capsules in the treatment of chronic myelogenous leukemia. Clin Cancer Res. 2002;8(5):935–942. [PubMed] [Google Scholar]
  • 33.Goldman JM. Chronic Myeloid Leukemia: A Historical Perspective. Seminars in Hematology. 2010;47:302–311. [DOI] [PubMed] [Google Scholar]
  • 34.Druker BJ. Translation of the Philadelphia chromosome into therapy for CML. Blood. 2008;112:4808–4817. [DOI] [PubMed] [Google Scholar]
  • 35.Nowell PC, Hungerford DA. National Academy of Sciences: A minute chromosome in human chronic granulocytic leukemia. Science. 1960;132:1488–1501.17739576 [Google Scholar]
  • 36.Caspersson T, Gahrton G, Lindsten J, Zech L. Identification of the Philadelphia chromosome as a number 22 by quinacrine mustard fluorescence analysis. Exp Cell Res. 1970;63:238–240. [DOI] [PubMed] [Google Scholar]
  • 37.O’Riordan ML, Robinson JA, Buckton KE, Evans HJ. Distinguishing between the chromosomes involved in Down’s Syndrome (Trisomy 21) and chronic myeloid leukaemia (Ph 1) by fluorescence. Nature. 1971;230:167. [DOI] [PubMed] [Google Scholar]
  • 38.Rowley JD. Letter: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature. 1973;243:290–293. [DOI] [PubMed] [Google Scholar]
  • 39.Davis RL, Konopka JB, Witte ON. Activation of the c-abl oncogene by viral transduction or chromosomal translocation generates altered c-abl proteins with similar in vitro kinase properties. Mol Cell Biol. 1985;5:204–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ben-Neriah Y, Daley GQ, Mes-Masson AM, Witte ON, Baltimore D. The chronic myelogenous leukemia-specific P210 protein is the product of the bcr/abl hybrid gene. Science. 1986;233:212–214. [DOI] [PubMed] [Google Scholar]
  • 41.Daley GQ, Etten RV, Baltimore D. Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome. Science. 1990;247:824–830. [DOI] [PubMed] [Google Scholar]
  • 42.Heisterkamp N, Jenster G, ten Hoeve J, Zovich D, Pattengale PK, Groffen J. Acute leukaemia in bcr/abl transgenic mice. Nature. 1990;344:251–253. [DOI] [PubMed] [Google Scholar]
  • 43.Bower H, Björkholm M, Dickman PW, Hoglund M, Lambert PC, Andersson TM- L. Life expectancy of patients With chronic myeloid leukemia approaches the life expectancy of the general population. J Clin Oncol. 2016;34:2851–2857. [DOI] [PubMed] [Google Scholar]
  • 44.Patel AB, Wilds BW, Deininger MW. Treating the chronic-phase chronic myeloid leukemia patient: which TKI, when to switch and when to stop? Expert Rev Hematol. 2017;10:659–674. [DOI] [PubMed] [Google Scholar]
  • 45.Rusch V, Baselga J, Cordon-Cardo C, et al. Differential expression of the epidermal growth factor receptor and its ligands in primary non-small cell lung cancers and adjacent benign lung. Cancer Res. 1993;53:2379–2385. [PubMed] [Google Scholar]
  • 46.Ranson M, Hammond LA, Ferry D, et al. ZD1839, a selective oral epidermal growth factor receptor-tyrosine kinase inhibitor, is well tolerated and active in patients with solid, malignant tumors: results of a phase I trial. J Clin Oncol. 2002;20:2240–2250. [DOI] [PubMed] [Google Scholar]
  • 47.Herbst RS, Maddox A- M, Rothenberg ML, et al. Selective oral epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 is generally well-tolerated and has activity in non-small-cell lung cancer and other solid tumors: results of a phase I trial. J Clin Oncol. 2002;20:3815–3825. [DOI] [PubMed] [Google Scholar]
  • 48.Baselga J, Rischin D, Ranson M, et al. Phase I safety, pharmacokinetic, and pharmacodynamic trial of ZD1839, a selective oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with five selected solid tumor types. J Clin Oncol. 2002;20:4292–4302. [DOI] [PubMed] [Google Scholar]
  • 49.Kris MG, Natale RB, Herbst RS, et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. JAMA. 2003;290:2149–2158. [DOI] [PubMed] [Google Scholar]
