PIIS147149991417301430
PIIS147149991417301430
PIIS147149991417301430
874 Trends in Molecular Medicine, October 2017, Vol. 23, No. 10 http://dx.doi.org/10.1016/j.molmed.2017.08.003
© 2017 Elsevier Ltd. All rights reserved.
genetic-based evidence for cancer occurrence and development. Improved technologies Sackler School of Medicine, Tel Aviv
University, Tel Aviv, 69978, Israel
enabling the detection of such mutations in non-neoplastic tissues (including bodily fluids) 4
Center for Bioinformatics and
has allowed the early detection of somatic oncogenic mutations, such as Ras mutations and Computational Biology, University of
hotspot p53 tumor suppressor mutations [6–8]. While these developments reflect advances Maryland, College Park, MD 20742,
USA
made already during the 1980s, it has taken another generation to better establish the 5
Technion Integrated Cancer Center,
importance of mutation frequency, its variability in the transformed tissue, and its causative Faculty of Medicine, Technion, Israel
role. This growing understanding has been a prerequisite for the introduction of mechanism- Institute of Technology, Haifa, 31096,
Israel
based therapies into clinical practice. Commonly known as targeted therapies, these 6
Center for Bioinformatics and
therapeutic approaches are based on small molecules or monoclonal antibodies that inhibit Computational Biology, University of
oncogenic drivers [9–14], or target genetic vulnerabilities [e.g., poly (ADP-ribose) polymerase Maryland, College Park, MD 20742,
USA
(PARP) inhibitors in tumors with homologous recombination deficiency [15]]. Several years
of clinical experience with targeted agents, and especially of the resistance to drugs, has led to
the recognition of the central role of genetic heterogeneity and plasticity of growth-promoting *Correspondence:
signaling pathways in determining a patient’s individual response. A notable example is the [email protected] (Z.A. Ronai).
targeting of BRAF mutations, which are present in more than 40% of melanomas [16]. Although
targeting recurrent BRAF mutation(s) by mutant-specific BRAF inhibitors demonstrated great
clinical success [9,17], understanding the complex feedback and crosstalk between key
players of the altered RAS/RAF/MEK/ERK signaling axis became necessary for optimizing
therapy. Accordingly, in terms of clinical outcomes, combined BRAF and MEK inhibition proved
superior over single-agent use [18]. Furthermore, new generations of specific BRAF inhibitors
are currently in the pipeline, finely tuned to overcome mutation-driven altered signaling events in
the RAS/RAF/MEK/ERK pathway [18]; these might be expected to outperform previous
inhibitors of this pathway. Similar undertakings may be required to target deregulated signaling
pathways arising from other mutations in different tumors, where a driver mutation is known,
and where drugs targeting a given driver may exist.
The search for cancer vulnerabilities in specific cancer types has been facilitated by numerous
technological advances yielding large-scale molecular profiling of major cancer types [23,24].
This system-based analysis of tumor samples, together with massive hypothesis-based
research, has significantly changed our understanding of cancer biology (Figure 1, Key Figure):
carcinogenesis is generally considered to be driven by the natural selection of continuously
acquired genetic and epigenetic variation in individual cells [25]. These converge on common
phenotypic characteristics for cancer cells, including sustained proliferation, migration, inva-
sion, and/or resistance to apoptosis [26]. Tissue microenvironments provide the fitness
selection defining spatial and temporal changes in environmental pressures. These influence
the evolutionary path of any given cancer cell, resulting in (epi-)genetically heterogeneous
subpopulations. Diversity within cancer cell populations is not limited to the genome, and
dynamic variations in differentiation hierarchies, transcriptional signals, and the proteomic
landscape add to the phenotypic heterogeneity observed within tumors [27]. Indeed, cancer
cells do not exist as isolated entities, but rather, engage in heterotypic interactions with stromal
cells and cooperate with adjacent tumor subclones; this is important, because it can result in
the increased robustness of a tumor [28].
The following sections describe related computational approaches; for an expanded summary
of references on these and additional topics (ITH and single cell analysis approaches) the reader
is referred to Tables S1–S3 in the supplemental information online.
IMPACT; M.D. Anderson All; advanced, Hotspot sequencing 1144 175 Mono and Higher ORR and longer
[193]; NCT00851032; refractory 11 genes; FISH combination; Phase PFS compared with
retrospective, (ALK) I therapies; N.S. unmatched therapy
nonrandomized
IMPACT/COMPACT [48]; Advanced MALDI-TOF MS 1640 245 N.S.; investigational Genotyped matched
NCT01505400; cancers and hotspot panel (23 agents of 277 trials, therapy improved
retrospective, Phase I genes) or targeted including 89 response
observational, candidates NGS panel (48 or 50 genotype-matched
nonrandomized genes) trials
Bisgrove [195]; All; advanced, IHC, FISH, gene 86 68 Mono and 27% with extended PFS
NCT00530192; refractory expression combination; N.S. of 30% compared with
prospective, single-arm previous therapy
Phase I
Genomic Profiling in Phase All; advanced, Panel NGS (236 339 122 Mono and High matching score
I [46]; NCT02437617; refractory genes); standard combination; Phase associated with higher
prospective, biomarker I/II therapies; N.S. SD 6 months/PR/CR,
nonrandomized longer PFS and survival
MOSCATO 01 [44]; All; advanced, CGH array; panel 843 199 Phase I drugs and 33% with extended PFS
NCT01566019 refractory NGS (WES and off-label drugs; N.S. of 30% compared with
prospective, RNAseq in 2014 previous therapy
nonrandomized included)
WinTher; NCT01856296; All; advanced, DNA (236 genes) To be 200 N.A. N.S. Chosen based Estimated completion in
prospective, refractory and RNA in tumor on DNA analysis, if 2018
nonrandomized and normal no actionable
matched tissue mutation, then
based on RNA
analysis
SHIVA [45]; NCT01771458 All; advanced, Panel hotspot NGS 741 99 Erlotinib, lapatinib Median PFS in
prospective, randomized refractory (46 genes); CNV + trastuzumab, experimental group
sorafenib, imatinib, (matched therapy) not
dasatinib, significantly longer than
vemurafenib, in control group
everolimus
NCI-MPACT; All, advanced NGS (4000 variants To be 700 N.A. Carboplatin, Estimated completion in
NCT01827384; solid tumors across 143 genes); everolimus, 2019; outcome
prospective, Phase II activation of RAS/ temozolomide, measures: ORR;
randomized feasibility RAF or PI3K trametinib, veliparib, compare genotype
study pathway; AZD1775 matched versus
inactivation of DNA physicians choice
repair pathway
a
Abbreviations: AZD1775, WEE1 inhibitor; CGH, comparative genomic hybridization; CR, complete response; FISH, fluorescent in situ hybridization; IHC, immu-
nohistochemistry; N.A., not applicable; N.S., not specified; NGS, next-generation sequencing; ORR, overall response rate; PFS, progression-free survival; PR, partial
response; Pts, patients; SD, stable disease; WES, whole-exome sequencing.
deep neural nets in areas of computer vision, natural language processing, and biology [84].
Initial molecular subtype studies have often focused on clustering samples into subtypes based
on gene expression in a single cancer type, which have provided robust biomarkers and
subgroups, coherent with patient survival profiles (e.g, in breast cancer [85] or CRC [86]). These
studies typically reveal a more refined set of clusters than those defined by known
BATTLE; Chemorefractory Non-NGS, mutation Mutation/amplification: Erlotinib, sorafenib, Better DCR for EGFR
NCT00409968, NSCLC analysis, FISH, IHC EGFR, KRAS/BRAF, vandetanib, erlotinib + erlotinib and KRAS/
NCT00411671, CCND1; protein + bexarotene BRAF +sorafenib
NCT00411632, expression: VEGF, RXR,
NCT00410059, cyclinD1
NCT00410189;
Prospective, adaptively
randomized, umbrella
trial
BATTLE-2; Advanced NSCLC NGS; DNA, mRNA, IHC: pAKT, PTEN, HIF1a, Erlotinib (ctrl) sorafenib, 8-week DCR
NCT01248247; 2- RPPA, IHC LKB1. Mutation: P13KCA, MK-2206 + erlotinib; MK-
stage Phase II umbrella BRAF, AKT1, HRAS, 2206 + selumetinib
design NRAS, MEK1, MET,
CTNNB1, LKB1;
Lung-MAP; Advanced, recurrent FMI FoundationOne PI3KCA, CDK4/6, CCND1/ Taselisib, palbociclib, PFS, ORR, OS
NCT02154490 squamous cell lung platform; IHC; 2/3, FGFR1/2/3, HGF/c- AZD4547, erlotinib,
randomized phase II/III carcinoma MET erlotinib + rilotumumab,
umbrella design ipilimumab, nivolumab,
durvalumab
I-SPY-2; Stage III breast cancer Conventional, MRI HER2, hormone receptor, HER2+: neratinib, MK2206 Pathological complete
NCT01042379; and Mammaprint + trastuzumab, T-DM1 response; several drugs
randomized open-label Bayesian marker-adaptive + pertuzumab, trebananib graduated to Phase III
Phase II umbrella trial designs + trastuzumab, trials
design; (adjuvant pertuzumab [196–198]
setting) + trastuzumab; HER2 :
veliparib, MK2206,
ganitumab + metformin,
trebananib
ALCHEMIST; Resectable NGS, tissue and EGFR mutation; ALK Erlotinib, crizotinib, OS
NCT02194738; nonsquamous NSCLC blood; germline rearrangements (FISH); nivolumab
NCT02193282; + somatic PD-L1 expression (IHC)
NCT02201992; alterations
NCT02595944;
screening study and
accrual to Phase III
randomized treatment
studies (adjuvant
setting)
SAFIR02 Breast; HER2 recurrent and/ CGH array, hotspot To be determined during AZD2014, AZD4547, PFS compared with
NCT02299999; Phase or metastatic breast sequencing study AZD5363, sapitinib, standard maintenance
II randomized umbrella cancer selumetinib, vandetanib,
design bicalutamide, olaparib,
durvalumab
SAFIR02 Lung; EGFR and ALK WT CGH array, hotspot To be determined during AZD2014, AZD4547, PFS compared with
NCT02117167; Phase recurrent and/or sequencing study AZD5363, sapitinib, standard maintenance
II randomized umbrella metastatic NSCLC selumetinib, vandetanib,
design durvalumab
Lung MATRIX; Advanced, pretreated 28-gene NGS Mutation: FGFR2/3, TSC1/ AZD4547, AZD2014, ORR, PFS
NCT02664935 Phase NSCLC platform 2, LKB1, KRAS + RbWT, palbociclib, crizotinib,
II; nonrandomized NF1, NRAS, PIK3CA, selumetinib, AZD5363,
umbrella design AKT1, EGFR + EGFRT790; osimertinib, durvalumab
LoF: p16 + RbWT, PTEN;
amplification: CDK4
+ RbWT, CCDN1 + RbWT,
MET, PIK3CA; rearranged:
ROS1
V-BASKET [49]; Solid tumors, multiple Mutation analysis BRAFV600 Vemurafenib monotherapy; Efficacy in NSCLC,
NCT01524978; flexible, myeloma with local method vemurafenib + cetuximab ECD, and LCH
early Phase II; basket in CRC
study
CUSTOM [47]; NSCLC, SCLC, thymic NGS Mutation: AKT1, BRAF, Erlotinib, selumetinib, Targeting EGFR and
NCT01306045; malignancy EGFR, ERBB2, HRAS, KIT, MK2206, lapatinib, ALK offers benefit;
biomarker-derived, KRAS, NRAS, PDGFRA, sunitinib design not feasible for
multiarm, multihistology PIK3CA, PTEN; most arms
Phase II, basket trial amplification: ERBB2,
PIK3CA, PDGFRA; fusion:
ALK
NCI-MATCH; Advanced, recurrent, NGS (4000 variants Mutations: AKT1, BRAF Ado-trastuzumab ORR
NCT02465060; refractory solid tumors, across 143 genes) V600; BRAF non-V600, emtansine, afatinib,
nonrandomized Phase lymphoma, myeloma BRCA1/2, cKIT, DDR2, AZD4547, AZD5363,
II; basket trial dMMR, EGFR, AZD1775, osimertinib,
EGFRT790 M, FGFR1/2/3, binimetinib, crizotinib,
GNAQ/GNA11, HER2, dabrafenib + trametinib,
MET ex14 sk, mTOR, NF1, dasatinib, defactinib,
NRAS, PI3KCA, PTEN, GSK2636771, larotectinib,
SMO/PTCH1, TSC1/2; nivolumab, palbociclib,
Translocations: ROS1, sunitinib, TAK-228,
ALK; amplification: taselisib, trametinib,
CCDN1/2/3, CDK4/6, trastuzumab, vismodegib
HER2, MET; loss: NF2,
PTEN; fusion: NFRK
a
Abbreviations: AZD4547, FGFR inhibitor; AZD5363, AKT1/2/3 inhibitor; AZD1775, WEE1 inhibitor; AZD2014, inhibitor of mTORC1/2; CGH, comparative genomic
hybridization; DCR, disease control rate; ECD, Erdheim–Chester disease; GSK2636771, inhibitor of PI3K beta; LCH, Langerhans’ cell histiocytosis; MK2206, inhibitor
of AKT1/2/3; NGS, next-generation sequencing; NSCLC, non-small cell lung cancer; ORR, overall response rate; OS, overall survival; PFS, progression-free survival;
RPPA, reverse-phase protein array; SCLC, small cell lung cancer; TAK-228, dual mTORC1/2 inhibitor.
histopathological markers, with more-coherent survival profiles of the samples and/or patients
composing them. While some of the molecular clusters strongly overlap with known histologi-
