Вы находитесь на странице: 1из 7

Seminars in Cancer Biology 42 (2017) 13–19

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Review

Roles of tumor heterogeneity in the development of drug resistance:


A call for precision therapy
Duojiao Wu ∗ , Diane C. Wang, Yunfeng Cheng, Mengjia Qian, Miaomiao Zhang, Qi Shen,
Xiangdong Wang
Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Shanghai, China

a r t i c l e i n f o a b s t r a c t

Article history: The drug resistance limits the optimal efficacy of drugs during target therapies for lung cancer and
Received 8 June 2016 requires the development of precision medicine to identify and develop new highly selective drugs
Accepted 8 November 2016 and more precise tailoring of medicine to the target population. Lung cancer heterogeneity as a poten-
Available online 10 November 2016
tial cause of drug resistance to targeted therapy may foster tumor evolution and adaptation and fade
personalized-medicine strategies. The present review elucidates the influence of tumor heterogeneity
Keywords:
on drug efficacy and resistance, and discusses potential strategies to combat heterogeneity for cancer
Tumor heterogeneity
treatment. There is an urgent need to discover and develop disease- and biology-specific biomarkers
Precision medicine
Genetic variations
for monitoring the existence and occurrence of lung cancer heterogeneity, testing targeted drugs in
Cancer clinical trials, and implementing precision medicine for patients. Better understanding of lung cancer
Drug resistance heterogeneity will strengthen therapeutic strategies and apply precision medicine to cure the disease.
Biomarker © 2016 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2. Disease heterogeneity and drug efficacy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
3. Micro-environment heterogeneity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4. Metastatic heterogeneity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
5. Functional heterogeneity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
6. Clinical potentials of precision medicine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
7. Functional and precision biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
8. Opportunities and challenges of precision therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18

and variants, or protein over-expression [4]. Precision medicine


1. Introduction requires an understanding of cancer genes, mutational processes,
or heterogeneity between cancer cells during tumor evolution.
Precision medicine is proposed as a new strategy to identify and The tumor heterogeneity in diverse cancer types was evidenced
develop new highly selective drugs against specific targets for the by a meta-analysis of 2957 whole exomes and 126 whole genomes
disease and more precise tailoring of medicines to the target popu- [5]. Inter-tumor heterogeneity with limited somatic alterations was
lations [1–3]. Precision medicine can be an important approach to noticed in histopathologic subtype tumors, while intra-tumor het-
create more novel and safer therapeutics for patients with gene erogeneity within individual tumor biopsies of the same tumor and
fusions and mutation, methylation and acetylation, aberrations temporally evolves during the disease course [6–8].
Patients with advanced solid tumors still have poor clinical out-
come, due to the resistance to chemotherapeutics and targeted
∗ Corresponding author. therapies [9].The tumor heterogeneity may exist between individ-
E-mail address: wuduojiao@126.com (D. Wu). uals, organs/tissues, locations, or cells [10,11] and contribute to

http://dx.doi.org/10.1016/j.semcancer.2016.11.006
1044-579X/© 2016 Elsevier Ltd. All rights reserved.
14 D. Wu et al. / Seminars in Cancer Biology 42 (2017) 13–19

