Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Pilot Study of a Next-Generation Sequencing-Based Targeted Anticancer Therapy in Refractory Solid Tumors at a Korean Institution

  • Hyung Soon Park,

    Affiliations Department of Pharmacology and Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea

  • Sun Min Lim,

    Affiliation Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea

  • Sora Kim,

    Affiliation Severance Biomedical Science Institute and Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea

  • Sangwoo Kim,

    Affiliation Severance Biomedical Science Institute and Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea

  • Hye Ryun Kim,

    Affiliation Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea

  • KyuBum Kwack,

    Affiliation Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea

  • Min Goo Lee,

    Affiliation Department of Pharmacology and Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea

  • Joo-Hang Kim,

    Affiliation Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea

  • Yong Wha Moon

    ymoon@cha.ac.kr

    Affiliation Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea

Abstract

We evaluated the preliminary efficacy and feasibility of a next-generation sequencing (NGS)-based targeted anticancer therapy in refractory solid tumors at a Korean institution. Thirty-six patients with advanced cancer underwent molecular profiling with NGS with the intent of clinical application of available matched targeted agents. Formalin-fixed paraffin-embedded (FFPE) tumors were sequenced using the Comprehensive Cancer Panel (CCP) or FoundationOne in the Clinical Laboratory Improvement Amendments-certified laboratory in the USA. Response evaluations were performed according to RECIST v1.1. Four specimens did not pass the DNA quality test and 32 specimens were successfully sequenced with CCP (n = 31) and FoundationOne (n = 1). Of the 32 sequenced patients, 10 (31.3%) were ≤40 years. Twelve patients (37.5%) had received ≥3 types of prior systemic therapies. Of 24 patients with actionable mutations, five were given genotype-matched drugs corresponding to actionable mutations: everolimus to PIK3CA mutation in parotid carcinosarcoma (partial response) and tracheal squamous cell carcinoma (stable disease; 21% reduction), sorafenib to PDGFRA mutation in auditory canal adenocarcinoma (partial response), sorafenib to BRAF mutation in microcytic adnexal carcinoma (progressive disease), and afatinib to ERBB2 mutation in esophageal adenocarcinoma (progressive disease). Nineteen of 24 patients with actionable mutations could not undergo targeted therapy based on genomic testing because of declining performance status (10/24, 41.7%), stable disease with previous treatment (5/24, 20.8%), and lack of access to targeted medication (4/24, 16.7%). NGS-based targeted therapy may be a good option in selected patients with refractory solid tumors. To pursue this strategy in Korea, lack of access to clinical-grade NGS assays and a limited number of genotype-matched targeted medications needs to be addressed and resolved.

Introduction

In Korea, a total of around 200,000 new solid tumor cases and around 66,000 solid tumor deaths were reported in 2012 [1]. According to solid tumor types, approximately 30% of patients had distant metastasis at the time of cancer diagnosis [2]. Systemic chemotherapy is the standard treatment for these advanced cancer patients. However, many patients have treatment failure after standard therapy. These refractory solid tumor patients have few anti-cancer treatment options. Many of these patients pay high costs for unproven treatments, such as traditional medicines, but still do not have an improved survival in advanced solid tumors [2]. In Korea, the cost of alternative medicine in cancer patients increased from 621 to 1,388 (million US$, per year) during 2000–2010 [3]. Therefore, development of effective therapeutic strategies for refractory solid tumors is a huge unmet medical need.

Currently, molecular-based targeted therapy is a standard approach in selected patients such as non-small cell lung cancer (NSCLC) with EGFR mutations in which a very high response rate of approximately 75% is observed [4]. More recently, the strategy of matching targeted drugs to biologically relevant targets using molecular profiling techniques is becoming better established, although many challenges remain [5,6]. A systematic review of phase II clinical trials in advanced/metastatic NSCLC showed that molecular matching of patients' tumors to drugs was independently associated with better outcomes as compared with those of unselected patients [7]. Moreover, in the phase I setting, molecular matching was associated with improved outcomes in multivariate analysis [8].