cally based clusters, others surprisingly comprise samples with distinct histopathological
markers, but with similar transcriptomic profiles and survival rates. More recent analyses have
clustered multiple platforms across multiple cancer types and, as outlined above, identified
molecular similarities between tumors of different tissue of origin. For example, one study
analyzed TCGA data from >3000 samples across 12 cancer types, and found that, while most
cancers could be classified based on their histology, approximately 10% could be classified as
belonging to an ‘integrated’ subtype, that is, including cancers from multiple tissues of origin
in the same subtype [37]. Furthermore, grouping samples from different tissue types yielded
improved predictive power for patient prognosis, potentially reflecting the value of molecular
features (such as common mutations) for predicting survival [37]. The US Food and Drug
Administration (FDA) recently approved the drug pembrolizumab (an immune-checkpoint PD-1
blockade), used across many cancer types, with demonstrated effectiveness in CRC, endo-
metrial, pancreas, thyroid, and eight other cancer types, based on the presence of a specific
(mismatch-repair deficiency) signature [87]. These studies demonstrate the promise of classi-
fying tumors using molecular features, which can give additional insights into prognosis and
treatment beyond tissue of origin.
Despite these advances, in most cancer data sets, there is a ‘long tail’ of genes with infrequent
mutations, where the drivers are statistically indistinguishable from passenger mutations [32].
One report illustrated the depth of this problem by estimating the number of samples required
to detect driver mutations with a given frequency in a given cancer type through saturation
analysis [34]. The cancer type in question was critical because of the high variance in
background mutation rates in different cancers (e.g., breast cancer, prostate cancer, etc.).
For example, the authors showed that up to 5300 samples would need to be sequenced to
detect drivers occurring at 2% above the high background mutation rate in melanoma [34]. This
presents a particular challenge for rare cancer types, especially since cancer types continue to
be divided into different subtypes [34].
Given this promising potential, extensive experimental efforts have aimed to tease out the wiring
of genetic interactions in cancer cells based on single (isogenic) cell lines [116–121] or on large-
scale genetic knockout-based screens [122–126]. However, due to the large combinatorial
space of pairwise interactions that need to be surveyed, these screens have probed only a
small fraction of the coverage offered by SLi: each screen typically scans a few thousand
candidate SL partners of just one ‘anchor’ cancer driver gene of interest (e.g., KRAS or VHL),
altogether covering a mere fraction of the 500 million gene pairings in the human genome. Yet,
with these screens, several SL interactions have been successfully uncovered to date; apart
from examining the effect of PARP inhibitors in patients with BRCA-mutated breast and
Aiming to bypass the limitations of current experimental techniques in probing the vast space of
potential SLi, various computational approaches have been developed to identify such candi-
date SLi (see references in Table S2D in the supplemental information online). These include
applying various machine-learning methodologies to predict genetic interactions in different
species [128–131], and in cancer (using yeast SLi) [119,132], utilizing metabolic modeling
[133,134], evolutionary characteristics [119,129], and transcriptomic profiles [101,135], and,
more recently, by mining cancer patient data [136–138] (Table S2D in the supplemental
information online). One recent study evaluated the TCGA copy number and transcriptomics
data to identify as candidate SLis, gene pairs that are almost never found inactivated in the
same tumors [136]. The study demonstrated that gene pair interactions (a subset of which was
validated in experimental screens) could be successfully used to predict the survival of patients
with breast cancer in an independent data set [136]. The pair was also used to predict in vitro
drug responses to identify novel drug repurposing indications for potentially treating renal
cancer [136]. Unlike the approach of using expression and copy number data [136], an
algorithm was recently developed to mine pan-cancer human tumor data and define muta-
tion-specific SL interactions for specific cancers [139]. Its SL predictions were validated against
published SL screens and one specific SL gene pair interaction between mutated IDH1 and the
gene encoding acetyl-CoA carboxylase 1 (ACACA) in leukemia was experimentally validated;
this interaction attenuating tumor growth in patient-derived xenografts(PDX) [139]. Finally,
certain predicted SL interactions were shown to successfully predict drug sensitivity, thus
serving as biologically interpretable biomarkers of the latter [139]. Overall, while these studies
have laid a solid basis for some of these genome-wide approaches, extensive research is
warranted to further elucidate the potential of SLi-based approaches in precision oncology.
Moreover, for clinical trials, there is an important unmet need to specifically design and test SLi-
based approaches that may uncover a range of tumor-specific vulnerabilities.
The Use of In Vitro and In Vivo Models for Guiding Precision Therapy
The increased functional annotation of genetic variants of unknown significance [140], along
with systematic high-throughput drug screens in 1000 cancer cell lines [141] or 1000 PDX
models [142], has significantly increased our understanding of the relationship between
genotype and drug sensitivity. We are beginning to understand the molecular profiles of
patients responding to conventional chemotherapy, such as to temozolomide [143] and
other DNA-damaging agents [144]. Due to their well-characterized clinical benefit in unstratified
patient cohorts, these remain equally valuable therapy choices in addition to targeted therapies.
With this ever-increasing number of validated biomarkers and available drugs, it is expected
that molecular profiling will reveal multiple potentially actionable alterations, which may be
treated with a multitude of drugs and/or drug combinations. Prioritizing predicted treatments
requires functional testing, especially in cases where the drug–biomarker association has not
been clinically validated. Undoubtedly, there will still be patients whose molecular analysis is
either not feasible or does not reveal targetable alterations, for which alternate routes to inform
therapy are necessary. For this purpose, several in vitro or in vivo patient-derived functional
platforms (e.g., PDX or organoid models) have been developed that mimic the native features of
tumors more closely than conventional cell culture drug-screening platforms [145].
PDX models offer one attractive approach, because tumor heterogeneity is maintained in these
models at least in early passages (for a comprehensive review, see [146]). In addition, clinical
studies have demonstrated remarkable correlations between drug activity in the PDX model
and a patient’s clinical outcome [146–149]. However, not all human tumor samples grow in
mice following subcutaneous or orthotopic implantation, and the long time span needed for
Patient-derived 3D organoids provide a practical alternative (see other models discussed in Box
2). Organoids are established by dissociating and embedding tissue in an cell-free extracellular
matrix (matrigel or collagen), which can be expanded in a growth factor-enriched medium [150].
Organoids from pancreatic [151], colon [152–154], gastric [155], prostate cancer [156], and
brain tumors and/or metastasis [157] have been established, and have the advantage of 3D
growth of normal and cancer tissue, recapitulating copy number and mutation spectra, as well
as other physiologically relevant aspects of disease progression in vitro [150–158]. Organoids
can be established in culture from needle biopsies within a relative short time period, and have
also been generated from circulating tumor cells(CTCs) [156]. Organoids can serve as a
model system to perform high-throughput screens within a clinically relevant time frame: in a
larger precision oncology study, organoids were established from fresh tissue available from
38% of 145 patients [158]. In addition, PDX models were successfully established from these
organoids in 19 of 22 attempts [158]. High-throughput drug screens were performed (160
drugs, including chemotherapy and targeted therapy) in 2D cultures from four patients, and the
best ‘hits’ (drugs that most effectively decreased cell viability in vitro) were verified in 3D
organoid cultures. Selected treatments were then tested in combination to identify effective
combination therapies. The best hits of single and combination therapies from two patients
were further tested in 3D organoids and PDX models, validating tumor responses in vivo, and
compared with the efficacy of current patient treatments [158]. In addition, potential drug
In organotypic slice cultures [216] or organ explants [217], either thin slices of the tumor sample or minced tumor tissues
are maintained in culture. The biggest advantage of these culture types is that they retain cancer associated stromal
cells, preserve tumor–stroma interactions, signaling pathways, and gene expression profiles [218]. Improvements
include the use of autologous serum and patient-specific stromal-matrix proteins to more closely resemble individual
microenvironmental conditions [219], aiming to accurately predict responses to anticancer drugs. However, not all
tissues are suitable to generating thin slices (e.g., soft, mucinous, or fatty tissue), where firm tissue consistency is
required [220,221]. Another drawback of slice cultures is the loss of viability within 5–7 days [218]. Given that the median
time frame for molecular profiling and data processing in precision oncology trials is 2–4 weeks, the method is not
suitable for testing genomics-guided therapies derived from the same biopsy, unless combined with other models.
Recently, organotypic slice cultures established from pancreatic ductal adenocarcinoma PDX models were used to
screen against clinically relevant drug regimens in a 96-well format, demonstrating consistency between sensitivity of
organotypic cultures and the clinical responses of donor patients [222].
Multiple resistance mechanisms (genetic and nongenetic) can act in concert to confer resis-
tance to targeted therapy (Figure 2). Indeed, in a recent melanoma study [186], analysis of
patient-matched melanoma tumors biopsied before therapy and during disease progression
demonstrated that, in contrast to heterogeneous genetic mechanisms that could result in
acquired resistance, transcriptomic signatures were highly recurrent in serial biopsies; these
indicated that a multitude of genetic and epigenetic events within the tumor compartment
converged on specific genes (c-MET, LEF1, and YAP1) and pathways to mediate resistance,
consistent with a canalization evolutionary process [186]. Additionally, this acquired
resistance signature correlated with changes in the tumor immune microenvironment, including
depletion of intratumoral CD8+ T cells, exhaustion of tumor-reactive CD8+ T cells, and loss of
antigen presentation; these have been previously linked to resistance to anti-PD-1 salvage
therapy in biopsies of patients with melanoma, in support of the presumed role of CD8+ T cell
exhaustion in the development of resistance [77,186]. Furthermore, these findings suggest that
The mere follow-up of genomic alterations in ctDNA is unlikely to result in the satisfactory
management of resistant disease because adaptive mechanisms on transcriptional and
signaling levels can be missed by genomic analysis alone (Figure 2). The use of CTCs may
be preferred to monitor resistant disease, because they have a better overall prognostic value,
provide an additional opportunity to characterizing genetic and nongenetic ITH, and are
suitable for functional studies [187]. The previously described in vitro and/or ex vivo models
may serve to detect relevant signaling nodes and counteract adaptive signaling by combina-
torial therapy to delay onset of resistance. As suggested for the selection of first-line therapies, a
comprehensive genomic, transcriptomic, and functional analysis of resistant disease is
required to overcome these challenges. In that case, it may be possible to mine the increasingly
available ‘omics data from large cohorts of patients and identify genetic interactions that
mediate network-wide signaling alterations conferring resistance. Such an approach could
exploit the much less-studied type of genetic interactions, termed synthetic rescues (SRs),
also known as suppression interactions [188–192]. Such SRs denote a functional interac-
tion between two genes where the targeting of one gene is compensated by the altered activity
of another gene (termed the ‘rescuer gene’); this can restore and rescue cell fitness, leading to
drug resistance. Similar to SLs, SR interactions could in principle be identified by mining ‘omics
data from large cohorts of pretreated tumor samples, taking into consideration that functional
alterations might already be occurring during the natural evolution of tumorigenic populations.
At present, ITH appears to be the biggest obstacle we need to overcome to achieve a sustained
therapeutic response. With rapid technological advances, we are acquiring the toolbox to
comprehensively characterize the complex heterogeneity of tumors at the single cell level (Box
4). However, computing the data from different platforms of 1000 cells or more remains a
challenge. Among the questions that emerge is whether it will be possible to develop multiplex-
based approaches, or use machine-learning techniques to compute tumor trends to predict
subcluster and clonal behaviors (see Outstanding Questions).
Among the first series of steps needed to provide important insights into the complex nature of
malignancies, the inclusion of technological advances and computational approaches in clinical
trials stands out. We should aim to map and analyze the impact of complex genetic and
nongenetic heterogeneity during cancer progression and therapy regimens to help pave the
Clinical experience with genomics-guided cancer therapy supports the notion that genomic profiling can improve
patient outcomes. The degree of success can be associated with the ability to verify the role of a targeted mutation and/
or alteration in tumor development, or vice versa; that is, whether the drug can efficiently attenuate the tumorigenic
effects orchestrated by the genetic alteration.
Precision oncology cannot be limited to genetics to predict responses to therapy, neither can it be limited to a single
‘omics-based approach. Multiple drivers can underlie tumor heterogeneity, which in turn can confer resistance,
metastasis, and dormancy. It also requires the targeting of master regulators that are influenced by epigenetic and
microenvironmental-based pathways. The inclusion of additional platforms, of which at this time the most suitable
appears to be transcriptomics, (with future inclusion of metabolome and microbiome analysis), is highly desirable to
identify such master regulators and design more-precise therapeutic modalities.
Advances in computational approaches to mine and integrate the multitude of data sets that become available to us are
expected to allow better subclassification of patient cohorts into subpopulations able to respond to a given therapy. In
addition, integration of multiple data platforms is expected to drive the identification of novel vulnerabilities, which will
further add to the armamentarium of current anticancer therapies. This may likely result in newer stratification methods
to identify patients who might benefit from a given precision oncology approach.
The implementation of powerful ex vivo or improved in vivo PDX models in the planning of clinical practice is encouraged.
By using these models, a multitude of available drugs and predictive biomarkers might be assessed to evaluate
therapeutic options, as well as their combinations and delivery sequence and/or approaches.
Supplemental information associated with this article can be found online at http://dx.doi.org/10.1016/j.molmed.2017.08.
How many single cells (and from how
003.
many tumors) will we need to
sequence to accurately depict the
References complex heterogeneity observed
1. Vargas, A.J. and Harris, C.C. (2016) Biomarker development in 14. Saltz, L.B. et al. (2004) Phase II trial of cetuximab in patients with
within patients? Can machine-learning
the precision medicine era: lung cancer as a case study. Nat. refractory colorectal cancer that expresses the epidermal
Rev. Cancer 16, 525–537 growth factor receptor. J. Clin. Oncol. 22, 1201–1208
approaches be used to predict the
behavior of a tumor?
2. Hyman, D.M. et al. (2017) Implementing genome-driven oncol- 15. Ledermann, J. et al. (2014) Olaparib maintenance therapy in
ogy. Cell 168, 584–599 patients with platinum-sensitive relapsed serous ovarian cancer:
3. Prasad, V. (2016) Perspective: the precision-oncology illusion. a preplanned retrospective analysis of outcomes by BRCA Are CTCs a reliable source to compre-
Nature 537, S63 status in a randomised phase 2 trial. Lancet Oncol. 15, 852–861
hensively map tumor heterogeneity
4. Tannock, I.F. and Hickman, J.A. (2016) Limits to personalized 16. Cancer Genome Atlas, N (2015) Genomic classification of cuta- and do they accurately define pheno-
cancer medicine. N. Engl. J. Med. 375, 1289–1294 neous melanoma. Cell 161, 1681–1696
typic heterogeneity of advanced dis-
5. Voest, E.E. and Bernards, R. (2016) DNA-guided precision 17. Hauschild, A. et al. (2012) Dabrafenib in BRAF-mutated meta-
ease? How can we improve the
medicine for cancer: a case of irrational exuberance? Cancer static melanoma: a multicentre, open-label, phase 3 random-
ised controlled trial. Lancet 380, 358–365 isolation of these valuable cells?
Discov. 6, 130–132
6. Karnoub, A.E. and Weinberg, R.A. (2008) Ras oncogenes: split 18. Johnson, D.B. et al. (2016) Emerging targeted therapies for
personalities. Nat. Rev. Mol. Cell Biol. 9, 517–531 melanoma. Expert Opin. Emerg. Drugs 21, 195–207 How can ITH be exploited
7. Sidransky, D. (1997) Nucleic acid-based methods for the detec- 19. Tang, X.H. and Gudas, L.J. (2011) Retinoids, retinoic acid therapeutically?
tion of cancer. Science 278, 1054–1059 receptors, and cancer. Annu. Rev. Pathol. 6, 345–364
8. Soussi, T. (2010) The history of p53: a perfect example of the 20. Jones, P.A. et al. (2016) Targeting the cancer epigenome for
drawbacks of scientific paradigms. EMBO Rep. 11, 822–826 therapy. Nat. Rev. Genet. 17, 630–641
9. Chapman, P.B. et al. (2011) Improved survival with vemurafenib 21. Ferrara, N. and Kerbel, R.S. (2005) Angiogenesis as a thera-
in melanoma with BRAF V600E mutation. N. Engl. J. Med. 364, peutic target. Nature 438, 967–974
2507–2516 22. Miller, J.F. and Sadelain, M. (2015) The journey from discoveries
10. Cobleigh, M.A. et al. (1999) Multinational study of the efficacy in fundamental immunology to cancer immunotherapy. Cancer
and safety of humanized anti-HER2 monoclonal antibody in Cell 27, 439–449
women who have HER2-overexpressing metastatic breast can- 23. Cancer Genome Atlas Research, N et al. (2013) The Cancer
cer that has progressed after chemotherapy for metastatic Genome Atlas Pan-Cancer analysis project. Nat. Genet. 45,
disease. J. Clin. Oncol. 17, 2639–2648 1113–1120
11. Druker, B.J. et al. (2001) Efficacy and safety of a specific inhibitor 24. International Cancer Genome, C et al. (2010) International net-
of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N. work of cancer genome projects. Nature 464, 993–998
Engl. J. Med. 344, 1031–1037 25. McGranahan, N. and Swanton, C. (2017) Clonal heterogeneity and
12. Kwak, E.L. et al. (2010) Anaplastic lymphoma kinase inhibition in tumor evolution: past, present, and the future. Cell 168, 613–628
non-small-cell lung cancer. N. Engl. J. Med. 363, 1693–1703 26. Hanahan, D. and Weinberg, R.A. (2011) Hallmarks of cancer:
13. Lynch, T.J. et al. (2004) Activating mutations in the epidermal the next generation. Cell 144, 646–674
growth factor receptor underlying responsiveness of non-small- 27. Marusyk, A. et al. (2012) Intra-tumour heterogeneity: a looking
cell lung cancer to gefitinib. N. Engl. J. Med. 350, 2129–2139 glass for cancer? Nat. Rev. Cancer 12, 323–334
31. Futreal, P.A. et al. (2004) A census of human cancer genes. Nat. 55. Hammerman, P.S. et al. (2011) Mutations in the DDR2 kinase
Rev. Cancer 4, 177–183 gene identify a novel therapeutic target in squamous cell lung
cancer. Cancer Discov. 1, 78–89
32. Garraway, L.A. and Lander, E.S. (2013) Lessons from the can-
cer genome. Cell 153, 17–37 56. Iyer, G. et al. (2012) Genome sequencing identifies a basis for
everolimus sensitivity. Science 338, 221
33. Hofree, M. et al. (2013) Network-based stratification of tumor
mutations. Nat. Methods 10, 1108–1115 57. Liao, R.G. et al. (2013) Inhibitor-sensitive FGFR2 and FGFR3
mutations in lung squamous cell carcinoma. Cancer Res. 73,
34. Lawrence, M.S. et al. (2014) Discovery and saturation analysis of
5195–5205
cancer genes across 21 tumour types. Nature 505, 495–501
58. Serra, V. et al. (2013) Clinical response to a lapatinib-based
35. Leiserson, M.D. et al. (2015) Pan-cancer network analysis iden-
therapy for a Li-Fraumeni syndrome patient with a novel
tifies combinations of rare somatic mutations across pathways
HER2V659E mutation. Cancer Discov. 3, 1238–1244
and protein complexes. Nat. Genet. 47, 106–114
59. Subbiah, V. et al. (2014) Targeted therapy by combined inhibi-
36. Vogelstein, B. et al. (2013) Cancer genome landscapes. Science
tion of the RAF and mTOR kinases in malignant spindle cell
339, 1546–1558
neoplasm harboring the KIAA1549-BRAF fusion protein. J.
37. Hoadley, K.A. et al. (2014) Multiplatform analysis of 12 cancer Hematol. Oncol. 7, 8
types reveals molecular classification within and across tissues
60. Van Allen, E.M. et al. (2015) Genomic correlate of exceptional
of origin. Cell 158, 929–944
erlotinib response in head and neck squamous cell carcinoma.
38. Hartmaier, R.J. et al. (2017) Genomic analysis of 63,220 tumors JAMA Oncol. 1, 238–244
reveals insights into tumor uniqueness and targeted cancer
61. Wagle, N. et al. (2014) Response and acquired resistance to
immunotherapy strategies. Genome Med. 9, 16
everolimus in anaplastic thyroid cancer. N. Engl. J. Med. 371,
39. Dienstmann, R. et al. (2015) Database of genomic biomarkers 1426–1433
for cancer drugs and clinical targetability in solid tumors. Cancer
62. Wagle, N. et al. (2014) Activating mTOR mutations in a patient
Discov. 5, 118–123
with an extraordinary response on a phase I trial of everolimus
40. Rubio-Perez, C. et al. (2015) In silico prescription of anticancer and pazopanib. Cancer Discov. 4, 546–553
drugs to cohorts of 28 tumor types reveals targeting opportu-
63. Chen, J.C. et al. (2014) Identification of causal genetic drivers of
nities. Cancer Cell 27, 382–396
human disease through systems-level analysis of regulatory
41. Biankin, A.V. et al. (2015) Patient-centric trials for therapeutic networks. Cell 159, 402–414
development in precision oncology. Nature 526, 361–370
64. Luo, J. et al. (2009) Principles of cancer therapy: oncogene and
42. Rashdan, S. and Gerber, D.E. (2016) Going into BATTLE: non-oncogene addiction. Cell 136, 823–837
umbrella and basket clinical trials to accelerate the study of
65. Alizadeh, A.A. et al. (2015) Toward understanding and exploiting
biomarker-based therapies. Ann. Transl. Med. 4, 529
tumor heterogeneity. Nat. Med. 21, 846–853
43. Le Tourneau, C. et al. (2016) Treatment algorithms based on
66. Robinson, D. et al. (2015) Integrative clinical genomics of
tumor molecular profiling: the essence of precision medicine
advanced prostate cancer. Cell 161, 1215–1228
trials. J. Natl. Cancer Inst. 108, djv362
67. Fishbein, L. et al. (2017) Comprehensive molecular characteri-
44. Massard, C. et al. (2017) High-throughput genomics and clinical
zation of pheochromocytoma and paraganglioma. Cancer Cell
outcome in hard-to-treat advanced cancers: results of the
31, 181–193
MOSCATO 01 trial. Cancer Discov. 7, 586–595
68. Kato, S. et al. (2017) RET Aberrations in diverse cancers: next-
45. Le Tourneau, C. et al. (2015) Molecularly targeted therapy based
generation sequencing of 4,871 patients. Clin. Cancer Res. 23,
on tumour molecular profiling versus conventional therapy for
1988–1997
advanced cancer (SHIVA): a multicentre, open-label, proof-of-
concept, randomised, controlled phase 2 trial. Lancet Oncol. 69. Stransky, N. et al. (2014) The landscape of kinase fusions in
16, 1324–1334 cancer. Nat. Commun. 5, 4846
46. Wheler, J.J. et al. (2016) Cancer Therapy directed by compre- 70. Alvarez, M.J. et al. (2016) Functional characterization of somatic
hensive genomic profiling: a single center study. Cancer Res. mutations in cancer using network-based inference of protein
76, 3690–36701 activity. Nat. Genet. 48, 838–947
47. Lopez-Chavez, A. et al. (2015) Molecular profiling and targeted 71. Tian, S. et al. (2013) A combined oncogenic pathway signature
therapy for advanced thoracic malignancies: a biomarker- of BRAF, KRAS and PI3KCA mutation improves colorectal
derived, multiarm, multihistology phase II basket trial. J. Clin. cancer classification and cetuximab treatment prediction. Gut
Oncol. 33, 1000–1007 62, 540–549
48. Stockley, T.L. et al. (2016) Molecular profiling of advanced solid 72. Vecchione, L. et al. (2016) A vulnerability of a subset of colon
tumors and patient outcomes with genotype-matched clinical cancers with potential clinical utility. Cell 165, 317–330
trials: the Princess Margaret IMPACT/COMPACT trial. Genome 73. Lord, C.J. and Ashworth, A. (2016) BRCAness revisited. Nat.
Med. 8, 109 Rev. Cancer 16, 110–120
49. Hyman, D.M. et al. (2015) Vemurafenib in multiple nonmelanoma 74. Konstantinopoulos, P.A. et al. (2010) Gene expression profile of
cancers with BRAF V600 mutations. N. Engl. J. Med. 373, BRCAness that correlates with responsiveness to chemother-
726–736 apy and with outcome in patients with epithelial ovarian cancer.
50. Kopetz, S. et al. (2010) PLX4032 in metastatic colorectal cancer J. Clin. Oncol. 28, 3555–3561
patients with mutant BRAF tumors. J. Clin. Oncol. 28, 3534 75. Larsen, M.J. et al. (2013) Classifications within molecular sub-
51. Kaufman, B. et al. (2015) Olaparib monotherapy in patients with types enables identification of BRCA1/BRCA2 mutation carriers
advanced cancer and a germline BRCA1/2 mutation. J. Clin. by RNA tumor profiling. PLoS One 8, e64268
Oncol. 33, 244–250 76. Van Allen, E.M. et al. (2015) Genomic correlates of response to
52. Al-Ahmadie, H. et al. (2014) Synthetic lethality in ATM-deficient CTLA–4 blockade in metastatic melanoma. Science 350,
RAD50-mutant tumors underlies outlier response to cancer 207–211
therapy. Cancer Discov. 4, 1014–1021 77. Hugo, W. et al. (2016) Genomic and transcriptomic features of
53. Banerji, U. et al. (2014) Abstract LB-66: results of two phase I response to anti-PD-1 therapy in metastatic melanoma. Cell
multicenter trials of AZD5363, an inhibitor of AKT1, 2 and 3: 165, 35–44
189. Miyamoto, D.T. et al. (2015) RNA-Seq of single prostate CTCs 214. Ridky, T.W. et al. (2010) Invasive three-dimensional organotypic
implicates noncanonical Wnt signaling in antiandrogen resis- neoplasia from multiple normal human epithelia. Nat. Med. 16,
tance. Science 349, 1351–1356 1450–1455
190. Motter, A.E. et al. (2008) Predicting synthetic rescues in meta- 215. Kenny, H.A. et al. (2015) Quantitative high throughput screening
bolic networks. Mol. Syst. Biol. 4, 168 using a primary human three-dimensional organotypic culture
predicts in vivo efficacy. Nat. Commun. 6, 6220
191. Rathert, P. et al. (2015) Transcriptional plasticity promotes pri-
mary and acquired resistance to BET inhibition. Nature 525, 216. Vaira, V. et al. (2010) Preclinical model of organotypic culture for
543–547 pharmacodynamic profiling of human tumors. Proc. Natl. Acad.
Sci. U. S. A. 107, 8352–8356
192. van Leeuwen, J. et al. (2016) Exploring genetic suppression
interactions on a global scale. Science 354, aag0839 217. Freeman, A.E. and Hoffman, R.M. (1986) In vivo-like growth of
human tumors in vitro. Proc. Natl. Acad. Sci. U. S. A. 83,
193. Tsimberidou, A.M. et al. (2012) Personalized medicine in a
2694–2698
phase I clinical trials program: the MD Anderson Cancer Center
initiative. Clin. Cancer Res. 18, 6373–6383 218. Meijer, T.G. et al. (2017) Ex vivo tumor culture systems for
functional drug testing and therapy response prediction. Future
194. Schwaederle, M. et al. (2016) Precision oncology: the UC San
Sci. OA 3, FSO190
Diego Moores Cancer Center PREDICT experience. Mol. Can-
cer Ther. 15, 743–752 219. Majumder, B. et al. (2015) Predicting clinical response to anti-
cancer drugs using anex vivo platform that captures tumour
195. Von Hoff, D.D. et al. (2010) Pilot study using molecular profiling
heterogeneity. Nat. Commun. 6, 6169
of patients’ tumors to find potential targets and select treat-
ments for their refractory cancers. J. Clin. Oncol. 28, 4877–4883 220. Holliday, D.L. et al. (2013) The practicalities of using tissue slices
as preclinical organotypic breast cancer models. J. Clin. Pathol.
196. Park, J.W. et al. (2016) Adaptive randomization of neratinib in
66, 253–255
early breast cancer. N. Engl. J. Med. 375, 11–22
221. Naipal, K.A. et al. (2016) Tumor slice culture system to assess
197. Rugo, H.S. et al. (2016) Adaptive randomization of veliparib-
drug response of primary breast cancer. BMC Cancer 16, 78
carboplatin treatment in breast cancer. N. Engl. J. Med. 375,
23–34 222. Roife, D. et al. (2016) Ex vivo testing of patient-derived xeno-
grafts mirrors the clinical outcome of patients with pancreatic
198. Tripathy, D. et al. (2015) Adaptively randomized trial of neo-
ductal adenocarcinoma. Clin. Cancer Res. 22, 6021–6030
adjuvant chemotherapy with or without the Akt inhibitor MK-
2206: graduation results from the I-SPY 2 Trial. J. Clin. Oncol. 223. Wang, Y. et al. (2014) Clonal evolution in breast cancer revealed
33, 524–524 by single nucleus genome sequencing. Nature 512, 155–160
199. Kaplan, R. et al. (2013) Evaluating many treatments and bio- 224. Patel, A.P. et al. (2014) Single-cell RNA-seq highlights intra-
markers in oncology: a new design. J. Clin. Oncol. 31, tumoral heterogeneity in primary glioblastoma. Science 344,
4562–4568 1396–1401
200. Liu, F. et al. (2015) EGFR mutation promotes glioblastoma 225. Tirosh, I. et al. (2016) Single-cell RNA-seq supports a develop-
through epigenome and transcription factor network remodel- mental hierarchy in human oligodendroglioma. Nature 539,
ing. Mol. Cell 60, 307–318 309–313
201. Zhu, J. et al. (2015) Gain-of-function p53 mutants co-opt chro- 226. Gawad, C. et al. (2016) Single-cell genome sequencing: current
matin pathways to drive cancer growth. Nature 525, 206–211 state of the science. Nat. Rev. Genet. 17, 175–188
202. Mertins, P. et al. (2016) Proteogenomics connects somatic 227. Vitak, S.A. et al. (2017) Sequencing thousands of single-cell
mutations to signalling in breast cancer. Nature 534, 55–62 genomes with combinatorial indexing. Nat. Methods 14,
302–308
203. Zhang, B. et al. (2014) Proteogenomic characterization of
human colon and rectal cancer. Nature 513, 382–387 228. Zahn, H. et al. (2017) Scalable whole-genome single-cell library
preparation without preamplification. Nat. Methods 14,
204. Zhang, H. et al. (2016) Integrated proteogenomic characteriza-
167–173
tion of human high-grade serous ovarian cancer. Cell 166,
755–765 229. Klein, A.M. et al. (2015) Droplet barcoding for single-cell tran-
scriptomics applied to embryonic stem cells. Cell 161,
205. Drake, J.M. et al. (2016) Phosphoproteome integration reveals
1187–1201
patient-specific networks in prostate cancer. Cell 166,
1041–1054 230. Macosko, E.Z. et al. (2015) Highly parallel genome-wide expres-
sion profiling of individual cells using nanoliter droplets. Cell 161,
206. Aebersold, R. and Mann, M. (2016) Mass-spectrometric explo-
1202–1214
ration of proteome structure and function. Nature 537, 347–355
231. Zheng, G.X. et al. (2017) Massively parallel digital transcriptional
207. Senft, D. and Ronai, Z.A. (2016) Adaptive stress responses
profiling of single cells. Nat. Commun. 8, 14049
during tumor metastasis and dormancy. Trends Cancer 2,
429–442 232. Tsoucas, D. and Yuan, G.C. (2017) Recent progress in single-
cell cancer genomics. Curr. Opin. Genet. Dev. 42, 22–32
208. Davidson, S.M. et al. (2016) Environment impacts the metabolic
dependencies of ras-driven non-small cell lung cancer. Cell
Metab. 23, 517–528
209. Hensley, C.T. et al. (2016) Metabolic heterogeneity in human
lung tumors. Cell 164, 681–694