low efficacy or failure of therapies through the development of tors determine drug half-life, vascular permeability to drugs or
drug resistance [12,13]. Acquired resistance could develop after acquired resistance to targeted drugs [28]. Cancers with low genetic
an initial phase and be assessed by clinical phenotypes [14,15]. mutational burden are associated with long lasting response to
Resistance mechanisms were involved in secondary pathway targeted therapy, whereas gene-unstable cancers with shorter
mutations or bypass mechanisms within the tumor cells, such as duration of responses. The smoking-associated cancers with high
EGFR(T790M)44 mutations or MET receptor amplification [16,17]. levels of carcinogen-associated genetic mutations may be less to
Understanding of lung cancer heterogeneity can help the develop- derive substantial long-term benefit from targeted therapy.
ment of predictive or prognostic biomarker strategies [18–21], to
monitor the influence of tumor subclones in therapeutic efficacies
3. Micro-environment heterogeneity
and outcomes.
The present review briefly define various heterogeneities and
The environment and/or microenvironment where the human
potential associations with drug efficacy and resistance. We then
and tumor cells live was recently identified as a factor to influence
overview new strategies to increase drug efficacy and minimal-
drug resistance and emphasized as the importance of the tumor
ize the drug resistance and toxicity. We emphasize the importance
cell extrinsic compartments [29,30]. The origin and influence of
to develop functional and precision biomarkers to monitor drug
the micro-environment heterogeneity were involved in the recruit-
efficacy and resistance, and define opportunities and challenges of
ment of fibroblasts, migration of immune cells, matrix remodeling,
precision medicine for clinical practice.
and development of vascular networks [31–34]. Phenotypic and
functional heterogeneity of cancer-associated fibroblasts could
2. Disease heterogeneity and drug efficacy promote tumorigenesis, extracellular matrix production, and
cytokine secretion, including stromal cell-derived factor 1,vascu-
Tumor heterogeneity exists and develops between patients with lar endothelial growth factor, platelet-derived growth factor, and
or without genetic factors. A sequencing analysis of 3281 samples hepatocyte growth factor (HGF) [35]. Cancer-associated stromal
from 12 cancer types demonstrated that acute myeloid leukaemia secretion of HGF could activate the HGF receptor MET, mitogen-
was associated with the lowest mutational burden, while lung activated protein kinase (MAPK), and phosphatidylinositol-3-OH
squamous cell carcinoma with the highest mutational burden kinase (PI3K/AKT) signaling pathways, to have the immediate
[22,23]. Some somatic mutations e.g. methylmalonyl-CoA mutase resistance to RAF inhibition [31]. The activation of MAPK and
−driver genes, caused interpatient tumor heterogeneity [24,25]. PI3 K/AKT-mTOR pathways regulates the formation of the eIF4F
The high mutational burden of lung cancer samples was explained eukaryotic translation complex in BRAF(V600)-mutant tumors.
by the mutagenic effect of carcinogen exposures to e.g. cigarette The persistent formation of the eIF4F complex, binding to the
smoke, and melanoma to ultraviolet radiation [26]. 7-methylguanylate cap (m(7)G) at the 5’ end of messenger RNA
Lung cancer is a molecularly heterogeneous disease. The muta- and modulating the translation of specific mRNAs, was associated
tional landscape of lung adenocarcinoma is substantially different with resistance to anti-BRAF, anti-MEK, and anti-BRAF plus anti-
from that of squamous cell carcinoma or small cell lung cancer MEK drug combinations in BRAF(V600)-mutant melanoma, colon
(SCLC), as shown in Table 1. About 10%-40% mutations in the and thyroid cancer cells [33]. Microvessel density was reported
tyrosine kinase domain of the epidermal growth factor recep- to be a significant prognostic factor for poor outcome in NSCLC
tor (EGFR) were noticed in lung adenocarcinomas [22,27], while [36]. The expression of vascular endothelial growth factor-A was
rarely in squamous cell carcinoma and SCLC [22]. A number of up-regulated with a worse prognosis in lung and renal cancers
TP53, KRAS, STK11, EGFR, and NF1 were mutated in adenocarci- [37]. Tumor infiltrating lymphocytes may recognize neo-antigens
nomas, different from those classified by smoke status and gender. presented on the surface of tumor cells as non-self, promoting
Mutations inTP53, KRAS, LKB1, NF1, and RBM10 were enriched in enhanced T cell activation and immune cell tumor infiltration [38].
transversion-high tumors, while EGFR, RB1, and PIK3CA, and in- T cell activation was involved with stimulatory and inhibitory
frame insertions in the receptor tyrosine kinases EGFR and ERBB2 checkpoint signals to precisely tune responses to prevent excessive
in transversion-low tumors. Mutations in EGFR are associated with damage and autoimmunity. The usurping cytotoxic T cells can be
women, while RBM10 with men. Host genetic or non-genetic fac- activated in tumors through continuous engagement of inhibitory

Table 1
Heterogeneity of lung cancer.

Features Adenocarcinoma Squamous cell carcinoma Small-cell lung cancer

Age-adjusted incidence (Incidence per 100,000 per year) 22.1 14.4 9.8
Napsin-A and TTF-1 immunostaining Napsin-A (+) Napsin-A (−) Napsin-A (−)
TTF-1 (+) TTF-1 (−) TTF-1 (+)
Genomic lateration Mutations TP53, KRAS, EGFR, NF1, BRAF, TP53, CDKN2A, PIK3CA, TP53, RB1, EP300, CREBBP,
MET, RIT NFE2L2, KEAP1, PTEN, SLIT2
CUL3, PTEN, NF1, NOTCH1,2,
and 3, DDR2,
EGFR
Fusions ALK, ROS1, FGFRs
RET,NTRK1,RASSF1A,FZR2
Somatic copy number alteration Gains: NKX2-1, TERT, EGFR, Gains: Chr 3q 26 (SOX2, Gains: MYC, MYCN, MYCL1,
MET, KRAS,ERBB2, MDM2 PIK3CA, TP63 etc) SOX2, FGFR1, KIT
Losses: LRP1B, PTPRD, and Losses: CDKN2A, PTEN Losses: Chr 3p (FHIT, FUS1,
CDKN2A RASSF1A)
Pathway alterations RTK/RAS/RAF Squamous diff erentiation Hedgehog, DNA repair,axonal
mTOR Oxidative stress response guidance and neuroendocrine
JAK-STAT PIK3CA diff erentiation, Cell cycle
DNA repair DNA repair regulation, epigenetic
Cell cycle regulation Cell cycle regulation deregulation,
Epigenetic deregulation Epigenetic deregulation
D. Wu et al. / Seminars in Cancer Biology 42 (2017) 13–19 15

Fig. 1. Interplay of tumor and microenvironment and the therapies of targetting the tumor microenvironment.
(A) Origins and influence of tumor local heterogeneity. Tumor formation involves the co-evolution of neoplastic cells together with extracellular matrix and vascular
endothelial, stromal and immune cells. Several chemokines secreted by cancer-associated fibroblasts(CAFs) modulate the stromal landscape: CXC-chemokine ligand 12
(CCL12) reples T cells, CXCL13 recruits B cells, and CC-chemokine ligand 2 (CCL2), CCL3, CCL4 and CCL5 recruit myeloid cells. Tumor-associated blood endothelial cells(BECs)
upregulated mucosal vascular addressin cell adhension molecule 1(MADCAM1) and CD166 expression, which bind to ␣4␤7 integrin and CD6, respectively, on regulatory T
(Treg) cells and promote their preferential extravasation through the activation of ␣4␤7 integrin. (B) The FDA-approved agents that target the tumor vascular network and
tumor infiltrating immune cells have been listed in the table of Fig. 1B. CSF1R, colony stimulating factor 1 receptor; CTLA-4, cytotoxic T-lymphocyte-associated antigen 4;
DC, dendritic cell; EGFR, epidermal growth factor receptor; FDA, Food and Drug Administration; FLT3, Fms-like tyrosine kinase 3; IFN, interferon; IL-2, interleukin 2; PAP,
prostatic acid phosphatase; PD-1, programmed death-1; PDGFR, Platelet-derived growth factor receptor; PDL1, programmed death ligand 1; PLGF, placental growth factor;
VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor.

receptors, e.g. cytotoxic T-lymphocyte-associated antigen-4 and et al. [48]examined the intratumoral heterogeneity between pri-
programmed death 1 by upregulation of their ligands [39]. mary renal cell carcinoma and its lung metastasis using single-cell
The complexity of the tumor results from continuous crosstalk RNA sequencing. At the single-cell level, metastatic renal carcinoma
between tumor cells and the environment, as explained in Fig. 1A, cells showed an extensive heterogeneity in the activation of the
in addition to the complication of temporal heterogeneity on top of EGFR and Src pathways which needed a combinatorial regimen co-
spatial heterogeneity. The Food and Drug Administration-approved targeting two mutually exclusive pathways for metastatic cancer
therapies targeting multiple compartments of micro-environment cells. The combinatorial strategy showed better efficacy than the
has been listed in Fig. 1B. The question is whether durable responses monotherapy during preclinical validation using patient-derived
result from multi-pronged approaches if tumor cells and their cell- xenograft in vitro and in vivo models.
extrinsic support can be targeted. It is still a challenge to define
the relationship between tumor heterogeneity and therapeutic
5. Functional heterogeneity
response.
The heterogeneity of gene-specific and associated functions,
4. Metastatic heterogeneity e.g. epigenetic changes, posttranslational modifications, or cell dif-
ferentiation, exists and drives the initiation and progression of
The heterogeneity exists between the origin and metastatic cancer, where mutations occur more frequently than expected by
locations or between metastatic locations. Metastasis may be orig- random chance [5,23,49]. Epigenetic molecular profiles of cancers
inated or evolved from a single clone or a cluster of subclones with may reflect the variation of cell sub-compartments in normal cell
founder mutations differing from primary tumors, which can be renewal systems from which cancers arise. Mutations in transcrip-
hardly removed by surgery. Metastatic tumor develop through the tional factors/regulators have tissue specificity, whereas histone
linear or branched evolution which is more responsible for the modifiers are often mutated across several cancer types [23,50].
induction of drug resistance. Tumors with high instability of genes Inherent functional variabilities in tumor propagation con-
evolve in the metastasis, of which a visible mass contains thou- tribute to cancer growth and therapy efficacy. Diffuse intrinsic
sands of cells resistant to drug [40]. The elimination of a subset pontineglioma (DIPG) is a fatal brain cancer in the brainstem,
of metastatic lesions did not improve the long-term survival of the of which78% contained K27M mutation in H3F3A encoding his-
patient, since the lesion commonly has 20 clonal genetic alterations tone H3.3, or related HIST1H3B encoding histone H3.1 [51]. DIPG
[41,42]. Mutations occurred in the founder cell of the metastasis, H3-K27M subgroup was highly mutated in H3F3A, HIST1H3B, or
which can be escaped from the primary tumor. HIST1H3C and had highly unstable genomes [52]. DIPG has novel
Cancer stem cells are a potential contributor for the heterogene- oncogenic mutations connecting tumorigenesis, chromatin regu-
ity and evolution of cell subpopulations or phenotypes [43,44]. The lation, or developmental signaling pathways. H3-K27M subgroup
heterogeneity among cancer stem cells in a tumor can be an another may require multi-modal therapies due to the complexity and het-
driver to form the resistance by genetically distinct subclones erogeneity of the gene, if therapies targeting the histone mutations
within the cancer stem cell compartment rather than in differenti- become available.
ated compartments of a given tumor [45]. Single cell sequencing Chromatin remodeling, histone and DNA methylation, acety-
can detect the heterogeneity between single cells [46,47]. Kim lation, ubiquitination, or pre-mRNA splicing can be recurrently
16 D. Wu et al. / Seminars in Cancer Biology 42 (2017) 13–19

