Clinical Cancer CCR New Strategies Research

New Strategies in Personalized Medicine for Solid Tumors: Molecular Markers and Clinical Trial Designs

Juliane M. Jurgensmeier,€ Joseph P. Eder, and Roy S. Herbst

Abstract The delineation of signaling pathways to understand tumor biology combined with the rapid develop- ment of technologies that allow broad molecular profiling and data analysis has led to a new era of personalized medicine in oncology. Many academic institutions now routinely profile patients and discuss their cases in meetings of personalized medicine tumor boards before making treatment recommendations. Clinical trials initiated by pharmaceutical companies often require specific markers for enrollment or at least explore multiple options for future markers. In addition to the still small number of targeted agents that are approved for the therapy of patients with histological and molecularly defined tumors, a broad range of novel targeted agents in development are undergoing clinical studies with companion profiling to determine the best-responding patient population. Although the present focus of profiling lies in genetic analyses, additional tests of RNA, protein, and immune parameters are being developed and incorporated in clinical research, and these methods are likely to contribute significantly to future patient selection and treatment approaches. As the advances in tumor biology and human genetics have identified promising tumor targets, the ongoing clinical evaluation of novel agents will now need to show if the promise can be translated into benefit for patients. Clin Cancer Res; 20(17); 4425–35. 2014 AACR.

Disclosure of Potential Conflicts of Interest J.M. Jurgensmeier€ was an employee of and reports receiving a commercial research grant from AstraZeneca. R.S. Herbst reports receiving a commercial research grant from Genentech and is a consultant/advisory board member for Biothera, Diatech, Kolltan, and N-of-One. No potential conflicts of interest were disclosed by the other author.

CME Staff Planners' Disclosures The members of the planning committee have no real or apparent conflict of interest to disclose.

Learning Objectives Upon completion of this activity, the participant should have a better understanding of the new era of personalized medicine in oncology, particularly the advancement of signaling pathways in tumor biology, the identification of promising tumor targets through rapid developments in molecular profiling and data analysis, and the ongoing clinical evaluation of novel agents.

Acknowledgment of Financial or Other Support This activity does not receive commercial support.

Background "-omics" approaches, have facilitated the discovery and Over the past decade, the delineation of signaling development of novel targeted anticancer therapeutics, or pathways to understand tumor biology has laid the foun- at least provided the scientific rationale for their devel- dation for the discovery of novel targets and development opment. This increased molecular understanding of of multiple therapies for cancer. The identification tumors has led to a new era of personalized medicine of driver mutations and critical pathway dependencies, that has begun to influence common practice for oncol- as well as genomic sequencing and other large-scale ogists beyond academic institutions. The National Cancer Institute (NCI) defines "person- alized medicine," often also called "precision medicine," Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut. as "a form of medicine that uses information about a Corresponding Author: Roy S. Herbst, Yale School of Medicine, 333 person’s genes, proteins, and environment to prevent, Cedar Street, WWW221, P.O. Box 208028, New Haven, CT 06520-8028. diagnose, and treat disease." Personalized medicine uses Phone: 203-785-6879; Fax: 203-737-5698; E-mail: [email protected] specific markers in patients’ tumors to diagnose partic- doi: 10.1158/1078-0432.CCR-13-0753 ular cancers or make treatment decisions. A specific 2014 American Association for Cancer Research. marker may have prognostic significance for biologic

www.aacrjournals.org 4425

Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2014 American Association for Cancer Research. Jurgensmeier€ et al.

behavior or predict therapeutic outcome with a particular Antihormonal agents such as tamoxifen have been used anticancer agent. This review focuses on predictive mar- for decades, initially without molecular profiling. Studies kers associated with tumors but does not discuss host later correlated outcome with the expression of estrogen genetic factors that also can determine treatment and receptor (ER), progesterone receptor (PR), and androgen prognosis. receptor (AR). The presence of ER, PR, and AR not only Traditionally, the site of tumor origin, together with categorizes breast and prostate cancers into different prog- histology, was used to make treatment decisions. This nostic groups, but also determines treatment. , approach has been changed to include molecular tumor a targeting human epithelial growth parameters. Markers presently used to guide decisions factor receptor-2 (HER-2), was approved for patient popu- for personalized medicine treatment with targeted agents lations with a specific molecular profile: HER-2–overexpres- are either protein based [immunohistochemistry (IHC)] sing breast and gastric cancer (3–5). The first agent targeting or based on detection of genetic aberrations and are sum- a chromosomal translocation, imatinib, was originally marized in Table 1. For genetic aberrations, a range of approved for the treatment of Philadelphia chromosome þ methods, including fluorescence in situ hybridization positive (Ph ) chronic myelogenous leukemia, based on (FISH) tests, polymerase chain reaction (PCR), sequenc- inhibiting the Bcr–Abl tyrosine kinase (6). In 2008, based ing—nowadays often next-generation sequencing (NGS) of on the additional activity of imatinib to inhibit c-Kit, either multiple selected genes in parallel or whole exome approval by the FDA for the treatment of adult patients sequencing—are used, in particular in early clinical trials or with c-Kit (CD117)–positive unresectable and/or metastat- for exploratory purposes. These large panels of genes offer ic gastrointestinal stromal tumors (GIST) followed (7, 8). the opportunity not only to detect aberrations for which Other recent successes in targeting molecularly defined tu- treatment options already exist but also to generate data on mor subtypes include the development of vemurafenib for additional driver mutations or resistance pathways (1, 2). advanced BRAFV600-mutated (9), and of crizo- Treatment decisions may be based on the data when tests tinib for patients with non–small cell lung cancer (NSCLC) have been performed in Clinical Laboratory Improvement with ALK translocations (10). Molecular profiling can also Amendment (CLIA)–certified laboratories. lead to a negative selection, as the retrospective studies

Table 1. Molecular selection markers for approved anticancer agents

Target Cancer Variation type Marker Drug Test EGFR Lung cancer Mutation Predict benefitto Erlotinib DNA EGFR TKIs Gefitinib ALK Lung cancer Rearrangement Predict response to Crizotinib FISH ALK inhibitors ROS Lung cancer Rearrangement Predict response to Crizotinib FISH TKIs RET Lung cancer Rearrangement Predict response to Vandetanib FISH TKIs BRAF Melanoma Mutation Predict response to Vemurafenib DNA BRAF inhibitors Dabrafenib KRAS Colorectal cancer Mutation Predict lack of response to DNA anti-EGFR antibodies HER2 Breast cancer Amplification Predict response to anti-HER2 Trastuzumab FISH, IHC Gastric cancer Overexpression antibodies Lapatinib KIT GIST Mutation Predict response to c-Kit Imatinib IHC inhibitors Estrogen receptor Breast cancer Overexpression Predict response Examestane IHC Fulvestrant Letrozole Tamoxifen Progesterone receptor Breast cancer Overexpression Predict response Examestane IHC Letrozole

4426 Clin Cancer Res; 20(17) September 1, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2014 American Association for Cancer Research. Molecular Markers for Personalized Medicine

showing consistent lack of benefit from the anti-EGFR feedback loop that increases EGFR expression (34). Multi- antibodies cetuximab or panitumumab in patients with ple ongoing studies are testing these hypotheses. Encour- colorectal cancer with KRAS codon 12/13 mutations led to aging preliminary results have come from studies of PI3K/ the recommendation to restrict the use of these antibodies AKT/mTOR inhibitors in PIK3CAmt-deficient or PTEN- to patients with wild-type KRAS tumors (11–14). deficient cancers (35). Lung cancer has become the prototype for genetically tailored cancer therapy. The remarkable advances in under- On the Horizon standing molecular drivers in NSCLC, together with the Many cancer centers now profile patients by genetic development of targeted agents, allow classification of testing, RNA expression profiling, or protein analyses and NSCLC on a molecular basis, and molecular profiling has use the obtained data to direct patients to clinical trials at become routine practice in thoracic oncology. EGFR muta- their centers or elsewhere (36, 37). Genetic profiling is tions were discovered in 2004 in parallel to the develop- booming, and molecular profiling is discussed in multi- ment of gefitinib, an EGFR tyrosine kinase inhibitor (TKI), disciplinary tumor boards and contributes to treatment when retrospective studies revealed that patients with recommendations. Some academic cancer centers now responses harbor exon 19 deletions or exon 21 point holdaweeklyprecisiontumorboardmeetinginwhich mutations (15, 16). These findings led to the use of the oncologists together with radiologists and molecular EGFR TKIs gefitinib and erlotinib as initial treatment for pathologists present cases. The multidisciplinary team, patients with these mutations. The discovery of EGFR muta- which also includes basic and translational scientists, tions and the large benefit of targeted treatment have surgeons, and nurses, discusses the cases and potential boosted profiling and the search for novel targets. The treatment options. Presently, molecular profiling is most- development of crizotinib to treat patients with ALK ly limited to genetic aberrations [mutations, transloca- fusions, detected by a FISH assay, highlights one of the tions, fusions, copy number variation (CNV)], but it is outstanding successes in lung cancer with rapid develop- planned to extend the profiling to include RNA and ment of a treatment (17). More recently, the additional immune profiling (Fig. 1). Although currently genetic activity of crizotinib in patients with ROS fusions (18, 19) profiling is only performed for patients for whom addi- was discovered, and patients with RET fusions are beginning tional data are critical to guide treatment, it is envisioned to be treated with the RET inhibitors vandetanib and car- that in the near future all patients treated at academic bozantinib (20–24). cancer centers will have a diagnostic tumor sample ana- Although these targeted therapies have brought signifi- lyzed for genetic aberrations, and with repeat-biopsy cant improvements, all patients eventually develop thera- programs in place, ideally also at progression. The ulti- peutic resistance. Multiple resistance mechanisms have mate goal of the profiling is to find the best available been characterized, such as secondary mutations preventing treatment for the patient, mostly by enrollment in a inhibitor binding, EGFR or HER2 gene amplifications, clinical trial, if such a trial exists. There is an urgent need HGF/Met pathway activations as well as PI3K and BRAF for more studies with molecularly targeted agents that are mutations (25, 26). Multiple resistance mechanisms have open for patients across tumor types to investigate if these also been identified in patients with ALK fusions who treatments can benefit other patient populations. progress on crizotinib. These include ALK mutations, copy Several institutions and companies have launched trials gain number, and mutations in alternative pathways, that assign patients based on molecular profiling of tumors including EGFR mutations. Second- and third-generation in specific cancer types (BATTLE I and II; ref. 38), but also inhibitors of EGFR and ALK, such as AZD9291 with activity independent of cancer type. These investigations include against EGFRT790M and alectinib or ceritinib, especially observational studies as well as nonrandomized and ran- active against the L11986M ALK mutation that confers domized studies (2, 39–41). Nonrandomized studies build- resistance to crizotinib, are now in development with initial ing on the ability of academic cancer centers to perform promising data (17, 27–32). molecular profiling in CLIA-certified laboratories have been Multiple deregulated pathways have been identified initiated in 2013 by pharmaceutical companies making a across a range of tumor types, which could potentially be number of agents available for use in molecularly defined targeted by novel agents (summarized in Table 2). However, patient populations. These include agents for which safety often several genetic aberrations are found in the same data already exist and a phase II dose is defined, and exclude tumor and it is not always clear which are driver mutations, patient populations for which the drugs are either already which are secondary changes, and which are the determi- registered or for which dedicated randomized studies exist nants of inherent or acquired resistance. Moreover, muta- or populations for which a lack of benefit for the agent was tions that clearly act as driver mutations in one tumor type already observed. For example, the Novartis SIGNATURE may not be of similar relevance in another tumor. For studies offer independent trials with investigational agents example, patients with malignant melanoma with BRAF from a pharmaceutical company for patients with a specific V600E mutations respond to treatment with vemurafenib, molecular profile (42). These agents would otherwise not be (33), whereas patients with colorectal cancer that harbor available for patients with many tumor types, as specific the same mutation seem to derive little benefit from BRAF studies in all tumor types do not exist. The NCI plans to inhibitors because of complex mechanisms that include a open a similar study in 2014, NCI-MATCH, which is

www.aacrjournals.org Clin Cancer Res; 20(17) September 1, 2014 4427

Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2014 American Association for Cancer Research. 4428 lnCne e;2(7 etme ,2014 1, September 20(17) Res; Cancer Clin J Downloaded from resee tal. et urgensmeier € Table 2. Genetic aberrations as putative predictive biomarkers for anticancer agents

Examples for Aberration type Putative or proven drugs in clinical Target/pathway in solid tumors Disease examples drugs development References

clincancerres.aacrjournals.org EGFR Mutation Lung cancer EGFR inhibitors Erlotinib (15, 16, 25, 26, 30) Amplification GBM Gefitinib Afatinib AZD9291 HER2 (ERB2) Mutation Breast cancer ERB2/ERB3 inhibitors Lapatinib (4, 5) Amplification Gastric cancer Neratinib Lung cancer Trastuzumab Trastuzumab-emtansine ALK Rearrangement Lung cancer ALK inhibitors Crizotinib (10, 22, 28, 29, 65, 66) Mutation Neuroblastoma Ceritinib Colorectal cancer Alectinib on September 23, 2021. © 2014American Association for Cancer RET Rearrangement Thyroid cancer RET inhibitors Vandetanib (19–21, 23) Research. Mutation Lung cancer Carbozantinib ROS1 Rearrangement Lung cancer ROS1 inhibitors Crizotinib (18, 19) DDR2 Mutation Lung cancer DDR2 inhibitors Dasatinib (67, 68) Nilotinib FGFR1-4 Amplification Lung cancer FGFR inhibitors Ponatinib (69, 70) Mutation Gastric cancer Dovitinib Breast cancer BGJ398 Bladder cancer AZD4547 MET/HGF Mutation Lung cancer Met inhibitors (71–73) Amplification Gastric cancer Crizotinib Colorectal cancer Foretinib HCC INC280 RCC lnclCne Research Cancer Clinical KIT Mutation GIST Kit inhibitors Imatinib (7, 8) Mastocytosis Nilotinib Melanoma Sunitinib Dasatinib Ponatinib

(Continued on the following page) www.aacrjournals.org

Downloaded from Table 2. Genetic aberrations as putative predictive biomarkers for anticancer agents (Cont'd )

Examples for Aberration type Putative or proven drugs in clinical Target/pathway in solid tumors Disease examples drugs development References PDGFRA and PDGFRB Mutation GIST PDGFR inhibitors Imatinib (8, 74) Translocation Sarcoma Sunitinib clincancerres.aacrjournals.org GBM Ponatinib Leukemia Dermatofibrosarcoma protuberans KRAS, NRAS, HRAS Mutation Most cancers, including Mek inhibitors Trametinib (75) (RAS-RAF-MEK) colorectal cancer, Selumetinib lung cancers BRAF Mutation Melanoma Braf inhibitors Vemurafenib (75, 76) (RAS-RAF-MEK) Colorectal cancer Mek inhibitors Dabrafenib HCC Trametinib

on September 23, 2021. © 2014American Association for Cancer Selumetinib

Research. PI3KCA Mutation Multiple, including: PI3K inhibitors BKM-120 (77, 78) (PTEN/PI3K/AKT/mTOR) Amplification Breast cancer AKT inhibitors BEZ235 Colorectal cancer, GBM BYL719 Lung cancer GDC0941 Endometrial GDC0032 MLN1117 AKT inhibitors (see below) PIK3R1 Mutation Endometrial cancer PI3K inhibitors PI3K inhibitors (see above) (78)

(PTEN/PI3K/AKT/mTOR) Colorectal cancer Medicine Personalized for Markers Molecular lnCne e;2(7 etme ,2014 1, September 20(17) Res; Cancer Clin AKT1-3 Mutation Breast cancer PI3K inhibitors GDC0086 (77–79) (PTEN/PI3K/AKT/mTOR) Amplification Colorectal cancer AKT inhibitors MK2206 Meningeal cancer AZD5363 Urinary tract cancers PI3K inhibitors (see above) Endometrial cancer PTEN Deletion Most cancers, including PI3K inhibitors PI3K and AKT inhibitors (77–80) (PTEN/PI3K/AKT/mTOR) Mutation breast cancer AKT inhibitors (see above) Lung cancer Colorectal cancer (Continued on the following page) 4429 4430 lnCne e;2(7 etme ,2014 1, September 20(17) Res; Cancer Clin J Downloaded from resee tal. et urgensmeier Table 2. Genetic aberrations as putative predictive biomarkers for anticancer agents (Cont'd ) €

Examples for Aberration type Putative or proven drugs in clinical Target/pathway in solid tumors Disease examples drugs development References mTOR Mutation Endometrial cancer mTOR inhibitors Everolimus (78, 81, 82)

clincancerres.aacrjournals.org (PTEN/PI3K/AKT/mTOR) RCC Temsirolimus Colorectal cancer MLN128 Lung cancer AZD2014 GDC00980 BEZ235

TSC1/2 Mutation Tuberous sclerosis mTOR inhibitors mTOR inhibitors (see above) (78, 83) (PTEN/PI3K/AKT/mTOR) Urinary tract cancers Endometrial cancer Cervical cancer HCC

on September 23, 2021. © 2014American Association for Cancer Colorectal cancer

Research. LKB1 Mutation Cervical cancer mTOR inhibitors mTOR inhibitors (see above) (78) (PTEN/PI3K/AKT/mTOR) Small intestine cancer Lung cancer Skin cancer SMO, PTCH1 Mutation Basal cell carcinoma Hedgehog inhibitors Vismodegib (84, 85) (Hedgehog) Medulloblastoma Meningioma Breast cancer MDM2 Amplification GBM MDM2 inhibitors/antagonists RG7388 (53, 54) Sarcoma disrupting p53-MDM2 interaction P53 Mutation Most tumors P53 activators (53, 54) NOTCH Mutation Breast cancer g-secretase inhibitors MK0752 (86, 87) Rearrangement Lung cancer ABs to notch receptors or PF03084014 lnclCne Research Cancer Clinical Ovarian cancer ligands GBM OMP59R5 H&N cancer OMP52M51 Enoticumab CDKS Amplification Sarcoma CDK inhibitors Flavopiridol (88) Mutation Melanoma Palbociclib Rearrangement GBM

(Continued on the following page) www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org

Table 2. Genetic aberrations as putative predictive biomarkers for anticancer agents (Cont'd )

Examples for Aberration type Putative or proven drugs in clinical Target/pathway in solid tumors Disease examples drugs development References CHK1/2 Mutation Multiple tumors, including CHK inhibitors RG7741 (88) those listed below: LY2606368 Colorectal cancer Gastric cancer on September 23, 2021. © 2014American Association for Cancer Endometrial cancer Research. Breast cancer AURKA Amplification Multiple tumors Aurora kinase inhibitors Alisertib (88, 89) (Aurora kinases) ATR Mutation Gastric cancer ATM inhibitors No ATM inhibitors in clinical development (88, 90) Deletion Breast cancer PARP inhibitors PARP inhibitors (see below) Endometrial cancer ATM Mutation Multiple tumors, including ATR inhibitors VX970 (88, 90) Deletion Breast cancer PARP inhibitors PARP inhibitors (see below) oeua akr o esnlzdMedicine Personalized for Markers Molecular lnCne e;2(7 etme ,2014 1, September 20(17) Res; Cancer Clin BRCA1/2 Mutation Breast cancer PARP inhibitors Olaparib (91, 92) Ovarian cancer Veliparib Rucaparib BMN673

Abbreviations: GBM, glioblastoma multiforme; H&N cancer, head and neck cancer; HCC, hepatocellular carcinoma; RCC, renal cell carcinoma. 4431 Jurgensmeier€ et al.

Protein detection Genetic RNA profiling Immune • Selected markers (e.g., • Expression PTEN, ER, PR, AR, and HER2) profiling • miRNA profiling • Broad profiling

Tumor board treatment decision Figure 1. The academic medical center precision medicine tumor board model.

Clinical trials, Clinical trials, Center-specific Treatment outside NCI studies company A company B studies of institution

© 2014 American Association for Cancer Research

expected to include agents from multiple companies (43). tance critical for future direction. Multiplexing also requires For the NCI-MATCH studies, genomic profiling will be less tissue than multiple single tests. The latter is becoming performed by a consortium of NCI-selected CLIA-certified an increasing problem for phase I trials, for which compa- laboratories using NGS of a defined number of genes as the nies require large amounts of tissue for testing to evaluate basis for enrollment. The arms of the NCI-MATCH study are eligibility that exhausts archival diagnostic tissue, resulting still under discussion. It is expected to include agents from in patients requiring new biopsies or even depriving them multiple companies under NCI sponsorship and will val- of enrollment if a new sample cannot be obtained. idate the proposed broad sequencing platform. The SHIVA Advanced technology has revealed intratumor and inter- study randomizes patients with a particular molecular tumor heterogeneity at the protein, genetic, and epigenetic abnormality with any type of cancer between the matched level (44, 45). Genetic analyses, RNA and protein profiling agent and conventional cytotoxic therapy, with crossover of primary carcinomas, and metastases from the same on progression (39). The SHIVA study is different from the patients with renal carcinomas showed a large degree of other two studies mentioned here in that it has system-on- heterogeneity with most detected genetic aberrations not Chip (SoC) comparator arms for each newly tested agent consistent between lesions and tumor regions that confirm and will therefore collect data on the outcome of SoC that multiple clonal subpopulations exist within a single therapies in molecular segments. More studies of this type lesion (46, 47). In contrast, genetic analyses of colorectal with agents in development would be of high interest, but and lung cancer lesions showed more concordant data for a such studies are difficult to initiate and coordinate if they limited gene set (48, 49). Indeed, the effectiveness of avail- involve multiple companies. The results of these ongoing able targeted treatments for patients with advanced cancer studies are likely to identify additional patient populations such as gefitinib in EGFRmt lung cancer and cetuximab or for targeted therapy and hopefully will aid registration for panitumumab in KRASwt colorectal cancer has largely been additional indications. Data collection is critical for the demonstrated in clinical trials that identified the mutations future direction of personalized medicine and may trigger from archival diagnostic biopsies. In light of the heteroge- additional basic research to fully understand the contribu- neity, it is questionable, if a single archival biopsy taken for tions of newly discovered mutations to tumor development diagnostic purposes in early stages of the disease is sufficient and progression. as a sample for molecular profiling to guide treatment. Even Is testing for certain mutations sufficient or should broad if a new biopsy is available for analysis, is one sample testing be applied to all samples? Single mutation or small sufficient, if multiple lesions may have a very different panel testing has the clear advantage of requiring smaller molecular profile? How many areas of a single tumor need amounts of tissue, is less costly, and makes interpretation of to be biopsied to obtain comprehensive information of the data simpler and hence often quicker. Although there are drivers of the tumor in a patient? Answers to these questions only a few tests that clearly direct patient treatment at this are urgently needed. time (see Table 1), broader testing will facilitate better Given the emergence of resistance as described above, it is understanding of tumor drivers and mechanisms of resis- expected that future patients’ tumors will need to be profiled

4432 Clin Cancer Res; 20(17) September 1, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2014 American Association for Cancer Research. Molecular Markers for Personalized Medicine

at several times during treatment in order to determine the Other areas presently being explored are RNA profiling best treatment options. Indeed, some academic cancer (mRNA and miRNA), epigenetic profiling, and immune centers have repeat-biopsy programs in place, allowing profiling (62–64). RNA analyses will strengthen the them to pursue new biopsies when patients progress on understanding of deregulated signaling pathways in can- targeted treatment, in particular with EGFR or ALK inhibi- cer that genetic profiling alone may not reveal and may tors. How realistic are multiple longitudinal biopsies for a lead to novel hypotheses for targets. Immune profiling majority of patients with cancer across tumor types, and do will become important as new therapies, especially we urgently need to increase our efforts to further explore immune checkpoint therapies, demonstrate striking activ- other technologies that may be able to assess tumor muta- ity in several cancer types. tions without multiple longitudinal biopsies or parallel Although great progress has been made toward molecular biopsies of different lesions? profiling for personalized cancer therapy, it is expected that To minimize invasive procedures, the analysis of cfDNA with rapidly developing technology, molecular determi- or circulating tumor cells should be explored further to nants other than genetic aberrations may emerge that will evaluate if these technologies can replace multiple biopsies contribute to treatment decisions. The next 5 years will for molecular analysis, especially as patients move through decide if personalized medicine will become standard prac- successive lines of treatment. Initial results are promising tice across all tumor types and if this will revolutionize (50–52). Is genetic profiling really enough? How do we treatment options for greater survival benefit for patients approach the many targets for which treatment options do with cancer. not yet exist, including the ones that are presently consid- ered undruggable, like TP53 and KRAS (53–56)? Do we Authors' Contributions need broader molecular profiling that includes other end- Conception and design: J.M. Jurgensmeier,€ J.P. Eder, R.S. Herbst points to strengthen scientific understanding and other Development of methodology: J.M. Jurgensmeier,€ R.S. Herbst selection methods, especially if they can be applied to Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): R.S. Herbst samples other than tumor biopsies? Analysis and interpretation of data (e.g., statistical analysis, biosta- Serum proteins have been established as general prog- tistics, computational analysis): J.M. Jurgensmeier,€ R.S. Herbst € nostic factors, for example PSA levels in patients with Writing, review, and/or revision of the manuscript: J.M. Jurgensmeier, J.P. Eder, R.S. Herbst prostate cancer. Carcinoembryonic antigen (CEA) is mea- Administrative, technical, or material support (i.e., reporting or orga- sured as a tumor marker across multiple tumor types, and nizing data, constructing databases): J.M. Jurgensmeier,€ R.S. Herbst € high levels have been shown to correlate with poor Study supervision: J.M. Jurgensmeier, J.P. Eder, R.S. Herbst prognosis (57–59). Attempts to identify serum proteins as predictors for response to targeted agents so far have Grant Support had limited success (60, 61), but this has not been This work was supported by the NIH (2P30CA016359-34; to J.P. Eder, through his institution) and the NIH/NCI (1R01CA155196; to assessed broadly. Furthermore, serum marker studies in R.S. Herbst). clinical trials should be encouraged, especially if tumor samples are available at the baseline of these trials and Received March 18, 2014; revised June 11, 2014; accepted June 12, 2014; couldbecomparedwithserumassays. published online August 14, 2014.

References 1. MacConaill LE. Existing and emerging technologies for tumor genomic 9. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, profiling. J Clin Oncol 2013;31:1815–24. et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N 2. Tran B, Dancey JE, Kamel-Reid S, McPherson JD, Bedard PL, Brown Engl J Med 2010;363:809–19. AM, et al. Cancer genomics: technology, discovery, and translation. J 10. Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, et al. Clin Oncol 2012;30:647–60. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N 3. BaselgaJ,TripathyD,MendelsohnJ,BaughmanS,BenzCC, Engl J Med 2010;363:1693–703. Dantis L, et al. Phase II study of weekly intravenous recombinant 11. Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, humanized anti-p185HER2 monoclonal antibody in patients with et al. Wild-type KRAS is required for panitumumab efficacy in patients HER2/neu-overexpressing metastatic breast cancer. J Clin Oncol with metastatic colorectal cancer. J Clin Oncol 2008;26:1626–34. 1996;14:737–44. 12. Adelstein BA, Dobbins TA, Harris CA, Marschner IC, Ward RL. A 4. Figueroa-Magalhaes MC, Jelovac D, Connolly RM, Wolff AC. Treat- systematic review and meta-analysis of KRAS status as the determi- ment of HER2-positive breast cancer. Breast 2014;23:128–36. nant of response to anti-EGFR antibodies and the impact of partner 5. de Mello RA, Marques AM, Araujo A. HER2 therapies and chemotherapy in metastatic colorectal cancer. Eur J Cancer gastric cancer: a step forward. World J Gastroenterol 2013;19: 2011;47:1343–54. 6165–9. 13. Allegra CJ, Jessup JM, Somerfield MR, Hamilton SR, Hammond EH, 6. Thambi P, Sausville EA. STI571 (imatinib mesylate): the tale of a Hayes DF, et al. American Society of Clinical Oncology provisional targeted therapy. Anticancer Drugs 2002;13:111–4. clinical opinion: testing for KRAS gene mutations in patients with 7. Ashman LK, Griffith R. Therapeutic targeting of c-KIT in cancer. Expert metastatic colorectal carcinoma to predict response to anti-epidermal Opin Investig Drugs 2013;22:103–15. receptor monoclonal antibody therapy. J Clin Oncol 8. Corless CL, Barnett CM, Heinrich MC. Gastrointestinal stromal 2009;27:2091–6. tumours: origin and molecular oncology. Nat Rev Cancer 2011;11: 14. Jimeno A, Messersmith WA, Hirsch FR, Franklin WA, Eckhardt SG. 865–78. KRAS mutations and sensitivity to epidermal growth factor receptor

www.aacrjournals.org Clin Cancer Res; 20(17) September 1, 2014 4433

Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2014 American Association for Cancer Research. Jurgensmeier€ et al.

inhibitors in colorectal cancer: practical application of patient selec- tumors to find potential targets and select treatments for their refrac- tion. J Clin Oncol 2009;27:1130–6. tory cancers. J Clin Oncol 2010;28:4877–83. 15. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, 37. Meric-Bernstam F, Farhangfar C, Mendelsohn J, Mills GB. Building a Brannigan BW, et al. Activating mutations in the epidermal growth personalized medicine infrastructure at a major cancer center. J Clin factor receptor underlying responsiveness of non-small-cell lung can- Oncol 2013;31:1849–57. cer to gefitinib. N Engl J Med 2004;350:2129–39. 38. Kim ES, Herbst RS, Wistuba II, Lee JJ, Blumenschein GR Jr, Tsao A, 16. Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer mutations in lung cancer: correlation with clinical response to gefitinib Discov 2011;1:44–53. therapy. Science 2004;304:1497–500. 39. Le Tourneau C, Kamal M, Tredan O, Delord JP, Campone M, Goncalves 17. Camidge DR, Bang YJ, Kwak EL, Iafrate AJ, Varella-Garcia M, Fox SB, A, et al. Designs and challenges for personalized medicine studies in et al. Activity and safety of crizotinib in patients with ALK-positive non- oncology: focus on the SHIVA trial. Target Oncol 2012;7:253–65. small-cell lung cancer: updated results from a phase 1 study. Lancet 40. Tsimberidou AM, Iskander NG, Hong DS, Wheler JJ, Falchook GS, Fu Oncol 2012;13:1011–9. S, et al. Personalized medicine in a phase I clinical trials program: the 18. Bergethon K, Shaw AT, Ou SH, Katayama R, Lovly CM, McDonald NT, MD Anderson Cancer Center initiative. Clin Cancer Res 2012;18: et al. ROS1 rearrangements define a unique molecular class of lung 6373–83. cancers. J Clin Oncol 2012;30:863–70. 41. Arnedos M, Andre F, Farace F, Lacroix L, Besse B, Robert C, et al. The 19. Gainor JF, Shaw AT. Novel targets in non-small cell lung cancer: ROS1 challenge to bring personalized cancer medicine from clinical trials into and RET fusions. Oncologist 2013;18:865–75. routine clinical practice: the case of the Institut Gustave Roussy. Mol 20. Kohno T, Ichikawa H, Totoki Y, Yasuda K, Hiramoto M, Nammo T, et al. Oncol 2012;6:204–10. KIF5B-RET fusions in lung adenocarcinoma. Nat Med 2012;18:375–7. 42. ClinicalTrials.gov. Novartis Pharmaceuticals, NCT01831726, NCT 21. Kohno T, Tsuta K, Tsuchihara K, Nakaoku T, Yoh K, Goto K. RET fusion 01833169, NCT 01885195, NCT 01981187, NCT 02002689. clinical- gene: translation to personalized lung cancer therapy. Cancer Sci trials.gov. 2013;104:1396–400. 43. Conley BA. Precision cancer medicine; exceptional responders; NCI- 22. Lipson D, Capelletti M, Yelensky R, Otto G, Parker A, Jarosz M, et al. MATCH. Available from: http://deainfo.nci.nih.gov/advisory/ctac/ Identification of new ALK and RET gene fusions from colorectal and 1113/Precision%20Cancer%20Medicine.pdf. lung cancer biopsies. Nat Med 2012;18:382–4. 44. Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and 23. Drilon A, Wang L, Hasanovic A, Suehara Y, Lipson D, Stephens P, et al. consequences of genetic heterogeneity in cancer evolution. Nature Response to Cabozantinib in patients with RET fusion-positive lung 2013;501:338–45. adenocarcinomas. Cancer Discov 2013;3:630–5. 45. Awada A, Aftimos PG. Targeted therapies of solid cancers: new 24. Gainor JF, Shaw AT. The new kid on the block: RET in lung cancer. options, new challenges. Curr Opin Oncol 2013;25:296–304. Cancer Discov 2013;3:604–6. 46. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos 25. Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, E, et al. Intratumor heterogeneity and branched evolution revealed by Fidias P, et al. Genotypic and histological evolution of lung cancers multiregion sequencing. N Engl J Med 2012;366:883–92. acquiring resistance to EGFR inhibitors. Sci Transl Med 2011;3: 47. Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant 75ra26. metastasis occurs late during the genetic evolution of pancreatic 26. Gainor JF, Shaw AT. Emerging paradigms in the development of cancer. Nature 2010;467:1114–7. resistance to tyrosine kinase inhibitors in lung cancer. J Clin Oncol 48. Vakiani E, Janakiraman M, Shen R, Sinha R, Zeng Z, Shia J, et al. 2013;31:3987–96. Comparative genomic analysis of primary versus metastatic colorectal 27. Solomon B, Wilner KD, Shaw AT. Current status of targeted therapy for carcinomas. J Clin Oncol 2012;30:2956–62. anaplastic lymphoma kinase-rearranged non-small cell lung cancer. 49. Jakobsen JN, Sorensen JB. Intratumor heterogeneity and chemother- Clin Pharmacol Ther 2013;95:15–23. apy-induced changes in EGFR status in non-small cell lung cancer. 28. Sakamoto H, Tsukaguchi T, Hiroshima S, Kodama T, Kobayashi T, Cancer Chemother Pharmacol 2012;69:289–99. Fukami TA, et al. CH5424802, a selective ALK inhibitor capable of 50. Narayan A, Carriero NJ, Gettinger SN, Kluytenaar J, Kozak KR, Yock TI, blocking the resistant gatekeeper mutant. Cancer Cell 2011;19: et al. Ultrasensitive measurement of hotspot mutations in tumor DNA in 679–90. blood using error-suppressed multiplexed deep sequencing. Cancer 29. Seto T, Kiura K, Nishio M, Nakagawa K, Maemondo M, Inoue A, et al. Res 2012;72:3492–8. CH5424802 (RO5424802) for patients with ALK-rearranged advanced 51. Perkins G, Yap TA, Pope L, Cassidy AM, Dukes JP, Riisnaes R, et al. non-small-cell lung cancer (AF-001JP study): a single-arm, open-label, Multi-purpose utility of circulating plasma DNA testing in patients with phase 1–2 study. Lancet Oncol 2013;14:590–8. advanced cancers. PLoS ONE 2012;7:e47020. 30. Ranson M PW, Planchard D, Ohe Y, Kim S-W, Kim DW, Felip E, et al. 52. Yu M, Stott S, Toner M, Maheswaran S, Haber DA. Circulating tumor AZD9291; an irreversible potent and selective tyrosine kinase inhibitor cells: approaches to isolation and characterization. J Cell Biol of activating (EGFRmþ) and resistance (EGFRmþ/T790Mþ) mutations 2011;192:373–82. in NSCLC. J Thorac Oncol 2013;8:S389. 53. Wade M, Li YC, Wahl GM. MDM2, MDMX and p53 in oncogenesis and 31. Yu HA, Pao W. Targeted therapies: Afatinib–new therapy option for cancer therapy. Nat Rev Cancer 2013;13:83–96. EGFR-mutant lung cancer. Nat Rev Clin Oncol 2013;10:551–2. 54. Shangary S, Wang S. Targeting the MDM2-p53 interaction for cancer 32. Berardi R, Santoni M, Morgese F, Ballatore Z, Savini A, Onofri A, et al. therapy. Clin Cancer Res 2008;14:5318–24. Novel small molecule EGFR inhibitors as candidate drugs in non-small 55. Maurer T, Garrenton LS, Oh A, Pitts K, Anderson DJ, Skelton NJ, et al. cell lung cancer. Onco Targets Ther 2013;6:563–76. Small-molecule ligands bind to a distinct pocket in Ras and inhibit 33. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, SOS-mediated nucleotide exchange activity. Proc Natl Acad Sci U S A et al. Improved survival with vemurafenib in melanoma with BRAF 2012;109:5299–304. V600E mutation. N Engl J Med 2011;364:2507–16. 56. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras(G12C) 34. Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, inhibitors allosterically control GTP affinity and effector interactions. et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition Nature 2013;503:548–51. through feedback activation of EGFR. Nature 2012;483:100–3. 57. Ebeling FG, Stieber P, Untch M, Nagel D, Konecny GE, Schmitt UM, 35. Janku F, Wheler JJ, Westin SN, Moulder SL, Naing A, Tsimberidou AM, et al. Serum CEA and CA 15-3 as prognostic factors in primary breast et al. PI3K/AKT/mTOR inhibitors in patients with breast and gyneco- cancer. Br J Cancer 2002;86:1217–22. logic malignancies harboring PIK3CA mutations. J Clin Oncol 58. Okamoto T, Nakamura T, Ikeda J, Maruyama R, Shoji F, Miyake T, et al. 2012;30:777–82. Serum carcinoembryonic antigen as a predictive marker for sensitivity 36. Von Hoff DD, Stephenson JJ Jr, Rosen P, Loesch DM, Borad MJ, to gefitinib in advanced non-small cell lung cancer. Eur J Cancer Anthony S, et al. Pilot study using molecular profiling of patients' 2005;41:1286–90.