Despite such advantages, genotype-matched therapy using molecular profiling in advanced cancer faces various obstacles in many countries. For instance, lack of access to clinical-grade next-generation sequencing (NGS) testing and targeted medication is the most common barrier. Therefore, we performed a pilot study to evaluate the preliminary efficacy and clinical feasibility of NGS-based targeted anticancer therapy at a Korean institution.

Materials and Methods

Study design

Fig 1 details the study schematic demonstrating the flow of the patients who consented for the current pilot study of NGS-based targeted anticancer therapy. First, sample quality, such as tissue fragmentation or DNA concentration, was checked using agarose gel or PicoGreenR [9]. After the sample quality check, somatic mutations were identified using FoundationOne by Foundation Medicine or Ion AmpliSeqTM Comprehensive Cancer Panel (CCP, Life Technology) by Macrogen, the Clinical Laboratory Improvement Amendments (CLIA)-certified laboratory, MCL, in Rockville, Maryland [10,11]. In this study, actionable mutation was defined as a mutation that was either the direct target or a pathway component that could be targeted by at least one approved or investigational drug. Patients who harbored actionable mutations were treated using genotype-matched targeted drugs. Approved drugs for the disease or another disease were given on or off-label, respectively. Drugs in clinical trials were considered if available. For instance, everolimus is approved for the treatment of renal cell carcinoma by Korea Food and Drug Administration; however, in this study, everolimus was used in parotid carcinosarcoma and tracheal squamous cell carcinoma with PIK3CA mutation. In this study, TP53 mutations were not considered actionable mutations as in a similar study [8].

thumbnail
Fig 1. Study scheme.

After quality check (QC), samples were sequenced by next generation sequencing (NGS). Medically fit patients with actionable mutation received matched targeted therapy, but patients who were not medically fit received chemotherapy or best supportive care (BSC).

https://doi.org/10.1371/journal.pone.0154133.g001

Study objectives

The primary objective of this study was the response rate (RR) as determine by RECIST v1.1 [12] in patients who received genotype-matched therapy. Firstly, radiologists read baseline CT scan, MRI or PET CT, and restaging images were read again after targeted therapy. Then, based on radiologist’ official report, medical oncologists evaluated the tumor response according to RECIST 1.1. Secondary objectives included time to progression (TTP) in the same subset and clinical feasibility of NGS-based targeted anticancer therapy.

Patient eligibility

From May 2014 to January 2015, patients with advanced cancer underwent molecular profiling with NGS with the intent of clinical application of available matched targeted agents at Severance hospital, Seoul, Korea. Key inclusion criteria were as follows: (1) age ≥ 19 years; (2) refractory solid tumor, which was defined as advanced solid tumor that was refractory to standard therapy and had no more evidence-based therapies; (3) available tumor tissue; (4) Eastern Cooperative Oncology Group (ECOG) performance status of 0–2 at enrollment; (5) appropriate organ functions which allow anti-cancer therapy; and (6) signed informed consent. The study protocol was approved by the Institutional Review Board of Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.

NGS

Tumor tissue was obtained from biopsy or surgery upon the initial diagnosis of cancer at the primary or metastasis site, and tumor genomic DNA was extracted from formalin-fixed and paraffin-embedded (FFPE) tumor tissue. Extracted tumor genomic DNA was sent to Foundation Medicine or Macrogen, the CLIA-certified laboratory in the USA, and library preparation using FoundationOne or CCP was conducted. CCP is a panel, targeting 409 genes which includes the exons of tumor suppressor genes and oncogenes frequently mutated [13]. The panel was designed to be amplification based capture with approximately 16,000 amplicons. Average amplicon size is 155 base pairs (125–175 base pairs), and it require a total of 40ng of DNA as a template for each sample. Sequencing was processed by Ion PGMTM system. FoundationOne is a pan-cancer panel, which is designed for 315 cancer related genes and 28 frequently rearranged genes [14].