mutated in types of cancers [53,54]. The mutated pathways and are directly involved in the development of acquired multidrug
components include PI3K/AKT-mTOR signaling, KEAP1-NRF2-CUL3 resistance by trans-activating endogenous ABCB1 [64]. Mutation-
apparatus, DNA methylation, p53-related pathways, or mRNA or epigenetics-associated or specific new drugs can target drug
processing. Correlations between DNA methylation with gene resistance-associated/driven gene mutation or epigenomes to pre-
mutation, expression and copy number profiles indicate clinical vent or reverse non-responsiveness to anti-cancer drugs. The
risks for patients. Somatic alterations of 475 driver genes across wild-type p53 represses ABCB1 promoter activity, while mutant
tumor genomes/exomes from nearly 7000 tumors were referred p53 acts oppositely. Preclinical evidence demonstrated that the
for the drug ability and therapeutic design [55]. Driver gene muta- restoration or mimicry of wild-type p53 function could induce a
tions may be subclonal in tumors and be benefited from targeted senescence program in liver carcinomas and sarcomas and apopto-
therapy (Deciphering Anti-tumor Response with Intratumour Het- sis in lymphoma, which consequently leads to tumor clearance or
erogeneity, NIH study trial registration number NCT02183883; regression [65,66].
clintrials.gov). Targeting intratumor heterogeneity and temporal Another strategy is the combination of target therapies as a new
acquisition of subclonal drivers and genomic cancer evolution alternative for the treatment. The combination of ALK and MAPK
could impact therapeutic response, disease relapse, progression, kinase inhibitors was active in an ALK-positive resistant tumor that
and development of precision medicine [56,57]. had developed a MAP2K1 activating mutation. The combination
It is possible to identify driver mutations and causal determi- of EGFR and fibroblast growth factor receptor (FGFR) inhibitors
nants of the disease, upstream genes of functional drivers, and was efficient in an EGFR mutant resistant cancer with a FGFR3
genetic determinants of tumor subtypes for designing new drugs mutation [67]. Combined ALK and SRC (pp60c-src) inhibition was
and detecting drug efficacy. Mutational intratumor heterogene- effective in several ALK-driven diseases. Those preclinical evidence
ity was seen in tumor-suppressor genes, while SETD2, PTEN, and demonstrates that the development of rational drug combinations
KDM5C underwent multiple distinct and spatially separated inac- is critical in anti-cancer and anti-resistance strategies.
tivating mutations within a single tumor [12]. Unfortunately, there A new wave of small-molecular machineries interfere with
is few targeting drugs associated with suppressor genes to balance the functional heterogeneity of cancer cell synthetases [60].
driver mutations. Chemotherapeutics can target a broad spectrum of cellular
Regulatory networks are considered as an important approach machineries (Fig. 3) by crosslinking nucleobases in DNA, block-
to understand the interaction between genes and proteins, define ing DNA replication, repair and nucleotide synthesis, interfering
signal pathways and factors involved in transcriptional regula- with DNA polymerization, or typically using antagonists of ribonu-
tions and intracellular signals, and contribute to development and cleotide reductases or thymidine synthetases. Drugs act on cellular
promotion of tumor heterogeneity [58]. The variation of driver enzyme complexes and target chromatin modification, e.g. histone
factor distributions within the tumor attribute to the occurrence deacetylases, bromodomain-containing proteins, or DNA methyl-
of heterogeneity through driver factor signaling. Tumor growth transferases (Fig. 2A). Drugs can inhibit HSP90, HSP70, HDAC6,
factor-␤-responding progenies differentiate and transcriptionally or different protease activities, e.g. chymotrypsin-like activity,
activate p21, to stabilize NRF2 enhancing glutathione metabolism trypsin-like activity and/or caspase-like activity, within the 26S
and diminishing effectiveness of anti-cancer therapies [59]. It proteasome to disrupt the protein degradation apparatus. The
can be a non-genetic paradigm that the driver factor signaling ubiquitylation machinery and ubiquitin retrieval can also be
contributes to tumor heterogeneity, characteristics, and drug resis- manipulated by small molecules, providing additional opportuni-
tance. ties for interfering with proteasomal degradation (Fig. 2B).
Cancer cell signaling pathways and processes are important
components of functional heterogeneity. Mutated driver genes of
6. Clinical potentials of precision medicine tumors were categorized into 12 pathways (Fig. 2C) to develop
agents and target functional heterogeneity of tumors [50]. Clinical
Anticancer chemotherapeutics have severe side effects, trials should be defined to match actionable oncogenic signatures
although having partial efficacy. Drugs targeting signaling onco- with personalized therapies. The interaction of genetic and non-
proteins were expected to increase specificity and reduce side genetic determinants could influence the functional diversity and
effects, while easily develop the resistance [60]. The drug resistance therapy response for cancers in future.
can be hardly predicted and protected, due to the limited under-
standing of signaling transduction, feedback loops, redundancy,
tumor heterogeneity, or target oncogenic roles. Structures of target 7. Functional and precision biomarkers
oncogenes and corresponding oncoproteins bound to the effectors
or regulators are critical for drug design. Therapeutic strategies There is a need to develop tumor heterogeneity-specific
can be developed on basis of knowledge on precise mechanisms by biomarkers to detect the development and formation of the resis-
which target molecules can be activated or activate elements of the tance to target therapies, and drug-specific biomarkers to detect
downstream as a new way against cancers. One way to overcome drug efficacy, dynamics, metabolisms, pharmacology, or kinetics.
the drug resistance may need to define epigenetic alterations and The epithelial-to-mesenchymal transition gene expression signa-
variations, through alterations of rapid kinetics, acquired drug ture was used as a predictive biomarker of resistance to the EGFR
resistance, or genetic mutations. Transcriptional states through inhibitor erlotinib or PI3K/AKT–mTOR signaling inhibitors [68]. The
reversible histone modifications and DNA methylation patterns failure to assess mutations that drive resistance limits the ability
may cause a dynamic formation and development of tumor to predict response to treatment. Such limitations are also depen-
heterogeneity. dent upon the sensitivity of the method or accuracy of mutations
The multidrug resistance transporter gene ABCB1 is one of present in less than 5% of cells. Chromosomal instability was con-
the most important mechanisms of drug resistance in cancer. sidered as prognostic biomarker of solid tumor and may be related
Histone acetyltransferases and histone deacetylases regulated to increased tumor cell heterogeneity to adapt to environmental
ABCB1 mRNA levels in human carcinoma cells [61]. The upreg- stresses [69]. We have emphasized the urgent need of biomarkers
ulation of ABCB1 by anticancer drugs was associated with a to monitor the processes of tautomerism, depurination, deamina-
dramatically induced acetyl-H3on the multidrug resistance1 P1 tion, slipped strand mispairing, error prone replication by-pass,
promoter [62,63]. Some transcription factors such as C/EBP␤ errors introduced during DNA repair, or induced mutation [70,71].
D. Wu et al. / Seminars in Cancer Biology 42 (2017) 13–19 17

Fig. 2. The drug development strategies based on functional heterogeneity of tumors.


There are emerging evidences of functional heterogeneity of tumor, including DNA methylation, acetylation, network, or signal pathways. The functional heterogeneity could
potentially be drug development strategies. To address these heterogeneity through the following broad mechanisms: (A) for DNA replication and integrity,by crosslinking
nucleobases in DNA and blocking DNA replication; by inhibiting DNA repair; by interfering with the polymerization of DNA; and by inhibiting nucleotide synthesis, typically
using antagonists of ribonucleoside diphosphate reductaseor thymidine synthetase. Chromatin modification can be targeted by drugs to disrupt chromatin contacts or
affect the recruitment of nonhistone proteins to chromatin. The cellular enzyme complexes such as histone deacetylases (HDACs), bromodomain-containing proteins and
DNA methyltransferases are potential targets. (B) Protein chaperones assist in refolding mutated or stress-misfolded proteins. Complexes consist of the heat shock proteins
HSP90 and HSP70, as well as HSP70/HSP90 organizing protein (HOP), multipleco-chaperones, adaptor proteins, the ubiquitin E3 ligase CHIP(carboxy terminus of HSP70-
interacting protein) and the associated HDAC6 (Histone Deacetylase 6). Drugs can inhibit HSP90, HSP70 or HDAC6. Drugs can inhibit different protease activities – for example,
chymotrypsin-like activity, trypsin-like activity and/or caspase-like activity – within the 26S proteasome to disrupt the protein degradation apparatus. Ubiquitination can
affect proteins in many ways: it can signal for their degradation via the proteasome, alter their cellular location, affect their activity, and promote or prevent protein
interactions. The ubiquitylation machinery and ubiquitin retrieval can also be manipulated by small molecules, providing additional opportunities for interfering with
proteasomal degradation. Finally, the cancer cell signaling pathways and the cellular process they regulate compose the complexity of functional heterogeneity of tumors as
well. Generally,the mutated driver genes found in tumors can be classified into one or more of 12 pathways (C) including DNA damage control,chromatin modification, etc.
These pathways can be further organized into three core cellular processes (genome maintenance, cell fate and cell survival). Recognition of these pathways also allow the
development of agents that target functional heterogeneity of tumors.

There is an increasing evidence to screen therapeutic targets and 8. Opportunities and challenges of precision therapy
drug resistance-conferring gene mutations on circulating tumor
cells (CTC) and cell-free circulating tumor DNA (ctDNA) released The high-throughput sequencing revealed that several hundred
from metastatic deposits [72], although values and accuracy to gene mutations underlie tumor heterogeneity. Those findings high-
assess efficacy and resistance of new drugs or drug combinations light the difficulties in developing newer therapies for the disease
remain to be further validated. CTC or ctDNA presents in the major- and suggest that targeted approaches will rely on better under-
ity of patients with cancer, and has independent diagnostic and standing of patient’s tumor. One of the resistance causes to targeted
prognostic significance for targeted therapies [73–75]. The “liquid therapies can be a challenge or invisible obstacle for achievement
biopsy” brings challenges and opportunities related to intra-tumor of precision medicine or “Moonshot” plan . Fig. 3 furthermore
heterogeneity and recurrence of cancer, to identify genetic and describes the influence of tumor heterogeneity in drug efficacy to
epigenetic aberrations and predict drug efficacy and acquired resis- bring opportunities and challenges for precision therapy.
tance. The immune repertoire sequencing on T/B lymphocytes is Tumor heterogeneity should be closely tracked to identify trunk
measured to determine B cell receptor and T cell receptor com- or heterogeneous events by more precise methodologies for under-
plementary diversity decision region and assess the relationship standing of resistance mechanisms. The heterogeneity should be
between the immune repertoire, drug efficacy, or disease [76,77]. qualified for optimal designs of combination therapies to counter-
Driver gene biomarkers can be developed by the integration of act the sub-modal resistant mutation. Specific and efficient drugs
genomic data on tumor heterogeneity, pinpoint driver genes from should be discovered and validated to target the potential escape
functional RNAi screens, cell biology, clinical bioinformatics, drug mechanisms and non-drugable driver mutations. Precise biomark-
sensitivity and specificity, and prognosis. Clinical practices evi- ers should be developed to monitor precision therapy and subclonal
denced that the detection of gene mutations highly depended dynamics of tumor architecture. Thus, we believe the heterogene-
upon the sensitivity of KRAS measurements in EGFR-non-sensitive ity can be a breakthrough point to perform precision medicine to
patients with metastatic colorectal cancer [78,79]. cure patients with cancer.
18 D. Wu et al. / Seminars in Cancer Biology 42 (2017) 13–19

Fig. 3. The challenges and opportunities presented by tumor heterogeneity for precision medicine.
Mutations introduced during primary tumor cell growth result in clonal and metastatic heterogeneity. At the right part, PI3K as representative signal transduction pathways
affected by mutations is illustrated. Activated PI3K-Akt signaling modulates the function of numerous substrates involved in the regulation of cellular growth. PI3K/Akt
signalling pathway components are frequently altered in human cancers. Here we take PI3K as an example to discuss the challenges and opportunities presented by tumor
heterogeneity for precision medicine as following: (1) Tracking Heterogeneity: Biopsies in one region of a heterogeneous tumor will identify trunk events but may also
identify more heterogeneous events. Comparison of paired metastatic samples may enhance the identification of trunk events for therapeutic targeting. (2) Quantifying
Heterogeneity: Measureing the extent ot heterogeneity in cancers will probably lead to rational design of combination therapies to counteract the sub-modal resistant
mutaiton. (3) Clarifying resistance mechanisms: A cataloguing of the potential resistance mechanisms-genetic,epigenetic and adaptive-to specific targeted therapies for each
individual oncogenic aberration. (4) Developing effective agents: The development of a panel of therapeutic agents to target the potential escape mechanisms, and to target
what are at present regarded as non-drugable driver mutations. (5) Monitoring drug efficacy: Developing of precision biomarkers to measure precision medicine and monitor
the subclonal dynamics of tumor architecture.

Conflict of interest [6] C. Swanton, Intratumor heterogeneity: evolution through space and time,
Cancer Res. 72 (19) (2012) 4875–4882.
[7] M. Dolsten, M. Søgaard, Precision medicine: an approach to R&D for
The authors declare that there are no conflicts of interest. delivering superior medicines to patients, Clin. Transl. Med. 1 (1) (2012) 7,
http://dx.doi.org/10.1186/2001-1326-1-7.
[8] M. He, J. Xia, M. Shehab, X.D. Wang, The development of precision medicine in
Acknowledgements clinical practice, Clin. Transl. Med. 4 (December (1)) (2015) 69, http://dx.doi.
org/10.1186/s40169-015-0069-y.
[9] C. Holohan, et al., Cancer drug resistance: an evolving paradigm, Nat. Rev
The work was supported by Shanghai Leading Academic Cancer 13 (10) (2013) 714–726.
Discipline Project (B115), Zhongshan Distinguished Professor [10] N.C. Turner, J.S. Reis-Filho, Genetic heterogeneity and cancer drug resistance,
Lancet Oncol. 13 (4) (2012) e178–85.
Grant (XDW), The National Nature Science Foundation of
[11] F. Niu, D.C. Wang, J.P. Lu, W. Wu, X.D. Wang, Potentials of single-cell biology
China (91230204, 81270099, 81320108001, 81270131, 81400035, in identification and validation of disease biomarkers, J. Cell. Mol. Med. (April)
81570075, 81500058, 81500025), The Shanghai Committee of Sci- (2016), http://dx.doi.org/10.1111/jcmm.12868.
ence and Technology (12JC1402200, 12431900207, 11410708600), [12] P.D. Thomas, M. Kahn, Kat3 coactivators in somatic stem cells and cancer
stem cells: biological roles, evolution, and pharmacologic manipulation, Cell
Zhejiang Provincial Natural Science Foundation (Z15H010002), Biol. Toxicol. 32 (February (1)) (2016) 61–81, http://dx.doi.org/10.1007/
Zhejiang Provincial Science Technology Department Foundation s10565-016-9318-0.
(WKJ-ZJ-1526). [13] N. McGranahan, C. Swanton, Biological and therapeutic impact of intratumor
heterogeneity in cancer evolution, Cancer Cell 27 (1) (2015) 15–26.
[14] X.D. Wang, Cancer Moonshot 2020: a new march of clinical and translational
medicine, Clin. Transl. Med. 5 (2016) 11.
References [15] Y. Zhu, M. Luo, M. Brooks, S.G. Clouthier, M.S. Wicha, Biological and clinical
significance of cancer stem cell plasticity, Clin. Transl. Med. 3 (December (1))
[1] X.D. Wang, Gene mutation-based and specific therapies in precision (2014) 32, http://dx.doi.org/10.1186/s40169-014-0032-3.
medicine, J. Cell Mol. Med. 20 (April ()4) (2016) 577–580, http://dx.doi.org/10. [16] X.D. Wang, K. Li, H. Chen, D. Wang, Y. Zhang, C. Bai, Does hepatocyte growth
1111/jcmm.12722. factor/c-Met signal play synergetic role in lung cancer? J. Cell. Mol. Med. 14
[2] F.S. Collins, H. Varmus, A new initiative on precision medicine, N. Engl. J. Med. (4) (2010) 833–839.
372 (9) (2015) 793–795. [17] M. Zwitter, M. Rajer, K. Stanic, et al., Intercalated chemotherapy and erlotinib
[3] E.A. Ashley, The precision medicine initiative: a new national effort, JAMA 313 for non-small cell lung cancer (NSCLC) with activating epidermal growth
(21) (2015) 2119–2120. factor receptor (EGFR) mutations, Cancer Biol. Ther. (June) (2016) 0.
[4] C. Chen, et al., Five critical elements to ensure the precision medicine, Cancer [18] R. Liu, X.D. Wang, K. Aihara, L. Chen, Early diagnosis of complex diseases by
Metastasis Rev. 34 (2) (2015) 313–318. molecular biomarkers: network biomarkers, and dynamical network
[5] M.S. Lawrence, et al., Mutational heterogeneity in cancer and the search for biomarkers, Med. Res. Rev. 34 (3) (2014) 455–478.
new cancer-associated genes, Nature 499 (7457) (2013) 214–218.
D. Wu et al. / Seminars in Cancer Biology 42 (2017) 13–19 19