  • 50.Fukuoka M, Yano S, Giaccone G, et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer. J Clin Oncol. 2003;21:2237–2246. [DOI] [PubMed] [Google Scholar]
  • 51.Bailey R, Kris M, Wolf M, et al. O-242 Gefitinib (‘Iressa’, ZD1839) monotherapy for pretreated advanced non-small-cell lung cancer in IDEAL 1 and 2: tumor response is not clinically relevantly predictable from tumor EGFR membrane staining alone. Lung Cancer. 2003;41:S71. [Google Scholar]
  • 52.Cohen MH, Williams GA, Sridhara R, et al. United States Food and Drug Administration drug approval summary: Gefitinib (ZD1839; Iressa) tablets. Clin Cancer Res. 2004;10:1212–1218. [DOI] [PubMed] [Google Scholar]
  • 53.Herbst RS, Giaccone G, Schiller JH, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial--INTACT 2. J Clin Oncol. 2004;22:785–794. [DOI] [PubMed] [Google Scholar]
  • 54.Giaccone G, Herbst RS, Manegold C, et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial--INTACT 1. J Clin Oncol. 2004;22:777–784. [DOI] [PubMed] [Google Scholar]
  • 55.Jänne PA, Gurubhagavatula S, Yeap BY, et al. Outcomes of patients with advanced nonll cell lung cancer treated with gefitinib (ZD1839, “Iressa”) on an expanded access study. Lung Cancer. 2004;44:221–230. [DOI] [PubMed] [Google Scholar]
  • 56.Miller VA, Kris MG, Shah N, et al. Bronchioloalveolar pathologic subtype and smoking history predict sensitivity to gefitinib in advanced non-small-cell lung cancer. J Clin Oncol. 2004;22:1103–1109. [DOI] [PubMed] [Google Scholar]
  • 57.Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci USA. 2004;101:13306–13311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–1500. [DOI] [PubMed] [Google Scholar]
  • 59.Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–2139. [DOI] [PubMed] [Google Scholar]
  • 60.Oxnard GR, Arcila ME, Sima CS, et al. Acquired resistance to EGFR tyrosine kinase inhibitors in EGFR-mutant lung cancer: distinct natural history of patients with tumors harboring the T790M mutation. Clin Cancer Res. 2011;17:1616–1622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Yu HA, Arcila ME, Rekhtman N, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res. 2013;19:2240–2247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yang JC- H, Ahn M- J, Kim D- W, et al. Osimertinib in pretreated T790M-positive advanced non-small-cell lung cancer: AURA study phase II extension component. J Clin Oncol. 2017;35:1288–1296. [DOI] [PubMed] [Google Scholar]
  • 63.Goss G, Tsai C- M, Shepherd FA, et al. Osimertinib for pretreated EGFR Thr790Met-positive advanced non-small-cell lung cancer (AURA2): a multicentre, open-label, single-arm, phase 2 study. Lancet Oncol. 2016;17:1643–1652. [DOI] [PubMed] [Google Scholar]
  • 64.Mok TS, Wu Y- L, Ahn M- J, et al. Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung Cancer. N Engl J Med. 2017;376:629–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Oxnard GR, Hu Y, Mileham KF, et al. Assessment of resistance mechanisms and clinical implications in patients with EGFR T790M-positive lung cancer and acquired resistance to osimertinib. JAMA Oncol. 2018;4:1527–1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Soria J- C, Ohe Y, Vansteenkiste J, et al. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med. 2018;378:113–125. [DOI] [PubMed] [Google Scholar]
  • 67.Gainor JF, Shaw AT, Sequist LV, et al. EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res. 2016;22:4585–4593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Lisberg A, Cummings A, Goldman JW, et al. A phase II study of pembrolizumab in EGFR-mutant, PD-L1+, tyrosine kinase inhibitor naive patients with advanced NSCLC. J Thorac Oncol. 2018;13:1138–1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lee CK, Man J, Lord S, et al. Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer-a meta-analysis. J Thorac Oncol. 2017;12:403–407. [DOI] [PubMed] [Google Scholar]
