Review of Precision Cancer Medicine: Evolution of the Treatment Paradigm (original) (raw)

Cancer Treat Rev. Author manuscript; available in PMC 2021 Jun 1.

Published in final edited form as:

PMCID: PMC7272286

NIHMSID: NIHMS1580737

Apostolia M. Tsimberidou, MD, PhD,1 Elena Fountzilas, MD, PhD,2 Mina Nikanjam, MD, PhD,3 and Razelle Kurzrock, MD3

Apostolia M. Tsimberidou

1The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX

Elena Fountzilas

2Department of Medical Oncology, Euromedica General Clinic, Thessaloniki, Greece

Mina Nikanjam

3Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA

Razelle Kurzrock

3Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA

1The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX

2Department of Medical Oncology, Euromedica General Clinic, Thessaloniki, Greece

3Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA

Authors’ contributions

All authors wrote and approved the paper.

Corresponding author: Apostolia-Maria Tsimberidou, MD, PhD, Tenured Professor, The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Unit 455, 1515 Holcombe Boulevard, Houston, TX 77030, Phone: 713-792-4259, Fax: 713-794-3249, gro.nosrednadm@rebmista

Abstract

In recent years, biotechnological breakthroughs have led to identification of complex and unique biologic features associated with carcinogenesis. Tumor and cell-free DNA profiling, immune markers, and proteomic and RNA analyses are used to identify these characteristics for optimization of anticancer therapy in individual patients. Consequently, clinical trials have evolved, shifting from tumor type-centered to gene-directed, histology-agnostic, with innovative adaptive design tailored to biomarker profiling with the goal to improve treatment outcomes. A plethora of precision medicine trials have been conducted. The majority of these trials demonstrated that matched therapy is associated with superior outcomes compared to non-matched therapy across tumor types and in specific cancers. To improve the implementation of precision medicine, this approach should be used early in the course of the disease, and patients should have complete tumor profiling and access to effective matched therapy. To overcome the complexity of tumor biology, clinical trials with combinations of gene-targeted therapy with immune-targeted approaches (e.g., checkpoint blockade, personalized vaccines and/or chimeric antigen receptor T-cells), hormonal therapy, chemotherapy and/or novel agents should be considered. These studies should target dynamic changes in tumor biologic abnormalities, eliminating minimal residual disease, and eradicating significant subclones that confer resistance to treatment. Mining and expansion of real-world data, facilitated by the use of advanced computer data processing capabilities, may contribute to validation of information to predict new applications for medicines. In this review, we summarize the clinical trials and discuss challenges and opportunities to accelerate the implementation of precision oncology.

Keywords: ctDNA, personalized, precision, molecular profile, matched therapy, genomic landscape

Background

The rapidly expanding body of knowledge about the roles of genomics and the immune system in cancer has enabled the development of therapies targeted to specific molecular alterations or other biologic characteristics, such as those implicated in immune suppression. However, genomics has also revealed a complicated reality about malignancies that requires a major shift in the therapy paradigm: away from tumor type-centered and toward gene-directed, histology-agnostic treatment, which is individualized for each patient on the basis of biomarker analysis. This paradigm shift is reflected by the emergence of precision medicine trials with innovative design.121 Next-generation sequencing (NGS) of advanced cancers has demonstrated that genomic alterations do not fall neatly into categories defined by the tumor organ of origin. Furthermore, metastatic tumors harbor tremendously complex and individually unique genomic and immune landscapes.22,23 Therefore, in order to target malignancies with “precision,” treatment needs to be personalized.

Historically, phase II and III oncology clinical trials have measured outcomes histologically, but histological assessment cannot always capture the effects of gene-targeted agents or immunotherapy. Precision medicine approaches analyze patients’ circulating DNA (liquid biopsy), as well as immune markers and other biologic features, to assess efficacy and make treatment decisions. Genomic biomarkers have been the most successful to date, but other biomarkers, including protein assays and transcriptomics, are being developed and tested.13,24,25 Several molecular alterations have been identified using sequencing and high-throughput technologies and have led to the approval of targeted agents by the Food and Drug Administration (FDA).26,27 Importantly, in recent years, the precision medicine paradigm has embraced immunotherapy and its interaction with genomics, as genomic characteristics, such as mismatch repair gene defects, are critical predictors of checkpoint blockade response.2830

Herein, we review the rapid evolution of precision medicine in oncology and, in particular, the challenge and opportunity that genomic science has revealed vis-à-vis the need for “N-of-1” treatments. This treatment model does not conform to either canonical trial design or clinical practice, which seek to find commonalities between patients and treat them alike; instead, its goal is to provide optimized individualized treatment for each patient on the basis of biomarker analysis.

History

Survival improvement with gene- or immune-directed therapy was accelerated by several major discoveries. In particular, the introduction of imatinib mesylate (Abl tyrosine kinase inhibitor) for patients with Philadelphia chromosome [t(9;22)]–positive chronic myelogenous leukemia producing the enzymatically aberrant Bcr-Abl31,32 resulted in near-normal life expectancy for patients with this previously fatal leukemia.

In 2001, the human genome was sequenced.33 Although this milestone represented an arduous and tremendously expensive endeavour, both the price and time required for sequencing have decreased precipitously, with technology advancing in a manner unparalleled in human history. A plethora of first- and second-generation precision medicine trials have since been conducted (Tables 1 and ​2). They include, but are not limited to, the first pan-histology biomarker-driven trial using mostly protein markers,1 the prospective molecular profiling of patients with advanced cancer in the phase I clinical trials setting (IMPACT trial)2,4, the SHIVA randomized trial,5 trials assessing customized combinations6,12, and trials including transcriptomics.13

Table 1:

Examples of Precision Medicine Trials: Design and Outcomes

Year First/Last author Trial name Trial type No of pts screened (N) Proportion of pts. matched Biomarker(s) Outcome Institute(s) Comments
Diverse treatment-refractory tumor types
20101 Von Hoff D Penny R Bisgrove Prospective, navigational 86 77% IHC, FISH, microarray 27% of 66 matched pts had a PFS2/PFS1 ratio* ≥1.3 (95% CI, 17% to 38%; p = 0.007). US (9 sites)
20122 Tsimberidou A Kurzrock R IMPACT, first cohort Registry type, Navigational 1144 15% PCR-based genomics, 9 genes Matched vs unmatchedRR, 27% vs. 5% (p<0.0001),TTF: median, 5.2 vs. 2.2 mos (p<0.0001)OS: median, 13.4 vs. 9.0 mos (p = 0.017) MD Anderson Cancer Center
20143 Tsimberidou A Berry D IMPACT, second cohort Registry type, navigational 1276 11% PCR-based genomics, 18–50 genes Matched vs unmatchedRR, 11.9% vs. 5% (p<0.0001),PFS: median, 3.9 vs. 2.2 mos, (p=0.001);OS: median, 11.4 vs. 8.6 mos (p=0.04) MD Anderson Cancer Center 2-month landmark analyses, matched therapy group: OS, responders 30.5 months vs. 11.3 months for non-responders (p = 0.01).
20174 Tsimberidou AM Kurzrock R IMPACT, third cohort Registry type, navigational 1436 27% PCR-based genomics and NGS, 11 to 182 genes Matched vs unmatchedHigher rates of ORR (p=0.0099),TTF (p=0.0015), and OS (p=0.04) MD Anderson Cancer Center
20155 Le Tourneau Paoletti X SHIVA Prospective, randomized 741 13% Targeted NGS, ~50 genes PFS not improved with matched therapy (p=0.41) Institut Curie, 8 French sites ~80% of patients received single-agent hormone modulators or everolimus
20166 Schwaederle M Kurzrock R PREDICT Registry type 347 25% NGS, 182 or 236 genes Matched vs unmatchedHigher rates of SD≥ 6 months/PR/CR (p=0.02) and PFS (p<0.04).Higher matching scores correlated with better OS: 15.7 vs 10.6 mos (p=0.04) University of California San Diego
20167 Wheler JJ Kurzrock R MD Anderson Personalized Cancer Therapy Initiative Prospective, navigational 500 24% NGS, 236 genes Higher matching scores correlated with higher rates of SD ≥6 months/PR/CR (p=0.024), TTF (p=0.0003), and OS (p=0.05) MD Anderson Cancer Center
20168 Stockley TL Bedard PL IMPACT/COMPACT Prospective 1893 5% Hot spot panel, 23 genes Matched vs unmatchedHigher ORR: 19% vs 9%, (p=0.026). PrincessMargaret,Canadian centers
20179 Massard C Soria JC MOSCATO Prospective 1035 19% Targeted NGS, 40–75 genes; aCGH; RNAseq PFS2/PFS1 ratio* was >1.3 in 33% (63/193) of patients InstitutGustaveRoussy
201810 Hainsworth JD Kurzrock R MyPathway Prospective, Phase 2 basket 251 Not available Genomic testing via any CLIA lab Matched patients, ORR: All, 23%_HER2_-altered, 38%_BR4F_-altered, 43% Multiple sites, Genentech 251 patients enrolled; 230 were treated; however, how many were screened pre-enrollment is unknown
201911 Tredan O Blay JY Profiler Prospective 2579 6% NGS, 69 genes RR = 13% (23 of 182 treated) Four institutes (France)
201912 Sicklick J Kurzrock R I-PREDICT Prospective, navigational 149 49% NGS, 315 genes; ctDNA; PDL1 IHC Higher matching scores correlated with increased rates of SD≥6 months/PR/CR: 50% vs 22.4% (p=0.028), PFS (p=0.0004), and OS (p=0.038) University of California San Diego and Avera First trial to administer customized combination therapy (“N-of-1” matching)
201913 Rodon J Kurzrock R WINTHER Prospective, navigational 303 35% NGS, 236 genes; transcriptomics Higher matching scores correlated with longer PFS (p=0.005) and OS (p= 0.03) Five countries (Spain, Israel, France, Canada, US) First solid tumor trial to include transcriptomics
Specific tumors—Lung
201114 Kim ES Hong WK BATTLE Prospective, adaptive, randomized 255 Not available 11 biomarkers 8-week disease control rate, 46% MD Anderson Cancer Center It is unclear how many patients were screened before consent
201415 Kris MG Bunn PA Lung cancer mutation consortium Prospective 1537 17% Multiplex genotyping, 10 genes Improved OS with matched vs unmatched therapy (p=0.006) 14 US sites
201616 Aisner D Kwiatkowski DJ Lung Cancer Mutation Consortium II Prospective 904 12% NGS, minimum of 14 genes Improved survival with matched therapy (p<0.001) 16 sites
201617 Papadimitrakopoulou V Herbst RS BATTLE-2 Prospective, adaptive, randomized 334 Non-applicable ALK, FISH, EGFR, and KRAS Sanger sequencing KRAS alterations: longer PFS without erlotinib (p=0.04); KRAS wild-type tumors: longer OS on erlotinib (p=0.03) MD Anderson Cancer Center
Specific tumors—Breast
201218 Esserman LJ Hylton N I-SPY 1 Neoadjuvant, correlative 237 Non-applicable IHC pCR differs by subset Multiple US sites Aim was to develop biomarkers of response to conventional therapy
201519 Andre F Bonnefoi H SAFIR01/UNICANCER Prospective 423 13% Sanger sequencing (2 genes: PIK3CA and AKT); aCGH Matched group, ORR 9% 18 centers in France
201620,21 Park JW Berry DARugo HS Esserman LJ I-SPY 2 Phase 2 adaptive design, neoadjuvant Non-applicable Non-applicable IHC, Mammaprint Improved pCR rates in 2 study arms with drug addition:HER2+, hormone receptor-negative: neratinib plus standard therapy (N=115) vs standard therapy (N=78): 56% vs 33%Triple-negative: veliparib plus carboplatin (N=72) with standard therapy vs standard therapy (N=44): 51% vs 26% Quantum-Leap Healthcare (US sites) Results for 2 arms of I-SPY-2 study available
Specific tumors—Gastric
2019122 Lee J WK Kang VICTORY Prospective 772 14% NGS, IHC, PDL1, MMR and EBV status Improved PFS and OS with matched vs unmatched therapy (p<0.0001) Republic of Korea The trial included 10 phase II trials that operated independently (based on eight biomarkers)

Table 2:

Selected ongoing studies of precision medicine

Year started Trial name Trial type Cancer type Biomarker NCT number Institute(s) Comment
201020,21 I-SPY 2 Prospective randomized Neoadjuvant breast cancer IHC, Mammaprint NCT01042379 Quantum-Leap Healthcare, US sites Ongoing study with preliminary results (see Table 1)
2012123 SPECTA-Color Registry type Advanced colorectal cancer NGS/IHC NCT01723969 European hospitals
2013 MPACT Prospective Advanced cancer NGS NCT01827384 NCI, US sites
2014124 ALCHEMIST Prospective Early stage non-small cell lung cancer Direct sequencing, FISH, CLIA certified genotyping NCT02194738 NCI, US sites
201434 Lung-MAP Prospective Advanced squamous cell lung cancer NGS NCT02154490NCT02785913NCT02965378NCT02785939NCT02785952NCT02926638NCT02766335 NCI, US sites
2014125 AURORA Registry type Metastatic breast cancer NGS/RNAseq NCT02102165 Institut JulesBordet,Brussels,Belgium,Europeanhospitals
2014126 Signature Prospective Advanced cancers Variable NCT02187783NCT02186821 Novartis, multiple sites
201410 MyPathway Prospective Advanced cancers Genomic testing NCT02091141 Genentech, US sites
2014 IMPACT2 Prospective, randomized Metastatic cancer Genomic testing NCT02152254 MD Anderson Cancer Center
2014127 Pangea Prospective Gastro-esophageal adenocarcinoma Tumor biomarker profiling/cell-free DNA NCT02213289 University of Chicago
2015128131 NCI-MATCH Prospective Advanced cancers NGS NCT02465060 NCI, US sites
201512 I-PREDICT Prospective navigational Advanced cancers including treatment-naïve patients CGP NCT02534675 UC San Diego Avera Ongoing study with preliminary results (see Table 1)
2016 DART Prospective Rare cancers NGS correlational testing: whole genomic, transcriptome, liquid biopsy (ctDNA), and immune signature NCT02834013 SWOG/NCI, multiple US sites
2016132 TAPUR Prospective Advanced cancers Genomic analysis or IHC NCT02693535 ASCO, US sites
2016 DRUP Prospective Advanced cancers NGS NCT02925234 Netherlands
2017 Pediatric MATCH Prospective Pediatric advanced Cancers CLIA-certified molecular testing NCT03155620 NCI-COG, US sites
2018 Columbia University N- of-1 Clinical Trials Prospective Metastatic cancer Computational strategies (OncoTarget and OncoTreat) Columbia University

Innovative clinical trial designs for precision medicine

Traditionally, oncology trials are drug-centered, aiming to identify common attributes among patients (e.g., their tumor type or, more recently, a shared genomic abnormality) and fit them into a trial with a specific drug regimen. The large variability in genomic subgroups, microenvironment, baseline characteristics, comorbidities, and other covariates resulted in tumor-specific clinical studies encompassing a tremendously heterogeneous population in histology-specific, gene-agnostic trials. Phase III randomized trials were often critical for regulatory approval of a novel agent/regimen, especially since the antitumor activity of a new drug/regimen was frequently only marginally better than the comparator arm (usually, conventional therapy), perhaps because the regimen was effective in only a small subgroup of the diverse population represented by any specific histology.

Basket, umbrella, platform, octopus, and master protocols:

More recently, basket designs have emerged that target a common genetic defect27. The 75% objective response rate noted across tumor types with larotrectinib, which targets NTRK fusions, best exemplifies the potential of the basket gene-directed, histology-agnostic model, though other single-gene targets have proven much less responsive.27 Umbrella trials involve a single histology and different treatments based on the genomic alterations in patient subgroups.34 Other trial designs include platform trials, which use a single analytic technique, such as NGS, to identify genomic or other biomarkers in tumors with multiple histologies; octopus trials (also referred to as “complete phase I trials”) that have multiple arms testing different combinations featuring a particular drug; and master protocols, which encompass trials with several histologic arms (previously, “broad phase II trials”) or multiple platform, basket, or umbrella trials or sub-trials.24,6 Randomization has also evolved, with the emergence of Bayesian adaptation, which allows dynamic modifications of randomization based on small numbers of patients and realtime outcomes.

From drug-centered to patient-centered studies:

The ultimate goal of precision medicine is an individualized, patient-centered (rather than drug-centered) trial based on the best available biomarkers. In “N-of-1” trials, each patient’s treatment is considered separately on the basis of molecular, immune, and other biologic characteristics. These trials involve customized drug combinations tailored to individual patients.12 Determining efficacy in “N-of-1” trials requires assessing the “strategy” of matching patients to drugs, rather than treatments, which differ from patient to patient.

Genomic and other biomarkers

Genomics has been the cornerstone of precision medicine studies. Beyond genomics, RNA and protein profiling, with proteins being the effectors of signaling, also appear to be important in mediating biologic impact. Interestingly, matching patients to drugs on the basis of genomics has proven more effective in improving outcome than matching on the basis of protein assays, perhaps for technical reasons24. Despite the current practical limitations, protein and transcript assays may provide essential information when integrated with genomics.13 Recently, panels that incorporate immune signatures, based on DNA, RNA, and/or proteins, have also gained clinical significance.35

Genomics:

Given the advances in NGS technologies and the large number of laboratories in the US that perform Clinical Laboratory Improvement Amendments (CLIA)-certified NGS, optimization of the accuracy, reproducibility, and standardization of sequencing methods; variant annotation; and data interpretation is critical. Guidelines for the validation of NGS panels36 and the interpretation and reporting of genomic variants have been developed37. Although whole-genome sequencing is not yet the standard practice in the clinic, the FDA has approved two NGS panels that include hundreds of genes.38

Most genomic sequencing involves tissue, but blood-derived circulating tumor DNA (ctDNA), circulating tumor cells39, and exosomes40 are increasingly used, with the latter two reflecting the contents of live cells.

Blood-derived cell-free DNA analysis:

Clinical-grade ctDNA testing, which is non-invasive and reflects tumor heterogeneity (because tumor DNA may be leaked into the bloodstream from multiple metastatic lesions), is increasingly being used to select anti-cancer therapy and to monitor subclone dynamics during treatment.41,42 The discordance noted in some cases between results of ctDNA testing and tumor tissue genotyping analysis43 could reflect technical issues but might be attributable to the following biologic reasons: (i) tumor NGS measures genomics in the small piece of tissue biopsied while ctDNA assesses shed DNA from multiple sites; (ii) ctDNA is associated with tumor load and can be detected at low levels.

Blood-derived circulating tumor cell (CTC) analysis:

The presence of CTCs, which are epithelial tumor cells, has been independently associated with worse survival in several types of cancer.4446 For example, in a prospective, multicenter, double-blind study, the number of CTCs in patients with untreated metastatic breast cancer correlated with shorter progression-free survival (PFS) and overall survival (OS).44 CTCs may also be a predictive biomarker for chemotherapy and immunotherapy.45,47 However, the use of CTCs in clinical practice has not been fully established.48 Finally, serial CTC analyses might enable real-time surveillance of the disease. A comparative study of five prospective randomized phase III trials in 6,081 patients with metastatic castration-resistant prostate cancer assessed the prognostic value of CTCs compared to prostate-specific antigen.49 CTC ≥0 at baseline and at week 13 from treatment initiation was associated with OS. The investigators demonstrated that CTC monitoring was a robust and meaningful response endpoint for early-phase clinical trials in this setting.49

Transcriptomics:

Transcriptomics refers to the study of RNA transcripts and their function. Transcriptomic analysis is performed using high-throughput technologies, including microarrays and RNA sequencing and it is a potentially valuable tool, particularly when there is discrepancy between genomic alterations and gene expression. Transcriptomics are utilized to identify prognostic and predictive gene expression signatures50,51, to explore miRNAs and their role in mRNA regulation52,53 and to identify the tissue of origin in cancer of unknown primary.5456 The first solid tumor precision medicine trial to use transcriptomics in the clinic--WINTHER---compared RNA expression in tumors to that in adjacent normal tissue and demonstrated that transcriptomics increased the number of patients that could be matched to therapy.13 Comparing tumor to normal tissue from the same patient may be necessary because of the large inter-patient variability in normal RNA expression. Other investigators have also used transcriptomics to select targeted treatments in patients with advanced solid tumors.57,58 Challenges that prevent extensive use of transcriptomic biomarkers are degradation and fragmentation of RNA in formalin-fixed, paraffin-embedded tissue samples, complexity of required bioinformatic analysis of profiling data and low reproducibility of the results.

Proteomics:

Proteomic analysis using immunohistochemical and other assays of tumors from patients with refractory metastatic cancer led to the identification of molecular targets that could guide therapeutic decisions and was associated with longer PFS compared to the patients’ PFS with their prior therapy (using patients as their own controls).1 Proteomic assays are used in clinical practice to identify prognostic or predictive biomarkers for targeted treatments (hormone receptor expression, HER2 overexpression, ALK expression). However, the weaker correlation of proteomic markers, compared to genomic markers, with clinical outcomes suggests that technical issues should be addressed.24 In a meta-analysis of phase 1 clinical trials of small molecules that used a genomic biomarker vs. those that used a protein biomarker, the median response rate was 41% vs. 25%, respectively (p = 0.05).24 Ongoing studies with targeted therapies include correlative analyses using peripheral blood and tumor tissue to identify proteomic biomarkers of response or resistance to treatment (LEEomic, NCT03613220 and BABST-C, NCT03743428).

Immunotherapy and cellular therapy

By reactivating the innate immune antitumor response, immunotherapy has provided a major breakthrough in oncology treatment.28,59 Several novel approaches are currently being explored: checkpoint blockade, oncolytic viruses, cell-based products, modified cytokines, CD3-bispecific antibodies, vaccine platforms, and adoptive cell therapy.60

Checkpoint blockade:

There are seven FDA-approved checkpoint inhibitors: ipilimumab, pembrolizumab, nivolumab, avelumab, cemiplimab, durvalumab, and atezolizumab. Selected patients with advanced disease have remarkable response, including durable complete remission (CR). Despite the significant benefit noted in patients with diverse tumor types treated with checkpoint inhibitors, approximately 80% of patients across cancers do not experience beneficial effects. In the era of precision medicine, genomics, transcriptomics and other technologies are employed for the identification of biomarkers that predict benefit from immunotherapy. Interestingly, biomarkers predicting checkpoint inhibitor responsiveness are genomic: high tumor mutational burden (TMB)28,59,61, mismatch gene repair defects resulting in high microsatellite instability (MSI-H) (and, thus, high TMB)29,62, PBRM1 alterations63,64, and PDL1 amplification.65 Specifically, TMB has been shown to predict clinical benefit from checkpoint inhibitors.66 In an analysis of 151 of 1,638 patients who were treated with immunotherapeutic regimens and had TMB evaluation, high (≥ 20 mutations/mb) TBM was independently associated with significant improvement in PFS and OS compared to low to intermediate TMB.66 Other studies have however questioned the use of TMB as a biomarker.67,68

Given its strong association with response to immunotherapy, MSI-H is an established biomarker for response to checkpoint inhibitors.69,70 MSI-H tumors have high TMB, often accumulating >1,000 non-synonymous genomic mutations, leading to tumor-specific proteins, known as neoantigens. Due to high clinical benefit rates, immunotherapeutic regimens have been approved by the FDA for the treatment of patients with advanced MSI-H colorectal cancer7173 or MSI-H tumors, irrespective of the organ of origin.74 Finally, defects in DNA proofreading proteins polymerase δ (POLD1) and polymerase ε (POLE) lead to increased TMB and are associated with response to immunotherapy.7577 For instance, of 4 patients with non–small cell lung cancer with deleterious mutations in POLD1 and POLE (whole-exome sequencing, [WES]), 3 patients with the highest TMB responded to pembrolizumab.75 Defects in other DNA repair systems might also be associated with response to immunotherapy. The predictive role of homologous recombination deficiency (HRD) is being evaluated in various tumors, including breast and ovarian cancer. Early phase clinical trials demonstrating that these patients may benefit from the addition of immunotherapy to poly ADP-ribose polymerase (PARP) inhibitors, should be confirmed with additional studies.78,79

Furthermore, PBRM1 molecular alterations are evaluated as genomic biomarkers predicting checkpoint inhibitor responsiveness. Specifically, PBRM1 alterations were evaluated in a study of 35 patients with metastatic renal cell cancer treated with anti-programmed death-1 (PD-1) regimens.63 WES revealed loss-of-function (LOF) mutations in the PBRM1 gene that predicted response to immunotherapy. Notably, the PBRM1 gene encodes for a protein of the chromatin remodeling complex, possibly interfering with hypoxia, and immune signaling pathways.63

Another biomarker that predicts benefit from immunotherapy is PD-L1 amplification.65 In a retrospective analysis, this marker was identified in 0.7% (843 of 118,187) patients of various tumor types and it did not always correlate with PD-L1 expression. Six of 9 (66.7%) patients with PD-L1-amplified solid tumors had an objective response to checkpoint inhibitors, and their median PFS was 15.2 months.65 PDL1 expression, assessed by immunohistochemistry on tumor cells or immune cells can be used as a response marker, albeit a suboptimal one.80 Approximately 20% of FDA approvals of immunotherapeutic agents are based on companion PD-L1 diagnostic testing.81

Genomic markers may also predict resistance---loss of JAK2 and beta 2 microglobulin mutations82—or hyper-progression (accelerated progression) after checkpoint blockade---MDM2 amplification and EGFR alterations.83 WES of tumor tissue from 4 patients with advanced melanoma whose disease was resistant to anti–PD1 therapy, demonstrated LOF mutations in genes involved in interferon-receptor signaling and in antigen presentation (JAK1/2, β2-microglobulin).82 Importantly, PTEN loss is associated with resistance to immunotherapy in patients with melanoma, suggesting that targeting the PI3K/AKT/mTOR pathway may overcome resistance to immunotherapy.84 In our opinion, it is plausible that when PI3K/AKT/mTOR pathway alterations or PTEN loss are the key drivers of the disease, immunotherapy may have limited, if any, antitumor activity. Similarly, STK11 mutations and β-catenin pathway alterations are reportedly associated with resistance to immunotherapy.85,86

In summary, the available biomarkers are insufficient to adequately predict response to immunotherapy. Novel strategies may enhance our ability to identify biomarkers longitudinally, incorporating ctDNA analysis87 or tumor tissue immune, genomic, transcriptomic, and proteomic analysis.

Adoptive cell therapy

Adoptive cell therapy (ACT) is an innovative personalized treatment approach that enhances a patient’s immune system leading to specific tumor cell killing. Immune cells derived from a patient’s blood or tissue are expanded in vitro and then reinfused into the patient. These immune cells may be reprogrammed to recognize tumor-specific antigens.60,88 Types of ACT include tumor-infiltrating lymphocyte (TIL) therapy, chimeric antigen receptor (CAR) T-cell therapy, engineered T-cell receptor (TCR) therapy and natural killer (NK) cell therapy.

TILs:

ACT of TILs is based on the use of T-cells that have infiltrated a patient’s tumor. Autologous cells are being harvested and administered to patients after their expansion and activation. This approach has shown promising results in metastatic melanoma8992, nasopharyngeal, and cervical carcinoma.93,94 In three sequential clinical trials in patients with metastatic melanoma who had failed standard therapy, the use of autologous TILs was associated with objective response rates of 49%, 52%, and 72%, respectively; durable CRs were reported in 22% (20 of 93) of patients; and clinical benefit was observed irrespectively of prior therapy.89 Ongoing clinical trials assess the role of TIL therapy in various solid tumors (NCT03645928, NCT03935893, NCT03108495, NCT03083873).

TCR therapy:

This approach uses T-cell receptor (TCR) engineered T-cells, and involves retroviruses that enable integration of new TCR transgene targeting antigens, which are expressed at high levels on different cancers into the genome of T-cells.98 TCR therapy has been assessed in hematologic and solid malignancies.99103 Current trials evaluate treatment-associated toxicity, binding affinity to tumor antigens and efficacy in carefully selected patients with increased tumor burden.

NK cell therapy:

Natural killer (NK) cells are cytotoxic lymphocytes that play a critical role in innate immunity. NK cells do not cause graft-versus-host disease, which makes them promising candidates for cancer treatment. Treatment of relapsed/refractory acute myeloid leukemia with haploidentical NK cells and recombinant human interleukin-15 induced CR in 32% of patients.104 Clinical trials are currently evaluating CAR-NK cells in hematologic (NCT03056339, NCT00995137) and solid (NCT03656705, NCT03383978) malignancies.

Personalized vaccines (vaccinomics):

The accumulation of somatic mutations in cancer can generate cancer-specific neo-epitopes. Autologous T-cells often identify these neo-epitopes as foreign bodies, which makes them ideal cancer vaccine targets. Every cancer has its own unique mutations, but a small number of neo-antigens are shared between cancers. Theoretically, technological advances will soon result in rapid mapping of mutations within a genome, rational selection of vaccine targets such as neo-epitopes, and on-demand production of vaccines tailored to a patient2019s individual tumor. Alternatively, off-the-shelf vaccines for tumors with shared epitopes might also be exploitable.

Several personalized vaccines are currently being evaluated in clinical trials.105,106 For example, investigators used computational prediction of neo-epitopes to design personalized RNA mutanome vaccines for patients with metastatic melanoma.105 Two of the five patients treated had objective responses to the vaccine alone, while a third patient had a CR to treatment with the vaccine combined with PD-1 blockade.105 In another study of vaccine-induced polyfunctional CD4+ and CD8+ T-cells targeting unique neoantigens in patients with melanoma106, four of six vaccinated patients had no recurrence at 25 months after vaccination.106

Sipuleucel-T, the first FDA-approved therapeutic cancer vaccine, is produced via ex vivo activation of autologous peripheral-blood mononuclear cells by a recombinant fusion protein comprised of prostatic acid phosphatase and granulocyte–macrophage colony-stimulating factor.107 Sipuleucel-T is used to treat metastatic castration-resistant prostate cancer on the basis of results of a randomized, double-blind, placebo-controlled phase III trial in which patients who received Sipuleucel-T had longer survival than those who received placebo (25.8 months vs. 21.7 months, respectively; p=0.03).107

Challenges and solutions for the optimal implementation of precision medicine

Genomic studies have unveiled the reality of tumors—they are tremendously heterogeneic and complex, and optimized therapy often does not result from classical clinical research and practice models.

Precision medicine studies (Tables 1 and ​2) demonstrate the major challenges in designing trials for this new paradigm. First, the rate of matching patients to drugs in these trials ranges from 5% to 49% and is mostly in the 15% to 20% range. Failure to match patients is attributed to (i) enrollment of individuals with end-stage disease, who deteriorate or die early; (ii) use of small gene panels that yield limited actionable alterations; (iii) delays in receiving and interpreting genomic results; and (iv) difficulty accessing targeted therapy drugs and/or limited drug availability. Some solutions provided by trials with higher matching rates, e.g., I-PREDICT12 (matching rate, 49%), include: (i) use of clinical trial navigators and medication acquisition specialists; (ii) application of a large NGS panel with >200 genes; (iii) creation of just-in-time electronic molecular tumor boards immediately upon physician request; and (iv) exploitation of biomarkers to match patients to chemotherapy, hormonal therapy, and immunotherapy (in addition to gene-targeted agents). The majority of these trials2,3,12,24 have shown improvement in clinical outcomes when treatments are matched to drugs compared to when they are not. Importantly, malignancies have complicated molecular biology, and use of personalized combinations of drugs that address a higher percentage of the aberrations present in an individual cancer is associated with better outcomes than more limited matching.6,7,12,13

Other major hurdles encountered in the implementation of precision medicine include the following: (i) Potential differences in response to matched therapy depending on histology and/or genomic co-alterations. In contrast to molecular abnormalities that predict tumor agnostic response to treatment (e.g., NTRK fusions, MSI-H)27,72,74, selected genomic biomarkers are predictive in specific tumor histologies.108,109 (ii) The heterogeneity, complexity, and constant evolution of genomic landscapes. Due to significant heterogeneity between primary tumor and metastatic sites, molecular profiling of tumor tissue obtained from a single lesion may not always be representative of the systemic disease.110,111 Additionally, under the pressure of targeted treatments, tumor molecular profile constantly evolves, with emerging resistant clones and new molecular alterations driving disease progression.112,113 (iii) The need to screen large numbers of patients in order to find specific/rare genomic defects (for instance, NTRK fusions).27,108,109 (iv) Incomplete biologic/molecular profiles with which to select therapy; suboptimal technology and resources to understand completely the drivers of cancer in individual patients; (v) Considerable delays in the activation of clinical trials; (vi) differences in the metabolism and adverse effects of study drugs in various ethnic groups; (vii) lack of agreement between assays from different diagnostic companies/laboratories; and (viii) most importantly, lack of access to drugs for patients with limited resources as well as excessive eligibility criteria that rule out large swaths of patients with real-world co-morbidities. Approximately 3–5% of patients with cancer are enrolled on clinical trials and accrual is limited by overly restrictive eligibility criteria and limited access to drugs.114 ASCO, the Friends of Cancer Research, and the FDA recommended to broaden eligibility criteria to allow more patients to participate in clinical trials and gain benefit from novel investigational therapies;115 and consequently participants will be representative of the actual patient population, increasing generalizability of the results. Patient enrollment could be enhanced by national and worldwide collaborations, as shown in multi-institutional trials.116,117 Finally, the Clinical Trials Transformation Initiative (CTTI), has been developed to examine the challenges and propose solutions to improve trial recruitment.118

Several initiatives might help overcome the challenges introduced by our emerging understanding of cancer biology: (i) molecular profiling (tissue, blood) should be used at the time of diagnosis and during the course of the disease, the latter to monitor response and resistance; (ii) completion of molecular profiling should be expedited; and (iii) bioinformatic analysis should be optimized to include the key drivers of carcinogenesis.

With the current excitement about the promise of immunotherapy, a large proportion of patients are assigned to immunotherapy trials without undergoing molecular profiling or immune marker identification. Although a significant minority of these patients will experience a clinical benefit and prolonged survival, the majority will have disease progression and/or significant adverse events. Therefore, the incorporation of biomarkers into the selection of patients for immunotherapy needs to be optimized.

Finally, the immense potential of real-world data needs to be addressed. Validation of database information can be performed by comparing outcomes of clinical trials that led to approval with those in the database; if outcomes are similar, real-world data can then be used to rapidly predict new applications for medicines.

Conclusions and future perspectives

Remarkable biotechnological advances are transforming cancer care. Tumor and cell-free DNA profiling using NGS, as well as proteomic and RNA analysis, and a better understanding of immune mechanisms are optimizing cancer treatment selection. A major challenge in the therapeutic management of patients with advanced metastatic cancer is the complexity of tumor biology. This complexity is attributed to highly variable patterns of genetic and epigenetic diversity and clonal architecture associated with spatial expansion, proliferative self-renewal, migration, and invasion. The complexity is amplified by the dynamic, Darwinian evolutionary character of cancer cells, which undergo sequential searches for mechanisms to escape environmental constraints. Such cellular evolution involves the interplay of advantageous “driver” lesions, neutral or “passenger/hitchhiker” abnormalities, molecular changes in the tumor cells that increase the rate of other genomic anomalies, and modifications to the microenvironment and immune machinery that alter the fitness effects of other variables.119 Strategies to address tumor complexity include targeting self-renewing cancer stem cells to overcome their plasticity and adaptability, impacting the microenvironment, and turning cancer into a chronic disease (using cytostatic drugs to suppress cell division and new mutations). The complicated nature of tumor biology is also the result of interactions between the tumor, host, and local ecosystem, including HLA type, genetic polymorphisms, microbiome, immune cell repertoire, and tumor microenvironment.120 New strategies, some of which now have a proven track record, include gene-directed therapies and a host of immune-targeted approaches (e.g., checkpoint blockade, CAR T-cells, personalized vaccinomics).120,121

An overarching theme is that optimized therapy may require the utilization of combinations of drugs and/or strategies that attack the tumor from multiple angles. It is time to recognize the possibility that advanced computer implementation could generate real-world data that expand our understanding of cancer, rapidly identify new treatments, and create personalized drugs or immune therapies.

Highlights

Funding

NIH/NCI, award number P30 CA016672

Footnotes

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

Competing interests

Dr. Apostolia-Maria Tsimberidou has the following financial relationships to disclose: Research Funding (Institution): Immatics, Parker Institute for Cancer Immunotherapy, Tempus, OBI Pharma, EMD Serono, Baxalta, ONYX, Bayer, Boston Biomedical, Placon Therapeutics, Karus Therapeutics, and Tvardi Therapeutics. Consulting or Advisory Role: Covance, Genentech, and Tempus.

Dr. Elena Fountzilas has the following financial relationships to disclose: Travel grant from Merck and K.A.M Oncology/Hematology; stock ownership Deciphera Pharmaceuticals, Inc.

Dr. Mina Nikanjam has the following financial relationships to disclose: Research Funding (Institution): Regeneron, Bristol Myers Squib, Immunocore, Idera, and Merck.

Dr. Razelle Kurzrock has the following financial relationships to disclose: Research Funding (Institution): Incyte, Genentech, Merck Serono, Pfizer, Sequenom, Foundation Medicine, Konica Minolta, Grifols, Biologic Dynamics, and Guardant. Consulting role: X-Biotech, Loxo, and Actuate Therapeutics. Speaker fees: Roche. Ownership interest: IDbyDNA and Curematch, Inc.

References

1. Von Hoff DD, Stephenson JJ Jr., Rosen P, et al. Pilot study using molecular profiling of patients’ tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol2010;28:4877–83. [PubMed] [Google Scholar]

2. Tsimberidou AM, Iskander NG, Hong DS, et al. Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative. Clin Cancer Res2012;18:6373–83. [PMC free article] [PubMed] [Google Scholar]

3. Tsimberidou AM, Wen S, Hong DS, et al. Personalized medicine for patients with advanced cancer in the phase I program at MD Anderson: validation and landmark analyses. Clin Cancer Res2014;20:4827–36. [PMC free article] [PubMed] [Google Scholar]

4. Tsimberidou AM, Hong DS, Ye Y, et al. Initiative for Molecular Profiling and Advanced Cancer Therapy (IMPACT): An MD Anderson Precision Medicine Study. JCO Precis Oncol2017;2017. [PMC free article] [PubMed] [Google Scholar]

5. Le Tourneau C, Delord JP, Goncalves A, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol2015;16:1324–34. [PubMed] [Google Scholar]

6. Schwaederle M, Parker BA, Schwab RB, et al. Precision Oncology: The UC San Diego Moores Cancer Center PREDICT Experience. Mol Cancer Ther2016;15:743–52. [PubMed] [Google Scholar]

7. Wheler JJ, Janku F, Naing A, et al. Cancer Therapy Directed by Comprehensive Genomic Profiling: A Single Center Study. Cancer Res2016;76:3690–701. [PubMed] [Google Scholar]

8. Stockley TL, Oza AM, Berman HK, et al. Molecular profiling of advanced solid tumors and patient outcomes with genotype-matched clinical trials: the Princess Margaret IMPACT/COMPACT trial. Genome Med2016;8:109. [PMC free article] [PubMed] [Google Scholar]

9. Massard C, Michiels S, Ferte C, et al. High-Throughput Genomics and Clinical Outcome in Hard-to-Treat Advanced Cancers: Results of the MOSCATO 01 Trial. Cancer Discov2017;7:586–95. [PubMed] [Google Scholar]

10. Hainsworth JD, Meric-Bernstam F, Swanton C, et al. Targeted Therapy for Advanced Solid Tumors on the Basis of Molecular Profiles: Results From MyPathway, an Open-Label, Phase IIa Multiple Basket Study. Journal of Clinical Oncology2018;36:536–42. [PubMed] [Google Scholar]

11. Tredan O, Wang Q, Pissaloux D, et al. Molecular screening program to select molecular-based recommended therapies for metastatic cancer patients: analysis from the ProfiLER trial. Ann Oncol2019;30:757–65. [PubMed] [Google Scholar]

12. Sicklick JK, Kato S, Okamura R, et al. Molecular profiling of cancer patients enables personalized combination therapy: the I-PREDICT study. Nat Med2019;25:744–50. [PMC free article] [PubMed] [Google Scholar]

13. Rodon J, Soria JC, Berger R, et al. Genomic and transcriptomic profiling expands precision cancer medicine: the WINTHER trial. Nat Med2019;25:751–8. [PMC free article] [PubMed] [Google Scholar]

14. Kim ES, Herbst RS, Wistuba II, et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov2011;1:44–53. [PMC free article] [PubMed] [Google Scholar]

15. Kris MG, Johnson BE, Berry LD, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA2014;311:1998–2006. [PMC free article] [PubMed] [Google Scholar]

16. Aisner DL, Sholl LM, Berry LD, et al. The Impact of Smoking and TP53 Mutations in Lung Adenocarcinoma Patients with Targetable Mutations-The Lung Cancer Mutation Consortium (LCMC2). Clin Cancer Res2018;24:1038–47. [PMC free article] [PubMed] [Google Scholar]

17. Papadimitrakopoulou V, Lee JJ, Wistuba II, et al. The BATTLE-2 Study: A Biomarker-Integrated Targeted Therapy Study in Previously Treated Patients With Advanced Non-Small-Cell Lung Cancer. J Clin Oncol2016;34:3638–47. [PMC free article] [PubMed] [Google Scholar]

18. Esserman LJ, Berry DA, DeMichele A, et al. Pathologic complete response predicts recurrence-free survival more effectively by cancer subset: results from the I-SPY 1 TRIAL--CALGB 150007/150012, ACRIN 6657. J Clin Oncol2012;30:3242–9. [PMC free article] [PubMed] [Google Scholar]

19. Andre F, Bachelot T, Commo F, et al. Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER). Lancet Oncol2014;15:267–74. [PubMed] [Google Scholar]

20. Park JW, Liu MC, Yee D, et al. Adaptive Randomization of Neratinib in Early Breast Cancer. N Engl J Med2016;375:11–22. [PMC free article] [PubMed] [Google Scholar]

21. Rugo HS, Olopade OI, DeMichele A, et al. Adaptive Randomization of Veliparib-Carboplatin Treatment in Breast Cancer. N Engl J Med2016;375:23–34. [PMC free article] [PubMed] [Google Scholar]

22. Wheler J, Lee JJ, Kurzrock R. Unique molecular landscapes in cancer: implications for individualized, curated drug combinations. Cancer Res2014;74:7181–4. [PMC free article] [PubMed] [Google Scholar]

23. Kurzrock R, Giles FJ. Precision oncology for patients with advanced cancer: the challenges of malignant snowflakes. Cell Cycle2015;14:2219–21. [PMC free article] [PubMed] [Google Scholar]

24. Schwaederle M, Zhao M, Lee JJ, et al. Association of Biomarker-Based Treatment Strategies With Response Rates and Progression-Free Survival in Refractory Malignant Neoplasms: A Meta-analysis. JAMA Oncol2016;2:1452–9. [PubMed] [Google Scholar]

25. Rosario SR, Long MD, Affronti HC, Rowsam AM, Eng KH, Smiraglia DJ. Pan-cancer analysis of transcriptional metabolic dysregulation using The Cancer Genome Atlas. Nat Commun2018;9:5330. [PMC free article] [PubMed] [Google Scholar]

26. Long GV, Stroyakovskiy D, Gogas H, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med2014;371:1877–88. [PubMed] [Google Scholar]

27. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N Engl J Med2018;378:731–9. [PMC free article] [PubMed] [Google Scholar]

28. Goodman AM, Kato S, Bazhenova L, et al. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol Cancer Ther2017;16:2598–608. [PMC free article] [PubMed] [Google Scholar]

29. Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med2015;372:2509–20. [PMC free article] [PubMed] [Google Scholar]

30. Subbiah V, Kurzrock R. The Marriage Between Genomics and Immunotherapy: Mismatch Meets Its Match. Oncologist2019;24:1–3. [PMC free article] [PubMed] [Google Scholar]

31. Kurzrock R, Shtalrid M, Romero P, et al. A novel c-abl protein product in Philadelphia-positive acute lymphoblastic leukaemia. Nature1987;325:631–5. [PubMed] [Google Scholar]

32. Kurzrock R, Gutterman JU, Talpaz M. The molecular genetics of Philadelphia chromosome-positive leukemias. N Engl J Med1988;319:990–8. [PubMed] [Google Scholar]

33. Venter JC, Adams MD, Myers EW, et al. The sequence of the human genome. Science2001;291:1304–51. [PubMed] [Google Scholar]

34. Herbst RS, Gandara DR, Hirsch FR, et al. Lung Master Protocol (Lung-MAP)-A Biomarker-Driven Protocol for Accelerating Development of Therapies for Squamous Cell Lung Cancer: SWOG S1400. Clin Cancer Res2015;21:1514–24. [PMC free article] [PubMed] [Google Scholar]

35. Pabla S, Conroy JM, Nesline MK, et al. Proliferative potential and resistance to immune checkpoint blockade in lung cancer patients. J Immunother Cancer2019;7:27. [PMC free article] [PubMed] [Google Scholar]

36. Jennings LJ, Arcila ME, Corless C, et al. Guidelines for Validation of Next-Generation Sequencing-Based Oncology Panels: A Joint Consensus Recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn2017;19:341–65. [PMC free article] [PubMed] [Google Scholar]

37. Li MM, Datto M, Duncavage EJ, et al. Standards and Guidelines for the Interpretation and Reporting of Sequence Variants in Cancer: A Joint Consensus Recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists. J Mol Diagn2017;19:4–23. [PMC free article] [PubMed] [Google Scholar]

39. Salami SS, Singhal U, Spratt DE, et al. Circulating Tumor Cells as a Predictor of Treatment Response in Clinically Localized Prostate Cancer. JCO Precision Oncology2019:1–9. [PMC free article] [PubMed] [Google Scholar]

40. Abd Elmageed ZY, Yang Y, Thomas R, et al. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem Cells2014;32:983–97. [PMC free article] [PubMed] [Google Scholar]

41. Chabon JJ, Simmons AD, Lovejoy AF, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun2016;7:11815. [PMC free article] [PubMed] [Google Scholar]

42. Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol2018;15:81–94. [PubMed] [Google Scholar]

43. Merker JD, Oxnard GR, Compton C, et al. Circulating Tumor DNA Analysis in Patients With Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. Arch Pathol Lab Med2018. [PubMed] [Google Scholar]

44. Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med2004;351:781–91. [PubMed] [Google Scholar]

45. Hiltermann TJ, Pore MM, van den Berg A, et al. Circulating tumor cells in small-cell lung cancer: a predictive and prognostic factor. Ann Oncol2012;23:2937–42. [PubMed] [Google Scholar]

46. Hofman V, Ilie MI, Long E, et al. Detection of circulating tumor cells as a prognostic factor in patients undergoing radical surgery for non-small-cell lung carcinoma: comparison of the efficacy of the CellSearch Assay and the isolation by size of epithelial tumor cell method. Int J Cancer2011;129:1651–60. [PubMed] [Google Scholar]

47. Tamminga M, de Wit S, Hiltermann TJN, et al. Circulating tumor cells in advanced non-small cell lung cancer patients are associated with worse tumor response to checkpoint inhibitors. J Immunother Cancer2019;7:173. [PMC free article] [PubMed] [Google Scholar]

49. Heller G, McCormack R, Kheoh T, et al. Circulating Tumor Cell Number as a Response Measure of Prolonged Survival for Metastatic Castration-Resistant Prostate Cancer: A Comparison With Prostate-Specific Antigen Across Five Randomized Phase III Clinical Trials. Journal of clinical oncology : official journal of the American Society of Clinical Oncology2018;36:572–80. [PMC free article] [PubMed] [Google Scholar]

50. Sparano JA, Gray RJ, Makower DF, et al. Adjuvant Chemotherapy Guided by a 21-Gene Expression Assay in Breast Cancer. N Engl J Med2018;379:111–21. [PMC free article] [PubMed] [Google Scholar]

51. Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med2004;351:2817–26. [PubMed] [Google Scholar]

52. Buffa FM, Camps C, Winchester L, et al. microRNA-associated progression pathways and potential therapeutic targets identified by integrated mRNA and microRNA expression profiling in breast cancer. Cancer research2011;71:5635–45. [PubMed] [Google Scholar]

53. Jacobsen A, Silber J, Harinath G, Huse JT, Schultz N, Sander C. Analysis of microRNA-target interactions across diverse cancer types. Nat Struct Mol Biol2013;20:1325–32. [PMC free article] [PubMed] [Google Scholar]

54. Michuda J, Igartua C, Taxter T, Bell JS, Pelossof R, White K. Transcriptome-based cancer type prediction for tumors of unknown origin. Journal of Clinical Oncology2019;37:3081–. [Google Scholar]

55. Bridgewater J, van Laar R, Floore A, Van’T Veer L. Gene expression profiling may improve diagnosis in patients with carcinoma of unknown primary. British Journal of Cancer2008;98:1425–30. [PMC free article] [PubMed] [Google Scholar]

56. Tothill RW, Shi F, Paiman L, et al. Development and validation of a gene expression tumour classifier for cancer of unknown primary. Pathology2015;47:7–12. [PubMed] [Google Scholar]

57. Weidenbusch B, Richter GHS, Kesper MS, et al. Transcriptome based individualized therapy of refractory pediatric sarcomas: feasibility, tolerability and efficacy. Oncotarget2018;9:20747–60. [PMC free article] [PubMed] [Google Scholar]

58. Worst BC, van Tilburg CM, Balasubramanian GP, et al. Next-generation personalised medicine for high-risk paediatric cancer patients - The INFORM pilot study. Eur J Cancer2016;65:91–101. [PubMed] [Google Scholar]

59. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science2015;348:124–8. [PMC free article] [PubMed] [Google Scholar]

60. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science2015;348:62–8. [PMC free article] [PubMed] [Google Scholar]

61. Jhaveri KL, Wang XV, Makker V, et al. Ado-trastuzumab emtansine (T-DM1) in patients with HER2-amplified tumors excluding breast and gastric/gastroesophageal junction (GEJ) adenocarcinomas: results from the NCI-MATCH trial (EAY131) subprotocol Q. Ann Oncol2019;30:1821–30. [PMC free article] [PubMed] [Google Scholar]

62. Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N Engl J Med2018;378:2093–104. [PMC free article] [PubMed] [Google Scholar]

63. Miao D, Margolis CA, Gao W, et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science2018;359:801–6. [PMC free article] [PubMed] [Google Scholar]

64. Otto GPBRM1 loss promotes tumour response to immunotherapy. Nature Reviews Clinical Oncology2018;15:134–5. [PubMed] [Google Scholar]

65. Goodman AM, Piccioni D, Kato S, et al. Prevalence of PDL1 Amplification and Preliminary Response to Immune Checkpoint Blockade in Solid Tumors. JAMA Oncol2018;4:1237–44. [PMC free article] [PubMed] [Google Scholar]

66. Goodman AM, Kato S, Bazhenova L, et al. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Molecular cancer therapeutics2017;16:2598–608. [PMC free article] [PubMed] [Google Scholar]

67. Langer C, Gadgeel S, Borghaei H, et al. OA04.05 KEYNOTE-021: TMB and Outcomes for Carboplatin and Pemetrexed With or Without Pembrolizumab for Nonsquamous NSCLC. Journal of Thoracic Oncology2019;14:S216. [Google Scholar]

68. Garassino M, Rodriguez-Abreu D, Gadgeel S, et al. OA04.06 Evaluation of TMB in KEYNOTE-189: Pembrolizumab Plus Chemotherapy vs Placebo Plus Chemotherapy for Nonsquamous NSCLC. Journal of Thoracic Oncology2019;14:S216–S7. [Google Scholar]

69. Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA Approval Summary: Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors. Clin Cancer Res2019;25:3753–8. [PubMed] [Google Scholar]

70. Kim ST, Cristescu R, Bass AJ, et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nature Medicine2018;24:1449–58. [PubMed] [Google Scholar]

71. Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol2017;18:1182–91. [PMC free article] [PubMed] [Google Scholar]

72. Le DT, Kim TW, Cutsem EV, et al. Phase II Open-Label Study of Pembrolizumab in Treatment-Refractory, Microsatellite Instability–High/Mismatch Repair–Deficient Metastatic Colorectal Cancer: KEYNOTE-164. Journal of Clinical Oncology2020;38:11–9. [PMC free article] [PubMed] [Google Scholar]

73. Overman MJ, Lonardi S, Wong KYM, et al. Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology2018;36:773–9. [PubMed] [Google Scholar]

74. Marabelle A, Le DT, Ascierto PA, et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair–Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. Journal of Clinical Oncology2020;38:1–10. [PMC free article] [PubMed] [Google Scholar]

75. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science2015;348:124–8. [PMC free article] [PubMed] [Google Scholar]

76. Gong J, Wang C, Lee PP, Chu P, Fakih M. Response to PD-1 Blockade in Microsatellite Stable Metastatic Colorectal Cancer Harboring a POLE Mutation. Journal of the National Comprehensive Cancer Network : JNCCN2017;15:142–7. [PubMed] [Google Scholar]

77. van Gool IC, Eggink FA, Freeman-Mills L, et al. POLE Proofreading Mutations Elicit an Antitumor Immune Response in Endometrial Cancer. Clin Cancer Res2015;21:3347–55. [PMC free article] [PubMed] [Google Scholar]

78. Domchek S, Postel-Vinay S, Im S, et al. Annals of Oncology (2019) 30 (suppl_5): v475–v532 101093/annonc/mdz253. [Google Scholar]

79. Konstantinopoulos PA, Waggoner S, Vidal GA, et al. Single-Arm Phases 1 and 2 Trial of Niraparib in Combination With Pembrolizumab in Patients With Recurrent Platinum-Resistant Ovarian Carcinoma. JAMA oncology2019;5:1141–9. [PMC free article] [PubMed] [Google Scholar]

80. Patel SP, Kurzrock R. PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Mol Cancer Ther2015;14:847–56. [PubMed] [Google Scholar]

81. Davis AA, Patel VG. The role of PD-L1 expression as a predictive biomarker: an analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. Journal for ImmunoTherapy of Cancer2019;7:278. [PMC free article] [PubMed] [Google Scholar]

82. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med2016;375:819–29. [PMC free article] [PubMed] [Google Scholar]

83. Kato S, Goodman A, Walavalkar V, Barkauskas DA, Sharabi A, Kurzrock R. Hyperprogressors after Immunotherapy: Analysis of Genomic Alterations Associated with Accelerated Growth Rate. Clin Cancer Res2017;23:4242–50. [PMC free article] [PubMed] [Google Scholar]

84. Peng W, Chen JQ, Liu C, et al. Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov2016;6:202–16. [PMC free article] [PubMed] [Google Scholar]

85. Koyama S, Akbay EA, Li YY, et al. STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvironment. Cancer Res2016;76:999–1008. [PMC free article] [PubMed] [Google Scholar]

86. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature2015;523:231–5. [PubMed] [Google Scholar]

87. Said R, Guibert N, Oxnard GR, Tsimberidou AM. Circulating tumor DNA analysis in the era of precision oncology. Oncotarget 2020;11:188–211. [PMC free article] [PubMed] [Google Scholar]

88. Schumacher TNM. T-cell-receptor gene therapy. Nat Rev Immunol2002;2:512–9. [PubMed] [Google Scholar]

89. Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clinical cancer research : an official journal of the American Association for Cancer Research2011;17:4550–7. [PMC free article] [PubMed] [Google Scholar]

90. Besser MJ, Shapira-Frommer R, Itzhaki O, et al. Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies. Clinical cancer research : an official journal of the American Association for Cancer Research2013;19:4792–800. [PubMed] [Google Scholar]

91. Andersen R, Donia M, Ellebaek E, et al. Long-Lasting Complete Responses in Patients with Metastatic Melanoma after Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes and an Attenuated IL2 Regimen. Clinical cancer research : an official journal of the American Association for Cancer Research2016;22:3734–45. [PubMed] [Google Scholar]

92. Forget M-A, Haymaker C, Hess KR, et al. Prospective Analysis of Adoptive TIL Therapy in Patients with Metastatic Melanoma: Response, Impact of Anti-CTLA4, and Biomarkers to Predict Clinical Outcome. Clinical cancer research : an official journal of the American Association for Cancer Research2018;24:4416–28. [PMC free article] [PubMed] [Google Scholar]

93. Comoli P, Pedrazzoli P, Maccario R, et al. Cell therapy of stage IV nasopharyngeal carcinoma with autologous Epstein-Barr virus-targeted cytotoxic T lymphocytes. Journal of clinical oncology : official journal of the American Society of Clinical Oncology2005;23:8942–9. [PubMed] [Google Scholar]

94. Stevanović S, Draper LM, Langhan MM, et al. Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells. Journal of clinical oncology : official journal of the American Society of Clinical Oncology2015;33:1543–50. [PMC free article] [PubMed] [Google Scholar]

95. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med2014;371:1507–17. [PMC free article] [PubMed] [Google Scholar]

97. Brown CE, Alizadeh D, Starr R, et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N Engl J Med2016;375:2561–9. [PMC free article] [PubMed] [Google Scholar]

98. Govers C, Sebestyén Z, Coccoris M, Willemsen RA, Debets R. T cell receptor gene therapy: strategies for optimizing transgenic TCR pairing. Trends Mol Med2010;16:77–87. [PubMed] [Google Scholar]

99. Johnson LA, Morgan RA, Dudley ME, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood2009;114:535–46. [PMC free article] [PubMed] [Google Scholar]

100. Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science (New York, NY)2006;314:126–9. [PMC free article] [PubMed] [Google Scholar]

101. Chodon T, Comin-Anduix B, Chmielowski B, et al. Adoptive transfer of MART-1 T-cell receptor transgenic lymphocytes and dendritic cell vaccination in patients with metastatic melanoma. Clinical cancer research : an official journal of the American Association for Cancer Research2014;20:2457–65. [PMC free article] [PubMed] [Google Scholar]

102. Kageyama S, Ikeda H, Miyahara Y, et al. Adoptive Transfer of MAGE-A4 T-cell Receptor Gene-Transduced Lymphocytes in Patients with Recurrent Esophageal Cancer. Clinical cancer research : an official journal of the American Association for Cancer Research2015;21:2268–77. [PubMed] [Google Scholar]

103. Parkhurst MR, Yang JC, Langan RC, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther2011;19:620–6. [PMC free article] [PubMed] [Google Scholar]

104. Cooley S, He F, Bachanova V, et al. First-in-human trial of rhIL-15 and haploidentical natural killer cell therapy for advanced acute myeloid leukemia. Blood Adv2019;3:1970–80. [PMC free article] [PubMed] [Google Scholar]

105. Sahin U, Derhovanessian E, Miller M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature2017;547:222–6. [PubMed] [Google Scholar]

106. Ott PA, Hu Z, Keskin DB, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature2017;547:217–21. [PMC free article] [PubMed] [Google Scholar]

107. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med2010;363:411–22. [PubMed] [Google Scholar]

108. Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N Engl J Med2015;373:726–36. [PMC free article] [PubMed] [Google Scholar]

109. Ross JS, Ali SM, Fasan O, et al. ALK Fusions in a Wide Variety of Tumor Types Respond to Anti-ALK Targeted Therapy. Oncologist2017;22:1444–50. [PMC free article] [PubMed] [Google Scholar]

110. Lovly CM, Salama AKS, Salgia R. Tumor Heterogeneity and Therapeutic Resistance. American Society of Clinical Oncology Educational Book2016:e585–e93. [PMC free article] [PubMed] [Google Scholar]

111. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing. New England Journal of Medicine2012;366:883–92. [PMC free article] [PubMed] [Google Scholar]

112. Kobayashi S, Boggon TJ, Dayaram T, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med2005;352:786–92. [PubMed] [Google Scholar]

113. Napolitano A, Vincenzi B. Secondary KIT mutations: the GIST of drug resistance and sensitivity. British Journal of Cancer2019;120:577–8. [PMC free article] [PubMed] [Google Scholar]

114. Murthy VH, Krumholz HM, Gross CP. Participation in cancer clinical trials: race-, sex-, and age-based disparities. Jama2004;291:2720–6. [PubMed] [Google Scholar]

115. Kim ES, Bruinooge SS, Roberts S, et al. Broadening Eligibility Criteria to Make Clinical Trials More Representative: American Society of Clinical Oncology and Friends of Cancer Research Joint Research Statement. J Clin Oncol2017;35:3737–44. [PMC free article] [PubMed] [Google Scholar]

116. Unger JM, Cook E, Tai E, Bleyer A. The Role of Clinical Trial Participation in Cancer Research: Barriers, Evidence, and Strategies. Am Soc Clin Oncol Educ Book2016;35:185–98. [PMC free article] [PubMed] [Google Scholar]

117. Trimble EL, Abrams JS, Meyer RM, et al. Improving cancer outcomes through international collaboration in academic cancer treatment trials. J Clin Oncol2009;27:5109–14. [PMC free article] [PubMed] [Google Scholar]

118. Huang GD, Bull J, Johnston McKee K, Mahon E, Harper B, Roberts JN. Clinical trials recruitment planning: A proposed framework from the Clinical Trials Transformation Initiative. Contemporary clinical trials2018;66:74–9. [PubMed] [Google Scholar]

120. Sahin U, Tureci O. Personalized vaccines for cancer immunotherapy. Science2018;359:1355–60. [PubMed] [Google Scholar]

121. van Rooij N, van Buuren MM, Philips D, et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol2013;31:e439–42. [PMC free article] [PubMed] [Google Scholar]

122. Lee J, Kim ST, Kim K, et al. Tumor Genomic Profiling Guides Patients with Metastatic Gastric Cancer to Targeted Treatment: The VIKTORY Umbrella Trial. Cancer Discov2019;9:1388–405. [PubMed] [Google Scholar]

123. Folprecht G, Aust DE, Roth A, et al. Improving access to molecularly defined clinical trials for patients with colorectal cancer: The EORTC SPECTAcolor platform. Journal of Clinical Oncology2015;33:575–575. [Google Scholar]

124. Gerber DE, Oxnard GR, Mandrekar SJ, et al. ALCHEMIST: a clinical trial platform to bring genomic discovery and molecularly targeted therapies to early-stage lung cancer. Journal of Clinical Oncology2015;33:TPS7583–TPS. [PMC free article] [PubMed] [Google Scholar]

125. Aftimos PG, Antunes De Melo e Oliveira AM, Hilbers F, et al. 152OFirst report of AURORA, the breast international group (BIG) molecular screening initiative for metastatic breast cancer (MBC) patients (pts). Annals of Oncology2019;30. [Google Scholar]

126. Slosberg ED, Kang BP, Peguero J, et al. Signature program: a platform of basket trials. Oncotarget2018;9:21383–95. [PMC free article] [PubMed] [Google Scholar]

127. Joshi SS, Maron SB, Lomnicki S, et al. Personalized antibodies for gastroesophageal adenocarcinoma (PANGEA): A phase II precision medicine trial (NCT02213289). Journal of Clinical Oncology2018;36:TPS198–TPS. [Google Scholar]

128. Krop IE, Jegede O, Grilley-Olson JE, et al. Results from molecular analysis for therapy choice (MATCH) arm I: Taselisib for PIK3CA-mutated tumors. Journal of Clinical Oncology2018;36:101–101. [Google Scholar]

129. Jhaveri KL, Wang XV, Makker V, et al. Ado-trastuzumab emtansine in patients with HER2-amplified tumors excluding breast and gastric/gastroesophageal junction adenocarcinomas: results from the NCI-MATCH trial (EAY131) subprotocol Q. Annals of Oncology2019. November 1;30(11):1821–1830 [PMC free article] [PubMed] [Google Scholar]

130. Chae YK, Vaklavas C, Cheng HH, et al. Molecular analysis for therapy choice (MATCH) arm W: Phase II study of AZD4547 in patients with tumors with aberrations in the FGFR pathway. Journal of Clinical Oncology2018;36:2503–2503. [Google Scholar]

131. Azad N, Overman M, Gray R, et al. Nivolumab Is Effective in Mismatch Repair-Deficient Noncolorectal Cancers: Results From Arm Z1D-A Subprotocol of the NCI-MATCH (EAY131) Study. Journal of Clinical Oncology2020. January 20;38(3):214–222. [PMC free article] [PubMed] [Google Scholar]

132. Mangat PK, Halabi S, Bruinooge SS, et al. Rationale and Design of the Targeted Agent and Profiling Utilization Registry (TAPUR) Study. JCO Precis Oncol2018;2018. [PMC free article] [PubMed] [Google Scholar]