Analysis platform of NGS data

Our in-house pipeline was applied to analyze CCP data. Sequencing data using CCP was aligned to the human reference genome build 19 and base calling was performed by Ion reporter 4.0 versions (Life Technologies). Variants acquired from the CCP panel were filtered by germline variants acquired from the Korean patients and 1000 genome data including Japanese and Chinese data [15]. We then annotated the variants using ANNOVAR [16], and non-coding regions and synonymous variants were filtered out. Mutations with low depth, which indicate ≤50x depths, were filtered out [17]. In addition, mutations with ≤5% variant allele frequency were filtered out [17]. Quality score, which is one parameter of the variant call format (VCF) using the phred scale, was used to filter out the variants, and Q30 was used for cut-off value [11,17]. Finally, we reviewed the mutation using the Broad’s Integrative Genomics Viewer [18]. Variants acquired from the CCP panel were validated by Sanger sequencing in selected actionable targets. In FoundationOne, a mutation list was provided by the service provider.

Results

Patient characteristics

A total of 36 patients were enrolled in the pilot study, and samples from 32 patients passed the quality control test for molecular analysis. Baseline characteristics are presented in Table 1. The median age was 48.5 years (range, 22–72 years), and 10 (31.3%) patients were ≤40 years old. Eleven NSCLC (34.4%) patients (10 adenocarcinomas and one neuroendocrine carcinoma) and four (12.4%) esophageal cancer patients were enrolled, and patients with various tumor types were enrolled in small numbers. Types of previous systemic therapy ranged from 1 to 5, and patients with ≥3 previous chemotherapy histories were 12 (37.5%).

thumbnail
Table 1. Patient characteristics at time when mutation profiling was performed.

https://doi.org/10.1371/journal.pone.0154133.t001

NGS test results

To identify somatic mutations, we performed the CCP platform on 31 samples and FoundationOne on one sample to a median depth of 823x and 562x, respectively. We identified 44 actionable mutations which were missense or truncation mutations. The list of actionable mutations in all patients is described in Table 2. Twenty-four (75%) of 32 patients had ≥1 actionable mutations; 11 (34.4%), 8 (25%), 3 (9.4%), and two (6.3%) patients had 1, 2, 3, and 4 actionable mutations, respectively. The frequency of each actionable mutation is shown in Fig 2, and the most common mutations were EGFR, ERBB2, ROS1, and PIK3CA mutations (3/32, 9.4%).

Outcome of genotype-matched therapy

Of 24 patients with actionable mutations, five received genotype-matched targeted therapy according to potential biological impact of mutations and availability of drugs in Korea, but 19 could not begin targeted therapy based on genomic tests because of declining performance status (10/24, 41.7%), stable disease with previous treatment (5/24, 20.8%), and lack of access to targeted medication (4/24, 16.7%). In particular, six of 11 NSCLC patients harbored actionable mutations but none were given genotype-matched therapy.

Of five patients who received genotype-matched targeted therapy, two showed partial response (PR), one showed stable disease (SD; −21%), but two had progressive disease (PD) just after initiation of matched therapy (Table 3). Everolimus was administered to PI3K/mTOR pathway genes-mutated patients. A parotid carcinosarcoma patient and tracheal squamous cell carcinoma patient who both had PIK3CA mutation (H1047R) showed PR and SD (−21%) to everolimus, respectively. The PIK3CA mutation (H1047R) in the parotid carcinoma patient was validated with Sanger sequencing. Before everolimus therapy, both patients received cisplatin based concurrent chemoradiotherapy, and tracheal squamous cell carcinoma patient additionally received gemcitabine and carboplatin chemotherapy, but they became refractory status. Two patients with PDGFRA or BRAF mutation received sorafenib which is known to target PDGFR and RAF. The external auditory canal adenocarcinoma patient, who had a PDGFRA mutation (L710F), achieved PR with sorafenib (Fig 3). Before sorafenib therapy, the patient received several lines of chemotherapy including cisplatin based CCRT, 5-fluorouracil/cisplatin, etoposide/cisplatin, and KX2-391/paclitaxel chemotherapy, but finally progressed to all regimens. However, the microcystic adnexal carcinoma of scalp patient with a BRAF mutation (D22G) showed PD to sorafenib within 1 month. Lastly, esophageal adenocarcinoma patient with ERBB2 mutation (P597H) received afatinib, which targets ERBB2, but showed PD within 1 month. Previous treatment history of patients with matched therapy was described in S1 Table. TTP in five patients who received genotype-matched therapy was 3.7 months (range, 0.7–6.7).

thumbnail
Fig 3. Tumor reduction in external auditory canal adenocarcinoma patient with PDGFRA mutation (L710F) after sorafenib treatment.

(A and B) multiple lung metastases disappeared and (C and D) huge anterior chest wall mass remarkably shrank after 4 weeks of sorafenib treatment.

https://doi.org/10.1371/journal.pone.0154133.g003

Discussion

For patients with refractory solid tumor, genome-based basket [19], umbrella [20], or phase I clinical trials [8] are drawing attentions. The basket trial allows patients with several tumor types with the same actionable target to be enrolled in trial. The other approach is umbrella trial, which is designed to test the impact of different drugs on different mutations in a single type of cancer, on the basis of molecular profile. However, these studies are only accessible in a limited number of countries. In a Korean institution, we evaluated the preliminary efficacy and clinical feasibility of NGS-based anticancer therapy in refractory solid tumors.

In this study, NGS-based genotype-matched therapy showed efficacy in patients with refractory solid tumors for which we did not have standard therapeutic options, although only small number of patients received genotype-matched therapy. Of five patients with refractory solid tumor who received genotype-matched therapy in this study, significant tumor reduction was seen in three patients (60%; two PRs, one decreasing SD by −21%). In a phase I Program at MD Anderson Cancer Center in the similar clinical setting, the response rate (RR) was 25% [8], whereas RR was only 4% in the patients with non-matched therapy [7]. However, a recently published SHIVA trial [21], which was a randomized, phase II trial to compare genome-based molecularly targeted therapy versus conventional therapy for advanced cancer, failed to show any improvement in survival or responses with genome-based targeted therapies. However, weakness of SHIVA trial can be found [22]: 1) patients with several co-existing molecular alterations are unlikely to respond to a single targeted therapy [23], 2) hormone monotherapy in heavily treated patients is unlikely to bring clinical response [24], 3) some targeted agents were incorrectly matched to molecular alterations [25], 4) treatment in the control group was offered at the discretion of physicians which may lead biases. Furthermore, two meta-analyses in 70,000 patients reported that trials with a personalized strategy led to a higher proportion of patients achieving responses and longer progression-free and overall survival than trials with unselected patients [26,27]. Therefore, it is hard to generally accept the conclusions of the SHIVA trial that precision therapy is disappointing and that the use of targeted drugs off-label should be discouraged. Taken together, although the current study is a small pilot trial in Korea, we suggest that NGS-based genotype-matched therapeutic approaches may be feasibly tested in larger trials and also may provide reasonable treatment options to refractory solid tumor patients in Korea in the future.