  • 70.Ciardiello F, Tortora G. A novel approach in the treatment of cancer: targeting the epidermal growth factor receptor. Clin Cancer Res. 2001;7:2958–2970. [PubMed] [Google Scholar]
  • 71.Cunningham D, Humblet Y, Siena S, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337–345. [DOI] [PubMed] [Google Scholar]
  • 72.Lièvre A, Bachet J-B, Le Corre D, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006;66:3992–3995. [DOI] [PubMed] [Google Scholar]
  • 73.Di Fiore F, Blanchard F, Charbonnier F, et al. Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by Cetuximab plus chemotherapy. Br J Cancer. 2007;96:1166–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lievre A, Bachet J- B, Boige V, et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol. 2008;26:374–379. [DOI] [PubMed] [Google Scholar]
  • 75.Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. NEngl J Med. 2008;359:1757–1765. [DOI] [PubMed] [Google Scholar]
  • 76.Allegra CJ, Rumble RB, Hamilton SR, et al. Extended RAS gene mutation testing in metastatic colorectal carcinoma to predict response to anti-epidermal growth factor receptor monoclonal antibody therapy: american society of clinical oncology provisional clinical opinion update 2015. J Clin Oncol. 2016;34:179–185. [DOI] [PubMed] [Google Scholar]
  • 77.Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hauschild A, Grob J- J, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380:358–365. [DOI] [PubMed] [Google Scholar]
  • 79.Paraiso KHT, Fedorenko IV, Cantini LP, et al. Recovery of phospho-ERK activity allows melanoma cells to escape from BRAF inhibitor therapy. Br J Cancer. 2010;102:1724–1730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sosman JA, Kim KB, Schuchter L, et al. Survival in BRAF V600-Mutant Advanced Melanoma Treated with Vemurafenib. N Engl J Med. 2012;366:707–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sosman JA, Pavlick AC, Schuchter LM, et al. Analysis of molecular mechanisms of response and resistance to vemurafenib (vem) in BRAFV600E melanoma. J Clin Oncol. 2012;30(15_suppl):8503–8503. [Google Scholar]
  • 82.Long GV, Stroyakovskiy D, Gogas H, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. NEngl J Med. 2014;371:1877–1888. [DOI] [PubMed] [Google Scholar]
  • 83.Long GV, Stroyakovskiy D, Gogas H, et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet. 2015;386:444–451. [DOI] [PubMed] [Google Scholar]
  • 84.Long GV, Flaherty KT, Stroyakovskiy D, 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. 2017;28:1631–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. NEngl J Med. 2015;372:30–39. [DOI] [PubMed] [Google Scholar]
  • 86.Holbeck SL, Camalier R, Crowell JA, et al. The National Cancer Institute ALMANAC: a comprehensive screening resource for the detection of anticancer drug pairs with enhanced therapeutic activity. Cancer Res. 2017;77:3564–3576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wolff AC, Hammond MEH, Allison KH, Harvey BE, McShane LM, Dowsett M. HER2 testing in breast cancer: american society of clinical oncology/college of american pathologists clinical practice guideline focused update summary. J Oncol Prac. 2018;14:437–441. [DOI] [PubMed] [Google Scholar]
  • 88.Roychowdhury S, Iyer MK, Robinson DR, et al. Personalized oncology through integrative high-throughput sequencing: a pilot study. Sci TranslMed. 2011;3:111ra121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Garraway LA, Lander ES. Lessons from the cancer genome. Cell. 2013;153:17–37. [DOI] [PubMed] [Google Scholar]
  • 90.CMS finalizes coverage of Next Generation Sequencing tests, ensuring enhanced access for cancer patients | CMS. https://www.cms.gov/newsroom/press-releases/cms-finalizes-coverage-next-generation-sequencing-tests-ensuring-enhanced-access-cancer-patients Accessed April 14, 2019.