[19] C.A. Klein, N.H. Stoecklein, Lessons from an aggressive cancer: evolutionary [50] B. Vogelstein, et al., Cancer genome landscapes, Science 339 (6127) (2013)
dynamics in esophageal carcinoma, Cancer Res. 69 (13) (2009) 5285–5288. 1546–1558.
[20] X. Wu, L. Chen, X.D. Wang, Network biomarkers, interaction networks and [51] G. Wu, et al., Somatic histone H3 alterations in pediatric diffuse intrinsic
dynamical network biomarkers in respiratory diseases, Clin. Transl. Med. pontine gliomas and non-brainstem glioblastomas, Nat. Genet. 44 (3) (2012)
(June (3)) (2014) 16. 251–253.
[21] L.A. Loeb, Human cancers express mutator phenotypes: origin, consequences [52] P. Buczkowicz, et al., Genomic analysis of diffuse intrinsic pontine gliomas
and targeting, Nat. Rev. Cancer 11 (6) (2011) 450–457. identifies three molecular subgroups and recurrent activating ACVR1
[22] S. Devarakonda, D. Morgensztern, R. Govindan, Genomic alterations in lung mutations, Nat. Genet. 46 (5) (2014) 451–456.
adenocarcinoma, Lancet Oncol. 16 (7) (2015) e342–51. [53] I.R. Watson, et al., Emerging patterns of somatic mutations in cancer, Nat. Rev.
[23] C. Kandoth, et al., Mutational landscape and significance across 12 major Genet. 14 (10) (2013) 703–718.
cancer types, Nature 502 (7471) (2013) 333–339. [54] Y. Sato, et al., Integrated molecular analysis of clear-cell renal cell carcinoma,
[24] P.J. Stephens, et al., The landscape of cancer genes and mutational processes Nat. Genet. 45 (8) (2013) 860–867.
in breast cancer, Nature 486 (7403) (2012) 400–404. [55] C. Rubio-Perez, et al., In silico prescription of anticancer drugs to cohorts of 28
[25] T. Shimizu, et al., The clinical effect of the dual-targeting strategy involving tumor types reveals targeting opportunities, Cancer Cell 27 (3) (2015)
PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced 382–396.
cancer, Clin. Cancer Res. 18 (8) (2012) 2316–2325. [56] C. Hiley, et al., Deciphering intratumor heterogeneity and temporal
[26] E. Hodis, et al., A landscape of driver mutations in melanoma, Cell 150 (2) acquisition of driver events to refine precision medicine, Genome Biol. 15 (8)
(2012) 251–263. (2014) 453.
[27] M.B. Carper, P.P. Claudio, Clinical potential of gene mutations in lung cancer, [57] M.et al. Jamal-Hanjani, Tracking genomic cancer evolution for precision
Clin. Transl. Med. 4 (1) (2015) 33, http://dx.doi.org/10.1186/s40169-015- medicine: the lung TRACERx study, PLoS Biol. 12 (7) (2014) e1001906.
0074-1. [58] X. Liu, J. Wang, L. Chen, Whole-exome sequencing reveals recurrent somatic
[28] A. Kreso, et al., Variable clonal repopulation dynamics influence mutation networks in cancer, Cancer Lett. 340 (2) (2013) 270–276.
chemotherapy response in colorectal cancer, Science 339 (6119) (2013) [59] N. Oshimori, D. Oristian, E. Fuchs, TGF-beta promotes heterogeneity and drug
543–548. resistance in squamous cell carcinoma, Cell 160 (5) (2015) 963–976.
[29] T. Zeng, D.C. Wang, X.D. Wang, F. Xu, L. Chen, Prediction of dynamical drug [60] M. Dobbelstein, U. Moll, Targeting tumour-supportive cellular machineries in
sensitivity and resistance by module network rewiring-analysis based on anticancer drug development, Nat. Rev. Drug Discov. 13 (3) (2014) 179–196.
transcriptional profiling, Drug Resist. Updat. 17 (3) (2014 Jul) 64–76. [61] S. Jin, K.W. Scotto, Transcriptional regulation of the MDR1 gene by histone
[30] M.B. Meads, R.A. Gatenby, W.S. Dalton, Environment-mediated drug acetyltransferase and deacetylase is mediated by NF-Y, Mol. Cell. Biol. 18 (7)
resistance: a major contributor to minimal residual disease, Nat. Rev. Cancer 9 (1998) 4377–4384.
(9) (2009) 665–674. [62] Y.H. Kim, D. Jeoung, Tubulin Beta3 serves as a target of HDAC3 and mediates
[31] R. Straussman, et al., Tumour micro-environment elicits innate resistance to resistance to microtubule-targeting drugs, Mol. Cells 38 (8) (2015) 705–714,
RAF inhibitors through HGF secretion, Nature 487 (7408) (2012) 500–504. http://dx.doi.org/10.14348/molcells.2015.0086.
[32] T.R. Wilson, et al., Widespread potential for growth-factor-driven resistance [63] E.K. Baker, et al., Epigenetic changes to the MDR1 locus in response to
to anticancer kinase inhibitors, Nature 487 (7408) (2012) 505–509. chemotherapeutic drugs, Oncogene 24 (54) (2005) 8061–8075.
[33] L. Boussemart, et al., eIF4F is a nexus of resistance to anti-BRAF and anti-MEK [64] G.K. Chen, et al., CCAAT/enhancer-binding protein beta (nuclear factor for
cancer therapies, Nature 513 (7516) (2014) 105–109. interleukin 6) transactivates the human MDR1 gene by interaction with an
[34] M.R. Junttila, F.J. de Sauvage, Influence of tumour micro-environment inverted CCAAT box in human cancer cells, Mol. Pharmacol. 65 (4) (2004)
heterogeneity on therapeutic response, Nature 501 (7467) (2013) 906–916.
346–354. [65] W. Xue, et al., Senescence and tumour clearance is triggered by p53