Two patients whose tumors responded to everolimus (one PR, one SD with 21% decrement) had the PIK3CA mutation (H1047R), which is already known to be a predictive marker for PI3K/AKT/mTOR pathway inhibitors [28]. An external auditory canal adenocarcinoma patient who showed PR to sorafenib harbored the PDGFRA mutation (L710F). The PDGFRA mutation is observed in approximately 7% of gastrointestinal stromal tumor, and 80% of the PDGFRA mutations are found in exon 18, which is located in the tyrosine kinase domain [29]. Our patient’s PDGFRA mutation (L710F) was located in the tyrosine kinase domain but was a novel mutation. A prediction of the functional effect of this novel mutation was performed by PolyPhen-2 [30] and possibly damaging was expected. Previous studies showed that sorafenib could inhibit the proliferation of cell lines or patient’s tumor with PDGFRA mutation [31,32]. The patient with a microcystic adnexal carcinoma of scalp who had a BRAF mutation (D22G) showed PD to sorafenib. That may be explained from the fact that the patient had a non-V600E BRAF mutation, which was not located in the kinase domain [33]. In addition, the patient’s poor general condition partly accounted for early PD. An esophageal adenocarcinoma patient with an ERBB2 mutation (P597H) also did not benefit from afatinib. Although the mutation locus was in the extracellular domain and could be related to ERBB2 activation [34,35], the patient also had a PIK3C2B mutation, which may cause PI3K pathway activation and subsequent resistance to afatinib.

In this study, we found several obstacles in terms of the clinical feasibility in performing NGS-based targeted anticancer therapy. First, there were no clinical-grade NGS assays available in Korea. Thus, we had to send samples for identifying patient’s mutation profile to a CLIA-certified laboratory in the USA. This made turn-around time about 4 weeks, which was too long for advanced cancer patients whose remaining survival time was not long. Clinical-grade NGS assays should be developed soon in Korea. Simpler hotspot cancer gene panels used in clinical practice may be sufficient to detect actionable mutations. The cost of NGS is also a challenging issue. It is currently not covered by either national health or private insurance in Korea. Therefore, in the near future, discussion should be started on the coverage of NGS by national health insurance or at least by private insurance once to twice per each cancer patient. Second, limited access to targeted medications was the most common reason that patients could not be treated. Genome-based basket or phase I trials, which can be offered to refractory solid tumor patients, are not enough in Korea. Thus, our patients were administered genotype-matched therapy off-label. Therefore, Korean medical oncologists should make vigorous efforts to quickly launch genome-based trials for refractory solid tumor patients.

This study has a few limitations. A very small number of patients were enrolled for evaluating the efficacy of genome-based matched therapy. We need more robust evidence for better clinical outcomes with NGS-based targeted therapy to recommend this approach in clinical practice. Second, paired normal samples were not sequenced for somatic mutation calling. This may be related to false-positive findings of somatic mutations [36]. To fix this problem, we used germline variants acquired from Korean patients and 1000 genome data to filter out false-positive findings in somatic mutations. Third, the novel mutation as an actionable target lacks evidence of functional changes in the protein, although the novel mutation is a missense mutation. It is difficult to predict the functional consequence of the novel mutation. Only functional in-vitro and in-vivo studies can identify the role of that mutation. As expected, the survival time of refractory solid tumor patients was short, and we did not have time to validate functional change of somatic mutations before we administered drugs to patients. However, some of actionable mutations are currently under functional validation.

In conclusion, NGS-based targeted therapy may be a good option in selected patients with refractory solid tumors. To pursue this strategy in Korea, lack of access to clinical-grade NGS assay and limited number of genotype-matched targeted medications needs to be addressed and resolved.

Supporting Information

S1 Table. Prior lines of therapies in patients who treated with NGS based targeted therapy.

https://doi.org/10.1371/journal.pone.0154133.s001

(DOCX)

Author Contributions

Conceived and designed the experiments: HSP HRK MGL JHK YWM. Performed the experiments: HSP YWM. Analyzed the data: HSP SRK SWK YWM. Contributed reagents/materials/analysis tools: HSP SML SRK SWK KK. Wrote the paper: HSP YWM.