  • 91.Jones S, Anagnostou V, Lytle K, et al. Personalized genomic analyses for cancer mutation discovery and interpretation. Sci TranslMed. 2015;7:283ra53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Yap TA, Sandhu SK, Workman P, de Bono JS. Envisioning the future of early anticancer drug development. Nat Rev Cancer. 2010;10:514–523. [DOI] [PubMed] [Google Scholar]
  • 93.Gainor JF, Longo DL, Chabner BA. Pharmacodynamic biomarkers: falling short of the mark? Clin Cancer Res. 2014;20:2587–2594. [DOI] [PubMed] [Google Scholar]
  • 94.Goldberg SB, Patel AA. Monitoring immunotherapy outcomes with circulating tumor DNA. Immunotherapy. 2018;10:1023–1025. [DOI] [PubMed] [Google Scholar]
  • 95.Tannock IF, Hickman JA. Limits to personalized cancer medicine. N Engl J Med. 2016;375:1289–1294. [DOI] [PubMed] [Google Scholar]
  • 96.Hyman DM, Taylor BS, Baselga J. Implementing genome-driven oncology. Cell. 2017;168:584–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168:613–628. [DOI] [PubMed] [Google Scholar]
  • 98.Rübben A, Araujo A. Cancer heterogeneity: converting a limitation into a source of biologic information. J Transl Med. 2017;15:190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Redig AJ, Jänne PA. Basket trials and the evolution of clinical trial design in an era of genomic medicine. J Clin Oncol. 2015;33(9):975–977. [DOI] [PubMed] [Google Scholar]
  • 100.Dienstmann R, Rodon J, Tabernero J. Optimal design of trials to demonstrate the utility of genomically-guided therapy: Putting Precision Cancer Medicine to the test. Mol Oncol. 2015;9:940–950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.André F, Bachelot T, Commo F, et al. Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER). Lancet Oncol. 2014;15:267–274. [DOI] [PubMed] [Google Scholar]
  • 102.Tsimberidou A- M, Iskander NG, Hong DS, et al. Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative. Clin Cancer Res. 2012;18:6373–6383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kris MG, Johnson BE, Berry LD, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA. 2014;311:1998–2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sohal DPS, Rini BI, Khorana AA, et al. Prospective clinical study of precision oncology in solid tumors. J Natl Cancer Inst. 2016;108(3). [DOI] [PubMed] [Google Scholar]
  • 105.Bedard PL, Oza A, Clarke B, et al. Abstract PR03: Molecular profiling of advanced solid tumors at Princess Margaret Cancer Centre and patient outcomes with genotype-matched clinical trials. Clin Cancer Res. 2016;22(1 Supplement):PR03–PR03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Le Tourneau C, Delord J- P, Gonqalves A, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16:1324–1334. [DOI] [PubMed] [Google Scholar]
  • 107.Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. NEngl J Med. 2015;372:2509–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Approved Drugs - FDA grants accelerated approval to pembrolizumab for first tissue/site agnostic indication. https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm560040.htm Accessed April 10, 2019.
  • 110.Approved Drugs - FDA approves larotrectinib for solid tumors with NTRK gene fusions. https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm626720.htm Accessed April 10, 2019.
  • 111.Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. NEngl J Med. 2015;373:726–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Approved Drugs - FDA grants regular approval to dabrafenib and trametinib combination for metastatic NSCLC with BRAF V600E mutation. https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm564331.htm Accessed April 10, 2019.
  • 113.Approved Drugs - FDA approves dabrafenib plus trametinib for anaplastic thyroid cancer with BRAF V600E mutation. https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm606708.htm Accessed April 15, 2019.
  • 114.Mateo J, Carreira S, Sandhu S, et al. DNA-repair defects and olaparib in metastatic prostate cancer. NEngl JMed. 2015;373:1697–1708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gruber JJ, Afghahi A, Hatton A, et al. Talazoparib beyond BRCA: A phase II trial of talazoparib monotherapy in BRCA1 and BRCA2 wild-type patients with advanced HER2-negative breast cancer or other solid tumors with a mutation in homologous recombination (HR) pathway genes. J Clin Oncol. 2019;37(suppl):abstr 3006. [Google Scholar]
  • 116.Kim ES, Herbst RS, Wistuba II, et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov. 2011;1:44–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Papadimitrakopoulou V, Lee JJ, Wistuba II, et al. The BATTLE-2 study: A biomarker-integrated targeted therapy study in previously treated patients with advanced non-small-cell lung cancer. J Clin Oncol. 2016;34:3638–3647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Janiaud P, Serghiou S, Ioannidis JPA. New clinical trial designs in the era of precision medicine: An overview of definitions, strengths, weaknesses, and current use in oncology. Cancer Treat Rev. 2019;73:20–30. [DOI] [PubMed] [Google Scholar]
  • 119.Conley BA, Doroshow JH. Molecular analysis for therapy choice: NCI MATCH. Semin Oncol. 2014;41:297–299. [DOI] [PubMed] [Google Scholar]
  • 120.Cunanan KM, Gonen M, Shen R, et al. Basket trials in oncology: a trade-off between complexity and efficiency. J Clin Oncol. 2017;35:271–273. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES