The aim of this study was to assess the frequency of potentially actionable genomic alterations in breast cancer that could be targeted with approved agents or investigational drugs in clinical trials using a next-generation sequencing-based genomic profiling assay. Systematic evaluation of the predictive value of these genomic alterations is critically important for further progress in this field.
Keywords: Next-generation sequencing, Precision medicine, Molecularly targeted therapy, Predictive markers
Abstract
Background.
The aim of this study was to assess the frequency of potentially actionable genomic alterations in breast cancer that could be targeted with approved agents or investigational drugs in clinical trials using a next-generation sequencing-based genomic profiling assay performed in a Clinical Laboratory Improvement Amendments-certified and College of American Pathologists-accredited commercial laboratory.
Methods.
Fifty-one breast cancers were analyzed, including primary tumor biopsies of 33 stage I–II and 18 stage IV cancers (13 soft tissue, 3 liver, and 2 bone metastases). We assessed 3,230 exons in 182 cancer-related genes and 37 introns in 14 genes often rearranged in cancer for base substitutions, indels, copy number alterations, and gene fusions. The average median sequencing depth was 1,154×.
Results.
We observed 158 genomic alterations in 55 genes in 48 of 51 (94%) tumors (mean 3.1, range 0–9). The average number of potentially therapeutically relevant alterations was similar in primary (1.6, range 0–4) and in heavily pretreated metastatic cancers (2.0, range 0–4) (p = .24). The most common actionable alterations were in PIK3CA (n = 9, phosphatidylinositol 3-kinase [PI3K]/mammalian target of rapamycin [mTOR] inhibitors), NF1 (n = 7, PI3K/mTOR/mitogen-activated protein kinase inhibitors), v-akt murine thymoma viral oncogene homolog 1-3 (n = 7, PI3K/mTOR/AKT inhibitors), BRCA1/2 (n = 6, poly[ADP-ribose] polymerase inhibitors), and CCND1,2 and CCNE (n = 8)/cycline dependent kinase (CDK)6 (n = 1) (CDK4/6 inhibitors), KIT (n = 1, imatinib/sunitinib), ALK (n = 1, crizotinib), FGFR1,2 (n = 5, fibroblast growth factor receptor inhibitors), and EGFR (n = 2, epidermal growth factor receptor inhibitors). Our sequencing assay also correctly identified all six cases with HER2 (ERBB2) amplification by fluorescence in situ hybridization when tumor content was adequate. In addition, two known activating HER2 mutations were identified, both in unamplified cases.
Conclusion.
Overall, 84% of cancers harbored at least one genomic alteration linked to potential treatment options. Systematic evaluation of the predictive value of these genomic alterations is critically important for further progress in this field.
Abstract
摘要
背景 本研究的目的是,在获得临床实验室改进修正案认证及美国病理学家学会认可的商业实验室中,使用一种新一代测序型基因组分析技术对乳腺癌进行测序分析,以确定癌灶内那些可作为已批准药物或临床试验性药物治疗靶标的基因组突变的发生频率。
方法 我们共分析了 51 例癌灶,包括 33 例 I-II 期原发性肿瘤活检组织和 18 例 IV 期癌灶活检组织(13 例为软组织灶,3 例为肝脏病灶,2 例为骨转移灶)。我们对 182 个癌症相关基因的 3230 个外显子和 14 个在癌症中常发生重排之基因的 37 个内含子进行了测序,以查找其中的碱基替代、插入缺失、拷贝数突变和基因融合。平均中位测序深度为 1154×。
结果 我们在 51 个瘤灶的 48 个 (94%) 内的 55 个基因中发现了 158 个基因组突变(平均 3.1,范围为 0-9)。原发性癌灶和进行过多重预先治疗的转移癌灶中可作为潜在治疗靶标的平均突变数大体相似(前者为 1.6,范围为 0-4;后者为 2.0,范围为 0-4)(p = 0.24)。最常见的可靶定突变为 PIK3CA (n =9,磷脂酰肌醇 3-激酶 [PI3K]/雷帕霉素的哺乳类动物靶标 [mTOR] 抑制剂)、NF1 (n =7,PI3K/mTOR/丝裂原活化蛋白激酶抑制剂)、v-akt 鼠科胸腺瘤病毒致癌基因同源体 1-3(n =7,PI3K/mTOR/AKT 抑制剂)、 BRCA1/2 (n =6,聚[ADP-核糖]聚合酶抑制剂)、CCND 1,2 和 CCNE (n =8)/细胞周期素依赖性抑制剂 (CDK)6 (n = 1)(CDK4/6 抑制剂)、KIT (n =1,伊马替尼/舒尼替尼)、ALK (n =1,克唑替尼)、FGFR1,2 (n =5,成纤维细胞生长因子受体抑制剂)以及 EGFR (n =2,表皮生长因子受体抑制剂)。在肿瘤量充足的情况下,我们的测序分析还通过荧光原位杂交法,准确地鉴定出了所有六例 HER2 (ERBB2) 扩增突变。此外,我们还检测出了两例已知的活化 HER2 突变,均出现在非扩增病例中。
结论 总体而言,84% 的癌症存在至少一种可作为潜在治疗靶标的基因组突变。对这些基因组突变的预测价值进行系统性评估,对推动这一领域的进步具有重要意义。The Oncologist 2014;19:453–458
Implications for Practice:
The technical ability to perform molecular profiling in the clinic is broadly available; it is now critically important to focus on assessing the clinical utility of molecular profiling as a patient selection tool.
Introduction
An increasing number of molecularly targeted drugs are available in the clinic as approved drugs (Table 1) or in the context of clinical trials (http://www.clinicaltrials.gov). These drugs target specific molecular abnormalities, including mutated protein kinases and amplified or rearranged genes. Cancers that carry these abnormalities often, but not always, respond to the corresponding targeted therapies. For example, the HER2 gene-amplified breast cancers benefit from HER2-targeted therapies [1]. Chronic myeloid leukemia with the BCR-ABL translocation responds to inhibitors of the BCR-ABL kinase [2]. Lung cancers with activating mutations of EGFR can benefit from epidermal growth factor receptor (EGFR) inhibitors [3], and lung cancers that carry an activating rearrangement of the ALK kinase often respond to anaplastic lymphoma kinase (ALK) inhibitors [4]. BRAF-mutant melanoma may respond to v-raf murine sarcoma viral oncogene homolog B1 (BRAF) inhibitors [5], and activating mutations in c-KIT or PDGFR render gastrointestinal stromal tumors sensitive to KIT inhibitors [6]. The repertoire of genomic abnormalities and their incidence differ across different histologic types of cancer, but most abnormalities are not unique to any particular cancer type [7]. Although the same genomic abnormality may play a more important driver role in one type of cancer compared with another, there is also evidence to support that different types of cancers could respond to the same biologically targeted agent if they harbor the sensitizing genomic abnormality. For example, HER2-targeted therapies are effective in gastric and esophageal adenocarcinomas that have HER2 gene amplification [8]. The BRAF inhibitor vemurafenib has shown promising results in patients with BRAF mutant metastatic papillary thyroid cancer and malignant histocytosis [9]. The purpose of the current study is to survey the potentially targetable genomic abnormalities in primary and metastatic breast cancers using a standardized, commercially available next-generation sequencing (NGS)-based genomic profiling assay on routine clinical tissue samples.
Table 1.
U.S. Food and Drug Administration-approved molecularly targeted drugs for cancer

Several studies have examined the mutational landscape of breast cancer using whole genome or partial genome sequencing [10–12]. Many of the initial whole genome and whole exome sequencing studies included very few patients and had limited sensitivity because of low sequencing depth. Other studies included a larger number of patients but restricted the analysis to a modest number of known oncogenic mutations [13–15]. The most comprehensive genomic analysis of breast cancer was recently reported by the Cancer Genome Atlas Network (TCGA) [16]. Whole exome sequencing was performed on 507 breast cancers with modest sequencing depth (30% of target sequences had coverage <20-fold). Low to moderate coverage limits the sensitivity to detect genomic events that may be restricted to relatively small tumor cell subpopulations. In aggregate, the above studies have established TP53 and PI3KCA as the most frequently mutated genes in breast cancer and also revealed a large number of low-frequency potentially druggable genomic anomalies in variable proportions of breast cancers. These observations raise the possibility that subsets of breast cancers may be candidates for targeted therapies aimed at rare genomic abnormalities. In this study, we performed full sequencing of 3,230 exons in 182 cancer-related genes and 37 introns in 14 genes that are frequently rearranged in cancers. As a result of the targeted sequencing method and the high, uniform coverage (>99% of exons covered ≥100×), our detection sensitivity for minor allele variants is significantly higher than those achieved in the TCGA. The analysis was performed using a standard operating procedure in a CLIA-certified (Clinical Laboratory Improvement Amendments, http://www.cms.gov/clia), CAP-accredited (College of American Pathologists) commercial molecular diagnostic laboratory; therefore, the results can be directly incorporated into clinical trials as patient selection criteria or used for medical decision making.
Materials and Methods
Patients and Samples
Fifty-one breast cancers were analyzed, including primary tumor biopsies in 1 stage I, 17 stage II, and 15 stage III primary breast cancers and 18 total stage IV metastasis biopsies of 13 soft tissue, 3 liver, and 2 bone tumors. The mean age was 52 years (range 28–86 years), 58% ER+, 20% HER2+, 31% triple negative. Fine-needle aspirations (FNA) of the primary cancers were obtained prospectively at the time of diagnosis before any therapy in the context of a prospective biomarker discovery protocol at MD Anderson Cancer Center. Metastatic cancer biopsies, also obtained through FNA, were collected in the context of a prospective biomarker validation trial [17]. Patients with metastatic breast cancers had received at least two prior lines of therapy for metastatic cancer and were exposed to an average of seven different drugs (range 5–17), including adjuvant therapy before the biopsies were obtained. Both tissue collection studies were approved by the Institutional Review Committee of the University of Texas MD Anderson Cancer Center, and all patients signed informed consent. Analysis was performed on the already collected tissue samples, and the results were not included in the medical records or used for treatment decision making.
Next-Generation Sequencing (NGS)-Based Genomic Profiling
Two FNA aspirates were pooled into a single vial for subsequent molecular analysis, and NGS-based targeted sequencing was performed by Foundation Medicine. Genomic DNA was extracted using the Maxwell 16 FFPE Plus LEV DNA Purification kit (Promega, Madison, WI, http://www.promega.com) and quantified using a PicoGreen fluorescence assay (Invitrogen, Carlsbad, CA, http://www.invitrogen.com). Fifty to 200 ng of DNA was sheared to 100–400 base pair fragments by sonication, followed by end-repair, dA-addition, and ligation of indexed, Illumina (San Diego, CA, http://www.illumina.com) sequencing adaptors. Sequencing libraries were hybridization captured using a pool of >24,000 individually synthesized 5′-biotinylated DNA oligonucleotides (Integrated DNA Technologies, Coralville, IA, http://www.idtdna.com/Home/Home.aspx) that were designed to target 182 cancer-related genes and 37 additional introns in 14 genes often rearranged in cancer, corresponding to 1.14 million total base pairs (supplemental online Table 1). DNA sequencing is performed using the HiSeq-2000 instrument (Illumina) with 49 × 49 paired-end reads. Point mutations (base substitutions), short insertions/deletions (indels), focal amplifications, homozygous deletions, and chromosomal rearrangements were analyzed using Bayesian algorithms, local assembly, comparison with process-matched normal controls, and analysis of chimeric pairs, respectively. The method was optimized to detect >5% mutant allele frequency (MAF) of base substitutions and >10% MAF of indels with >99% accuracy. The validated accuracy of copy number alterations was >95%. Methodological details and analytical validity of the assay were reported previously [18]. The results were annotated and interpreted through dbSNP, COSMIC, and medical literature to assemble the final report of actionable genomic alterations. We did not analyze matched normal specimens; however, all reported mutations have been identified in previously published cancer-sequencing studies and were therefore considered somatic events. An alteration was categorized as potentially “actionable” if linked to an approved therapy in breast cancer or another solid tumor, or if it mapped to a pathway targetable with an approved drug or a drug in clinical trials.
Results
All biopsies yielded the 50 ng of DNA required for analysis. The average median sequencing depth was 1,154-fold with >99% of nucleotides covered ≥100-fold. The time from DNA processing to final report required 7–14 days (Fig. 1). We observed 158 genomic alterations in 48 of 51 (94%) tumors (mean 3.1, range 0–9) in 55 genes. In total, 30 of 33 (91%) primary and 18 of 18 (100%) metastatic breast cancers had at least one genomic alteration. The 158 genomic alterations observed included 45 base substitutions (15%), 24 short insertions/deletions (28%), 63 focal amplifications (40%), 18 homozygous deletions (11%), and 8 rearrangements (5%) (Fig. 2). Overall, 84% of samples harbored at least one actionable alteration (Fig. 3). These actionable targets included the following: PIK3CA (n = 9, 18%), HER2 (n = 9, 18%), NF1 (n = 7, 14%), MCL1 (n = 6, 12%), and PTEN (n = 5, 10%). Twenty-five other genes with actionable alterations were identified, occurring at lower frequencies (n = 4 or fewer, <10%).
Figure 1.

Schematic illustrating tumor sample preparation, sequencing library preparation, the analysis pipeline, and clinical reporting. Reprinted with modification from Nature Biotech 31:1023-1031, 2013.
Figure 2.
Pie chart of the alteration classes in the 51 breast cancers sequenced in this study.
Figure 3.

Distribution of alterations observed in this study. For abbreviations and information on these genes, please see http://www.genecards.org.
Abbreviations: FGFR, fibroblast growth factor receptor; MEK, mitogen-activated protein kinase kinase; PARP, poly(ADP-ribose) polymerase; PI3, phosphatidylinositol 3; PI3K, phosphatidylinositol 3-kinase.
The specific alterations that were observed in the most frequently actionable targets included the following: PIK3CA C420R, E542K, E545A, E545K, H1047R (H1047R occurred in four cases); HER2 amplification and HER2 S310F and L755S mutations; NF1 truncation; MCL1 amplification; PTEN truncation; and PTEN S170I mutation. Sequencing-based HER2 copy number assessment correctly identified HER2 gene amplification in all six cases that were HER2-positive by routine clinical testing and had adequate tumor cellularity for NGS analysis. Additional mutations in other well-known oncogenes were observed, including AKT1 E17K (three cases); ALK V757M; DNMT3A R882C; ESR1 Y537C, Y537N, and D538G; KIT K642E; and KRAS G12D. Numerous other alterations with no available therapeutic implications were also observed in each cancer, including MYC amplification (n = 8, 16%) and TP53 alteration (n = 32, 63%: 31 mutations and 1 homozygous deletion), which were the most frequent alterations observed.
Thirty-nine (76%) tumors exhibited multiple alterations (Fig. 4). Only nine (18%) tumors exhibited single genetic alterations, and three (6%) tumors exhibited no detectable alterations. The average number of alterations was 2.9 in primary tumors and 3.4 in heavily pretreated metastatic cancers (p = .31). The average number of actionable alterations was 1.6 in primary (range 0–4) and 2.0 (range 0–4) in heavily pretreated metastatic cancers (p = .24).
Figure 4.

Comprehensive annotation of genomic alterations of 51 breast cancers.
Discussion
This study, along with numerous other reports, demonstrates that comprehensive sequencing of potentially druggable genes can be performed on small, routine, diagnostic needle biopsies in a clinically relevant time frame. The incorporation of NGS to profile cancer biopsies for therapeutic targets has become a reality in the clinic. Our results also indicate that a large fraction of breast cancers contains genomic abnormalities that may render them susceptible to approved or investigational therapies. The distribution and frequency of the most frequent genomic alterations observed in this study are similar to the findings reported by the TCGA breast cancer project including TP53 and PI3KCA mutations and HER2 amplification [16]. Importantly, sequencing-based assessment of HER2 gene copy number correctly identified six of the six clinically HER2-amplified cases when assay requirements were met. We also identified functional mutations in the HER2 gene (S310F, L755S) in two cases in the absence of amplification. Both of these mutations have previously been reported in the literature; S310F is an activating mutation [19], and L755S is a mediator of resistance to lapatinib [20]. We also detected numerous, less frequent but directly targetable alterations in KIT (K642E, an activating mutation commonly seen in gastrointestinal stromal tumors [21] and in acral and mucosal melanomas [22]), ALK (V757M, mutation in the extracellular domain that was previously observed in colorectal cancer [10]; ALK translocations cause non-small cell lung cancer, and mutations cause neuroblastoma [23]), and AKT1 mutation (E17K, a constitutively activating mutation that was also seen in colorectal, endometrial, lung, and ovarian cancers and acute leukemias [24]). We observed amplifications of the EGFR, FGFR1, FGFR2, AKT2, and AKT3 genes, all of which can be directly targeted by approved or investigational drugs.
A second collectively large group includes alterations that are not direct drug targets but represents biological pathways that could be targeted by drugs. These include deletion and truncation of neurofibromin/NF1 (Y1625fs*5, A2646fs*14, Q1399*, W1559*, and c.1393-1G>T splice site mutation), a negative regulator of the RAS signal transduction pathway, and a mutation in KRAS (G12D, a mutation that confers resistance to anti-EGFR therapy in colorectal cancer). Amplification of MCL1 gene that encodes BCL2-like protein inhibits apoptosis and amplifications in cyclins E and D, and MYC, as well as homozygous loss of PTEN, BRCA2, and truncation of BRCA1 (S1253fs*10, Q1756fs*74, K1759fs*70) and BRCA2 (V1610fs*4, L2092fs*7).
It is also important to recognize that almost all cancers harbored multiple abnormalities. The average number of targetable alterations per sample was surprisingly similar, 1.6 in primary versus 2.0 in heavily treated metastatic breast cancers. The multiplicity of anomalies suggests that ultimately combinatorial therapies may be required for optimal efficacy. Studying the functional interaction between the multiple somatic alterations that tumors acquire and the equally large number of functional germline polymorphisms that we all carry represents a very important and fertile ground for research. However, it is also important to remember that, despite the multiplicity of functional genomic alterations, targeting single alterations can provide clinical benefit (e.g., antiestrogens, HER2-targeted therapies, etc.), which justifies clinical testing of targeted therapies.
The increasing clinical availability of NGS-based profiling assays and the results that they generate raise several important clinical questions. The most important one is how to act on the results. Currently, all U.S. Food and Drug Administration (FDA)-approved therapies that target a specific molecular abnormality in breast cancer are HER2-targeted agents (trastuzumab, lapatinib, pertuzumab, and ado-trastuzumab-emtansine). However, there are 26 other FDA-approved drugs (Table 1) that target specific somatic molecular abnormalities seen in cancer, most of which are encountered at low frequencies in breast cancer. It is attractive to consider the use of these targeted agents in tumor types in which they have not been approved to evaluate the predictive utility of target profiling [25, 26]. The safety profiles of already approved agents are already well established, and the key challenge is to assess clinical benefit. Many academic institutions pursue “molecular triaging” or “basket” studies that involve performing molecular target profiling and using the results to steer patients to clinical trials that test targeted therapies [27]. The U.S. National Cancer Institute recently announced plans for the MATCH trial that aims to provide access to therapies that target the specific mutations found in a cancer [28]. Pharmaceutical companies are also considering providing a portfolio of their approved drugs and some investigational agents for molecular triaging studies that encompass multiple cancer types; however, no such clinical trial is open yet. The fragmentation of the patient population into very small, molecularly defined subsets will be a challenge during the implementation of these trials. It is hard to envisage efficiently running separate studies for BRAF mutant, HER2 mutant, AKT mutant, ALK mutant, cKIT mutant, and FGFR-amplified breast cancer subsets. More importantly, only a minority of cancer patients receive their care through large academic centers and have access to these studies; overall, only 5%–10% of U.S. cancer patients participate in clinical trials [29].
One could also consider other innovative ways to harness the broad scientific interest in this field and the motivation of patients with incurable diseases to broaden their treatment options. Establishment of a nationwide registry of molecularly targeted therapies of rare genomic abnormalities could rapidly move the field forward. The registry would collect basic clinical and molecular information and simple, but informative, outcome data such as duration of therapy (i.e., a composite endpoint of tolerability and cancer control). Such registry could even be linked to coverage for treatment and maintained by the Center for Medicaid and Medicare Services or other third-party payers. Drug activity could be assessed and made public at predefined milestones, for example, after accruing the first 100 patients nationwide with a particular molecular anomaly. The registry would not replace rigorous clinical trials but could identify clinical scenarios in which further trials are needed.
Conclusion
In summary, NGS-based genomic profiling of DNA from breast cancer needle biopsies to assess potential therapeutic targets is readily available. Target profiling showed a high frequency of genomic alterations linked to potential treatment option with approved or investigational drugs. These alterations include multiple different types of abnormalities, including gene amplification and deletions, frame shifts, small insertions and deletions (indels), single-nucleotide substitutions, and gene rearrangements with fusion genes. These results raise a broad spectrum of distinct clinically testable therapeutic hypotheses for individual patients. Testing the clinical efficacy of these treatment options represents a major challenge for the traditional clinical trial system as a result of the varied and individually small patient subsets. Innovative approaches to provide access to potentially effective drugs and to capture systematically the outcome of therapy are needed to move the field forward and to provide benefit to the greatest number of patients without long delays.
See http://www.TheOncologist.com for supplemental material available online.
Supplementary Material
Author Contributions
Conception/Design: Roman Yelensky, Kai Wang, Stacy Moulder, W. Fraser Symmans, Philip Stephens, Yun Wu, Baliang Wang, Rony Avritscher, Lajos Pusztai, Vincent Miller
Provision of study material or patients: Kai Wang, Stacy Moulder, W. Fraser Symmans, Philip Stephens, Yun Wu, Baliang Wang, Rony Avritscher, Lajos Pusztai
Collection and/or assembly of data: Neil Vasan, Gary Palmer, Lajos Pusztai
Data analysis and interpretation: Neil Vasan, Roman Yelensky, Hannah Dzimitrowicz, Gary Palmer, Lajos Pusztai, Maureen Cronin
Manuscript writing: Neil Vasan, Roman Yelensky, Lajos Pusztai
Final approval of manuscript: Gary Palmer, Lajos Pusztai, Maureen Cronin, Vincent Miller
Disclosures
Lajos Pusztai: Foundation Medicine (RF); Roman Yelensky: Foundation Medicine (E, IP, OI); Kai Wang: Foundation Medicine (E, OI); Gary Palmer: Foundation Medicine (E, OI); Philip Stephens: Foundation Medicine (E, OI); Maureen Cronin: Celgene (E); Celgene, Foundation Medicine (OI); Vincent Miller: Foundation Medicine (E, OI). The other authors indicated no financial relationships.
(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board
References
- 1.Hudis CA. Trastuzumab—mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51. doi: 10.1056/NEJMra043186. [DOI] [PubMed] [Google Scholar]
- 2.Capdeville R, Buchdunger E, Zimmermann J, et al. Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov. 2002;1:493–502. doi: 10.1038/nrd839. [DOI] [PubMed] [Google Scholar]
- 3.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: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
- 4.Gandhi L, Jänne PA. Crizotinib for ALK-rearranged non-small cell lung cancer: A new targeted therapy for a new target. Clin Cancer Res. 2012;18:3737–3742. doi: 10.1158/1078-0432.CCR-11-2393. [DOI] [PubMed] [Google Scholar]
- 5.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: 10.1056/NEJMoa1103782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Joensuu H, Roberts PJ, Sarlomo-Rikala M, et al. Effect of the tyrosine kinase inhibitor STI571 in a patient with a metastatic gastrointestinal stromal tumor. N Engl J Med. 2001;344:1052–1056. doi: 10.1056/NEJM200104053441404. [DOI] [PubMed] [Google Scholar]
- 7.Kan Z, Jaiswal BS, Stinson J, et al. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature. 2010;466:869–873. doi: 10.1038/nature09208. [DOI] [PubMed] [Google Scholar]
- 8.Bang Y-J, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687–697. doi: 10.1016/S0140-6736(10)61121-X. [DOI] [PubMed] [Google Scholar]
- 9.Kim KB, Cabanillas ME, Lazar AJ, et al. Clinical responses to vemurafenib in patients with metastatic papillary thyroid cancer harboring BRAF(V600E) mutation. Thyroid. 2013;23:1277–1283. doi: 10.1089/thy.2013.0057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Stephens PJ, Tarpey PS, Davies H, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486:400–404. doi: 10.1038/nature11017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Thomas RK, Baker AC, Debiasi RM, et al. High-throughput oncogene mutation profiling in human cancer. Nat Genet. 2007;39:347–351. doi: 10.1038/ng1975. [DOI] [PubMed] [Google Scholar]
- 12.Shah SP, Morin RD, Khattra J, et al. Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature. 2009;461:809–813. doi: 10.1038/nature08489. [DOI] [PubMed] [Google Scholar]
- 13.MacConaill LE, Campbell CD, Kehoe SM, et al. Profiling critical cancer gene mutations in clinical tumor samples. PLoS One. 2009;4:e7887. doi: 10.1371/journal.pone.0007887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Santarpia L, Qi Y, Stemke-Hale K, et al. Mutation profiling identifies numerous rare drug targets and distinct mutation patterns in different clinical subtypes of breast cancers. Breast Cancer Res Treat. 2012;134:333–343. doi: 10.1007/s10549-012-2035-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jurinke C, Oeth P, van den Boom D. MALDI-TOF mass spectrometry: A versatile tool for high-performance DNA analysis. Mol Biotechnol. 2004;26:147–164. doi: 10.1385/MB:26:2:147. [DOI] [PubMed] [Google Scholar]
- 16.Cancer Genome Atlas Network Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70. doi: 10.1038/nature11412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Moulder S. Personalized Treatment Selection for Metastatic Breast Cancer (NCT00780676). Available at http://clinicaltrials.gov/ct2/show/NCT00780676?term=nct00780676&rank=1Accessed March 17, 2014.