References

  1. 1. Jung KW, Won YJ, Kong HJ, Oh CM, Cho H, Lee DH, et al. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2012. Cancer Res Treat. 2015;47: 127–141. pmid:25761484
  2. 2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65: 5–29. pmid:25559415
  3. 3. Lee KS, Chang HS, Lee SM, Park EC. Economic Burden of Cancer in Korea during 2000–2010. Cancer Res Treat. 2015;47: 387–398. pmid:25672582
  4. 4. Riely GJ, Pao W, Pham D, Li AR, Rizvi N, Venkatraman ES, et al. Clinical course of patients with non-small cell lung cancer and epidermal growth factor receptor exon 19 and exon 21 mutations treated with gefitinib or erlotinib. Clin Cancer Res. 2006;12: 839–844. pmid:16467097
  5. 5. Parkinson DR, Johnson BE, Sledge GW. Making personalized cancer medicine a reality: challenges and opportunities in the development of biomarkers and companion diagnostics. Clin Cancer Res. 2012;18: 619–624. pmid:22298894
  6. 6. Munoz J, Swanton C, Kurzrock R. Molecular profiling and the reclassification of cancer: divide and conquer. Am Soc Clin Oncol Educ Book. 2013: 127–134. pmid:23714478
  7. 7. Janku F, Berry DA, Gong J, Parsons HA, Stewart DJ, Kurzrock R. Outcomes of phase II clinical trials with single-agent therapies in advanced/metastatic non-small cell lung cancer published between 2000 and 2009. Clin Cancer Res. 2012;18: 6356–6363. pmid:23014530
  8. 8. 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: 6373–6383. pmid:22966018
  9. 9. Ahn SJ, Costa J, Emanuel JR. PicoGreen quantitation of DNA: effective evaluation of samples pre- or post-PCR. Nucleic Acids Res. 1996;24: 2623–2625. pmid:8692708
  10. 10. Singh RR, Patel KP, Routbort MJ, Aldape K, Lu X, Manekia J, et al. Clinical massively parallel next-generation sequencing analysis of 409 cancer-related genes for mutations and copy number variations in solid tumours. Br J Cancer. 2014;111: 2014–2023. pmid:25314059
  11. 11. Pant S, Weiner R, Marton MJ. Navigating the rapids: the development of regulated next-generation sequencing-based clinical trial assays and companion diagnostics. Front Oncol. 2014;4: 78. pmid:24860780
  12. 12. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45: 228–247. pmid:19097774
  13. 13. Weren RD, Ligtenberg MJ, Kets CM, de Voer RM, Verwiel ET, Spruijt L, et al. A germline homozygous mutation in the base-excision repair gene NTHL1 causes adenomatous polyposis and colorectal cancer. Nat Genet. 2015;47: 668–671. pmid:25938944
  14. 14. Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31: 1023–1031. pmid:24142049
  15. 15. Abecasis GR, Auton A, Brooks LD, DePristo MA, Durbin RM, Handsaker RE, et al. An integrated map of genetic variation from 1,092 human genomes. Nature. 2012;491: 56–65. pmid:23128226
  16. 16. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38: e164. pmid:20601685
  17. 17. Wang L, Yamaguchi S, Burstein MD, Terashima K, Chang K, Ng HK, et al. Novel somatic and germline mutations in intracranial germ cell tumours. Nature. 2014;511: 241–245. pmid:24896186
  18. 18. Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29: 24–26. pmid:21221095
  19. 19. Hyman DM, Puzanov I, Subbiah V, Faris JE, Chau I, Blay JY, et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N Engl J Med. 2015;373: 726–736. pmid:26287849