[35] U.M. Polanska, A. Orimo, Carcinoma-associated fibroblasts: non-neoplastic restoration in murine liver carcinomas, Nature 445 (7128) (2007) 656–660.
tumour-promoting mesenchymal cells, J. Cell. Physiol. 228 (8) (2013) [66] A. Ventura, et al., Restoration of p53 function leads to tumour regression in
1651–1657. vivo, Nature 445 (7128) (2007) 661–665.
[36] R. Berardi, F. Morgese, A. Onofri, et al., Role of maspin in cancer, Clin. Transl. [67] A.S. Crystal, et al., Patient-derived models of acquired resistance can identify
Med. 2 (1) (2013) 8, http://dx.doi.org/10.1186/2001-1326-2-8. effective drug combinations for cancer, Science 346 (6216) (2014) 1480–1486.
[37] P.S. Hegde, et al., Predictive impact of circulating vascular endothelial growth [68] L.A. Byers, et al., An epithelial-mesenchymal transition gene signature
factor in four phase III trials evaluating bevacizumab, Clin. Cancer Res. 19 (4) predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a
(2013) 929–937. therapeutic target for overcoming EGFR inhibitor resistance, Clin. Cancer Res.
[38] J. Zhou, Z.T. Zhu, C.X. Ba, H.Z. Sun, X.D. Wang, Proteomic profiling of 19 (1) (2013) 279–290.
lymphocytes in autoimmunity, inflammation and cancer, J. Transl. Med. 12 [69] C. Swanton, et al., Chromosomal instability determines taxane response, Proc.
(2014) 6. Natl. Acad. Sci. U. S. A. 106 (21) (2009) 8671–8676.
[39] D.M. Pardoll, The blockade of immune checkpoints in cancer immunotherapy, [70] D. Wu, X. Wang, Application of clinical bioinformatics in lung cancer-specific
Nat. Rev. Cancer 12 (4) (2012) 252–264. biomarkers, Cancer Metastasis Rev. 34 (2) (2015) 209–216.
[40] B.J. Monk, T.J. Herzog, K.S. Tewari, Evolution of Chemosensitivity and [71] X. Wang, A.A. Adjei, Lung cancer and metastasis: new opportunities and
resistance assays as predictors of clinical outcomes in epithelial ovarian challenges, Cancer Metastasis Rev. 34 (2) (2015) 169–171.
cancer patients, Curr. Pharm. Des. (May) (2016) 41. [72] K. Pantel, C. Alix-Panabieres, Real-time liquid biopsy in cancer patients: fact
[41] S. Heerboth, G. Housman, M. Leary, et al., EMT and tumor metastasis, Clin. or fiction? Cancer Res. 73 (21) (2013) 6384–6388.
Transl. Med. 4 (2015) 6, http://dx.doi.org/10.1186/s40169-015-0048-3. [73] M. Westphal, K. Lamszus, Circulating biomarkers for gliomas, Nat. Rev.
[42] M.F. Clarke, et al., Cancer stem cells—perspectives on current status and Neurol. 11 (10) (2015) 556–566.
future directions: AACR Workshop on cancer stem cells, Cancer Res. 66 (19) [74] J. Tabernero, et al., Analysis of circulating DNA and protein biomarkers to
(2006) 9339–9344. predict the clinical activity of regorafenib and assess prognosis in patients
[43] J. Gu, X.D. Wang, New future of cell biology and toxicology: thinking deeper, with metastatic colorectal cancer: a retrospective, exploratory analysis of the
Cell Biol. Toxicol. 1 (2016) 1–3, http://dx.doi.org/10.1007/s10565-016-9313-5 CORRECT trial, Lancet Oncol. 16 (8) (2015) 937–948.
(Epub 2016 Feb 13). [75] P.P. Lin, Integrated EpCAM-independent subtraction enrichment and iFISH
[44] K. Anderson, et al., Genetic variegation of clonal architecture and propagating strategies to detect and classify disseminated and circulating tumors cells,
cells in leukaemia, Nature 469 (7330) (2011) 356–361. Clin. Transl. Med. 4 (1) (2015) 38, http://dx.doi.org/10.1186/s40169-015-
[45] B. Bakker, A. Taudt, M.E. Belderbos, et al., Single-cell sequencing reveals 0081-2.
karyotype heterogeneity in murine and human malignancies, Genome Biol. [76] J.R. McDaniel, et al., Ultra-high-throughput sequencing of the immune
17 (1) (2016) 115, http://dx.doi.org/10.1186/s13059-016-0971-7. receptor repertoire from millions of lymphocytes, Nat. Protoc. 11 (3) (2016)
[46] S. Ling, et al., Extremely high genetic diversity in a single tumor points to 429–442.
prevalence of non-Darwinian cell evolution, Proc. Natl. Acad. Sci. U. S. A. 112 [77] H. Robins, Immunosequencing: applications of immune repertoire deep
(47) (2015) E6496–E6505. sequencing, Curr. Opin. Immunol. 25 (5) (2013) 646–652.
[47] K.T. Kim, et al., Application of single-cell RNA sequencing in optimizing a [78] F. Molinari, et al., Increased detection sensitivity for KRAS mutations
combinatorial therapeutic strategy in metastatic renal cell carcinoma, enhances the prediction of anti-EGFR monoclonal antibody resistance in
Genome Biol. 17 (1) (2016) 80. metastatic colorectal cancer, Clin. Cancer Res. 17 (14) (2011) 4901–4914.
[49] B.L. Brücher, Y. Li, P. Schnabel, et al., Genomics, microRNA, epigenetics, and [79] W. De Roock, et al., Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the
proteomics for future diagnosis, treatment and monitoring response in upper efficacy of cetuximab plus chemotherapy in chemotherapy-refractory
GI cancers, Clin. Transl. Med. 5 (1) (2016) 13, http://dx.doi.org/10.1186/ metastatic colorectal cancer: a retrospective consortium analysis, Lancet
s40169-016-0093-6. Oncol. 11 (8) (2010) 753–762.

Вам также может понравиться