4434 Clin Cancer Res; 20(17) September 1, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2014 American Association for Cancer Research. Molecular Markers for Personalized Medicine

59. Jurgensmeier€ JM, Schmoll HJ, Robertson JD, Brooks L, Taboada M, 76. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Morgan SR, et al. Prognostic and predictive value of VEGF, sVEGFR-2 Mutations of the BRAF gene in human cancer. Nature 2002;417: and CEA in mCRC studies comparing cediranib, and 949–54. chemotherapy. Br J Cancer 2013;108:1316–23. 77. Chalhoub N, Baker SJ. PTEN and the PI3-kinase pathway in cancer. 60. Hanrahan EO, Ryan AJ, Mann H, Kennedy SJ, Langmuir P, Natale RB, Annu Rev Pathol 2009;4:127–50. et al. Baseline vascular endothelial growth factor concentration as a 78. Polivka J Jr, Janku F. Molecular targets for cancer therapy in the PI3K/ potential predictive marker of benefit from vandetanib in non-small cell AKT/mTOR pathway. Pharmacol Ther 2014;142:164–75. lung cancer. Clin Cancer Res 2009;15:3600–9. 79. Carpten JD, Faber AL, Horn C, Donoho GP, Briggs SL, Robbins CM, 61. Spencer SK, Pommier AJ, Morgan SR, Barry ST, Robertson JD, Hoff et al. A transforming mutation in the pleckstrin homology domain of PM, et al. Prognostic/predictive value of 207 serum factors in colo- AKT1 in cancer. Nature 2007;448:439–44. rectal cancer treated with cediranib and/or chemotherapy. Br J Cancer 80. Hollander MC, Blumenthal GM, Dennis PA. PTEN loss in the continuum 2013;109:2765–73. of common cancers, rare syndromes and mouse models. Nat Rev 62. Costa V, Aprile M, Esposito R, Ciccodicola A. RNA-Seq and human Cancer 2011;11:289–301. complex diseases: recent accomplishments and future perspectives. 81. Sato T, Nakashima A, Guo L, Coffman K, Tamanoi F. Single amino-acid Eur J Hum Genet 2013;21:134–42. changes that confer constitutive activation of mTOR are discovered in 63. Hauptman N, Glavac D. Long non-coding RNA in cancer. Int J Mol Sci human cancer. Oncogene 2010;29:2746–52. 2013;14:4655–69. 82. Hardt M, Chantaravisoot N, Tamanoi F. Activating mutations of TOR 64. Weichenhan D, Plass C. The evolving epigenome. Hum Mol Genet (target of rapamycin). Genes Cells 2011;16:141–51. 2013;22:R1–6. 83. Chan JA, Zhang H, Roberts PS, Jozwiak S, Wieslawa G, Lewin- 65. Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Kowalik J, et al. Pathogenesis of tuberous sclerosis subependymal et al. Identification of the transforming EML4-ALK fusion gene in non- giant cell astrocytomas: biallelic inactivation of TSC1 or TSC2 small-cell lung cancer. Nature 2007;448:561–6. leads to mTOR activation. J Neuropathol Exp Neurol 2004;63: 66. Chen Y, Takita J, Choi YL, Kato M, Ohira M, Sanada M, et al. 1236–42. Oncogenic mutations of ALK kinase in neuroblastoma. Nature 84. Xie J, Johnson RL, Zhang X, Bare JW, Waldman FM, Cogen PH, et al. 2008;455:971–4. Mutations of the PATCHED gene in several types of sporadic extra- 67. Day E, Waters B, Spiegel K, Alnadaf T, Manley PW, Buchdunger E, et al. cutaneous tumors. Cancer Res 1997;57:2369–72. Inhibition of collagen-induced discoidin domain receptor 1 and 2 85. Xie J, Murone M, Luoh SM, Ryan A, Gu Q, Zhang C, et al. Activating activation by imatinib, nilotinib and dasatinib. Eur J Pharmacol smoothened mutations in sporadic basal-cell carcinoma. Nature 2008;599:44–53. 1998;391:90–2. 68. Hammerman PS, Sos ML, Ramos AH, Xu C, Dutt A, Zhou W, et al. 86. Takebe N, Nguyen D, Yang SX. Targeting Notch signaling pathway in Mutations in the DDR2 kinase gene identify a novel therapeutic target cancer: clinical development advances and challenges. Pharmacol in squamous cell lung cancer. Cancer Discov 2011;1:78–89. Ther 2014;141:140–9. 69. Turner N, Grose R. Fibroblast growth factor signalling: from develop- 87. Shih Ie M, Wang TL. Notch signaling, gamma-secretase inhibitors, and ment to cancer. Nat Rev Cancer 2010;10:116–29. cancer therapy. Cancer Res 2007;67:1879–82. 70. Kelleher FC, O'Sullivan H, Smyth E, McDermott R, Viterbo A. Fibroblast 88. Lapenna S, Giordano A. Cell cycle kinases as therapeutic targets for growth factor receptors, developmental corruption and malignant cancer. Nat Rev Drug Discov 2009;8:547–66. disease. Carcinogenesis 2013;34:2198–205. 89. Lens SM, Voest EE, Medema RH. Shared and separate functions of 71. GherardiE,BirchmeierW,BirchmeierC,VandeWoudeG.TargetingMET polo-like kinases and aurora kinases in cancer. Nat Rev Cancer in cancer: rationale and progress. Nat Rev Cancer 2012;12:89–103. 2010;10:825–41. 72. Sierra JR, Tsao MS. c-MET as a potential therapeutic target and 90. Fokas E, Prevo R, Hammond EM, Brunner TB, McKenna WG, Muschel biomarker in cancer. Ther Adv Med Oncol 2011;3:S21–35. RJ. Targeting ATR in DNA damage response and cancer therapeutics. 73. Ghiso E, Giordano S. Targeting MET: why, where and how? Curr Opin Cancer Treat Rev 2014;40:109–17. Pharmacol 2013;13:511–8. 91. Rosen EM, Pishvaian MJ. Targeting the BRCA1/2 Tumor Suppressors. 74. Board R, Jayson GC. Platelet-derived growth factor receptor Curr Drug Targets 2014;15:17–31. (PDGFR): a target for anticancer therapeutics. Drug Resist Updat 92. Hennessy BT, Timms KM, Carey MS, Gutin A, Meyer LA, Flake 2005;8:75–83. DD II, et al. Somatic mutations in BRCA1 and BRCA2 could 75. Neuzillet C, Tijeras-Raballand A, de Mestier L, Cros J, Faivre S, expand the number of patients that benefitfrompoly(ADPribose) Raymond E. MEK in cancer and cancer therapy. Pharmacol Ther polymerase inhibitors in ovarian cancer. J Clin Oncol 2010;28: 2014;141:160–71. 3570–6.

www.aacrjournals.org Clin Cancer Res; 20(17) September 1, 2014 4435

Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2014 American Association for Cancer Research. New Strategies in Personalized Medicine for Solid Tumors: Molecular Markers and Clinical Trial Designs

Juliane M. Jürgensmeier, Joseph P. Eder and Roy S. Herbst

Clin Cancer Res 2014;20:4425-4435.

Updated version Access the most recent version of this article at: http://clincancerres.aacrjournals.org/content/20/17/4425

Cited articles This article cites 90 articles, 28 of which you can access for free at: http://clincancerres.aacrjournals.org/content/20/17/4425.full#ref-list-1

Citing articles This article has been cited by 1 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/20/17/4425.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/20/17/4425. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2014 American Association for Cancer Research.