- 18.Frampton GM, Fichtenholtz A, Otto GA, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31:1023–1031. doi: 10.1038/nbt.2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Greulich H, Kaplan B, Mertins P, et al. Functional analysis of receptor tyrosine kinase mutations in lung cancer identifies oncogenic extracellular domain mutations of ERBB2. Proc Natl Acad Sci USA. 2012;109:14476–14481. doi: 10.1073/pnas.1203201109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Bose R, Kavuri SM, Searleman AC, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 2013;3:224–237. doi: 10.1158/2159-8290.CD-12-0349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Willmore C, Holden JA, Zhou L, et al. Detection of c-kit-activating mutations in gastrointestinal stromal tumors by high-resolution amplicon melting analysis. Am J Clin Pathol. 2004;122:206–216. doi: 10.1309/4E6U-YBY6-2N2F-CA6N. [DOI] [PubMed] [Google Scholar]
- 22.Lutzky J, Bauer J, Bastian BC. Dose-dependent, complete response to imatinib of a metastatic mucosal melanoma with a K642E KIT mutation. Pigment Cell Melanoma Res. 2008;21:492–493. doi: 10.1111/j.1755-148X.2008.00475.x. [DOI] [PubMed] [Google Scholar]
- 23.Shaw AT, Solomon B. Targeting anaplastic lymphoma kinase in lung cancer. Clin Cancer Res. 2011;17:2081–2086. doi: 10.1158/1078-0432.CCR-10-1591. [DOI] [PubMed] [Google Scholar]
- 24.Bleeker FE, Felicioni L, Buttitta F, et al. AKT1(E17K) in human solid tumours. Oncogene. 2008;27:5648–5650. doi: 10.1038/onc.2008.170. [DOI] [PubMed] [Google Scholar]
- 25.Conti RM, Bernstein AC, Villaflor VM, et al. Prevalence of off-label use and spending in 2010 among patent-protected chemotherapies in a population-based cohort of medical oncologists. J Clin Oncol. 2013;31:1134–1139. doi: 10.1200/JCO.2012.42.7252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Dienstmann R, Serpico D, Rodon J, et al. Molecular profiling of patients with colorectal cancer and matched targeted therapy in phase I clinical trials. Mol Cancer Ther. 2012;11:2062–2071. doi: 10.1158/1535-7163.MCT-12-0290. [DOI] [PubMed] [Google Scholar]
- 27.Pusztai L. Breast Cancer Molecular Analysis Protocol (MAP-IT) (NCT01855503). Available at http://clinicaltrials.gov/ct2/show/NCT01855503?term=NCT01855503&rank=1Accessed March 17, 2014.
- 28.Willyard C. ‘Basket studies’ will hold intricate data for cancer drug approvals. Nat Med. 2013;19:655. doi: 10.1038/nm0613-655. [DOI] [PubMed] [Google Scholar]
- 29.Murthy VH, Krumholz HM, Gross CP. Participation in cancer clinical trials: Race-, sex-, and age-based disparities. JAMA. 2004;291:2720–2726. doi: 10.1001/jama.291.22.2720. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.