  20. 20. Kim ES, Herbst RS, Wistuba II, Lee JJ, Blumenschein GR Jr., Tsao A, et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov. 2011;1: 44–53. pmid:22586319
  21. 21. Le Tourneau C, Delord JP, Goncalves A, Gavoille C, Dubot C, Isambert N, 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. pmid:26342236
  22. 22. Tsimberidou AM, Kurzrock R. Precision medicine: lessons learned from the SHIVA trial. Lancet Oncol. 2015;16: e579–580. pmid:26678197
  23. 23. Janku F, Hong DS, Fu S, Piha-Paul SA, Naing A, Falchook GS, et al. Assessing PIK3CA and PTEN in early-phase trials with PI3K/AKT/mTOR inhibitors. Cell Rep. 2014;6: 377–387. pmid:24440717
  24. 24. Prat A, Baselga J. The role of hormonal therapy in the management of hormonal-receptor-positive breast cancer with co-expression of HER2. Nat Clin Pract Oncol. 2008;5: 531–542. pmid:18607391
  25. 25. Skinner MA, Safford SD, Freemerman AJ. RET tyrosine kinase and medullary thyroid cells are unaffected by clinical doses of STI571. Anticancer Res. 2003;23: 3601–3606. pmid:14666655
  26. 26. Schwaederle M, Zhao M, Lee JJ, Eggermont AM, Schilsky RL, Mendelsohn J, et al. Impact of Precision Medicine in Diverse Cancers: A Meta-Analysis of Phase II Clinical Trials. J Clin Oncol. 2015;33: 3817–3825. pmid:26304871
  27. 27. Jardim DL, Schwaederle M, Wei C, Lee JJ, Hong DS, Eggermont AM, et al. Impact of a Biomarker-Based Strategy on Oncology Drug Development: A Meta-analysis of Clinical Trials Leading to FDA Approval. J Natl Cancer Inst. 2015;107.
  28. 28. Janku F, Wheler JJ, Naing A, Falchook GS, Hong DS, Stepanek VM, et al. PIK3CA mutation H1047R is associated with response to PI3K/AKT/mTOR signaling pathway inhibitors in early-phase clinical trials. Cancer Res. 2013;73: 276–284. pmid:23066039
  29. 29. Corless CL, Schroeder A, Griffith D, Town A, McGreevey L, Harrell P, et al. PDGFRA mutations in gastrointestinal stromal tumors: frequency, spectrum and in vitro sensitivity to imatinib. J Clin Oncol. 2005;23: 5357–5364. pmid:15928335
  30. 30. Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, et al. A method and server for predicting damaging missense mutations. Nat Methods. 2010;7: 248–249. pmid:20354512
  31. 31. Heinrich MC, Marino-Enriquez A, Presnell A, Donsky RS, Griffith DJ, McKinley A, et al. Sorafenib inhibits many kinase mutations associated with drug-resistant gastrointestinal stromal tumors. Mol Cancer Ther. 2012;11: 1770–1780. pmid:22665524
  32. 32. Roubaud G, Kind M, Coindre JM, Maki RG, Bui B, Italiano A. Clinical activity of sorafenib in patients with advanced gastrointestinal stromal tumor bearing PDGFRA exon 18 mutation: a case series. Ann Oncol. 2012;23: 804–805. pmid:22294526
  33. 33. Gronych J, Korshunov A, Bageritz J, Milde T, Jugold M, Hambardzumyan D, et al. An activated mutant BRAF kinase domain is sufficient to induce pilocytic astrocytoma in mice. J Clin Invest. 2011;121: 1344–1348. pmid:21403401
  34. 34. Roskoski R Jr. The ErbB/HER family of protein-tyrosine kinases and cancer. Pharmacol Res. 2014;79: 34–74. pmid:24269963
  35. 35. Herter-Sprie GS, Greulich H, Wong KK. Activating Mutations in ERBB2 and Their Impact on Diagnostics and Treatment. Front Oncol. 2013;3: 86. pmid:23630663
  36. 36. Jones S, Anagnostou V, Lytle K, Parpart-Li S, Nesselbush M, Riley DR, et al. Personalized genomic analyses for cancer mutation discovery and interpretation. Sci Transl Med. 2015;7: 283ra253.