ArticlePDF Available

Navigating the Challenge of Tumor Heterogeneity in Cancer Therapy

Authors:

Abstract and Figures

The future of cancer treatment lies in personalized strategies designed to specifically target tumorigenic cell populations present in an individual. Although recent advances in directed therapies have greatly improved patient outcomes in some cancers, intuitive drug design is proving more difficult than expected owing largely to the complexity of human cancers. Intratumoral heterogeneity, the presence of multiple genotypically and/or phenotypically distinct cell subpopulations within a single tumor, is a likely cause of drug resistance. Advances in systems biology are helping to unravel the mysteries of cancer progression. In this issue of Cancer Discovery, Zhao and colleagues define a path for functional validation of computational modeling in the context of heterogeneous tumor populations and their potential for drug response and resistance. Cancer Discov; 4(2); 146–8. ©2014 AACR. See related article by Zhao et al., p. 166
Content may be subject to copyright.
2014;4:146-148. Cancer Discovery
Clare Fedele, Richard W. Tothill and Grant A. McArthur
Therapy
Navigating the Challenge of Tumor Heterogeneity in Cancer
Updated version
http://cancerdiscovery.aacrjournals.org/content/4/2/146
Access the most recent version of this article at:
Cited Articles
http://cancerdiscovery.aacrjournals.org/content/4/2/146.full.html#ref-list-1
This article cites by 10 articles, 3 of which you can access for free at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.org
To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at
Permissions
.permissions@aacr.org
To request permission to re-use all or part of this article, contact the AACR Publications Department at
Research.
on February 6, 2014. © 2014 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from
Research.
on February 6, 2014. © 2014 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from
146|CANCER DISCOVERY"FEBRUARY 2014 www.aacrjournals.org
VIEWS
Authors’ Affi liations: 1 Cancer Therapeutics Program, Division of Cancer
Research, Peter MacCallum Cancer Centre, East Melbourne;
2 Sir Peter
MacCallum Department of Oncology and
3 Department of Pathology, Uni-
versity of Melbourne, Parkville; and
4 Department of Medicine, St Vincent’s
Hospital, University of Melbourne, Fitzroy, Victoria, Australia
Corresponding Author: Grant A. McArthur, Divisions of Cancer Medicine
and Research, Peter MacCallum Cancer Centre, St Andrews Place, Locked
Bag 1, A’Beckett Street, East Melbourne, VIC 8006, Australia. Phone: 61-3-
9656-1954; Fax: 61-3-9656-3717; E-mail: grant.mcarthur@petermac.org
doi: 10.1158/2159-8290.CD-13-1042
©2014 American Association for Cancer Research.
IN THE SPOTLIGHT
Navigating the Challenge of Tumor Heterogeneity
in Cancer Therapy
Clare Fedele
1 , 2 , 3 , Richard W. Tothill
1 , 3 , and Grant A. McArthur
1 , 2 , 3
,
4
Summary: Th e f ut u r e o f c a n c e r t r e a t m e n t l i e s i n p e r s on a l i z e d s tr at e g i e s d e si g n e d t o sp e c i c a ll y t ar g e t t u m o r-
igenic cell populations present in an individual. Although recent advances in directed therapies have greatly
improved patient outcomes in some cancers, intuitive drug design is proving more diffi cult than expected owing
largely to the complexity of human cancers. Intratumoral heterogeneity, the presence of multiple genotypically
and/or phenotypically distinct cell subpopulations within a single tumor, is a likely cause of drug resistance.
Advances in systems biology are helping to unravel the mysteries of cancer progression. In this issue of Cancer
Discovery , Z ha o a n d co l l e a g u e s d e n e a p a t h f o r f u n c t io n a l v a l i da t i o n o f c o m p u ta t i o n a l m od e l i n g i n t h e c o n t e x t
of heterogeneous tumor populations and their potential for drug response and resistance. Cancer Discov; 4(2);
146–8. ©2014 AACR.
See related article by Zhao et al., p. 166 (5).
As rst proposed by pioneers of cancer evolution the-
ory, such as Nowell and Vogelstein, cancer results from the
sequential acquisition of heritable genetic and epigenetic
events, which under selective pressures leads to propaga-
tion of dominant cancer cell populations ( 1, 2 ). Although
we usually think of cancer as having a dominant clone, it
is becoming increasingly apparent that the propensity for
cancer evolution can also lead to signifi cant genetic hetero-
geneity in an individual patient’s cancer ( 3 ). This is perhaps
the greatest challenge to the concept of personalized cancer
medicine, as one drug targeting a single genetic driver may
not be suffi cient for adequate disease control. This concept
is not new, and the use of multiagent chemotherapy may pre-
vent the emergence of drug resistance ( 4 ), just as multiagent
antibiotics prevent the emergence of resistance in tuberculo-
sis. The advent of new technologies such as next-generation
sequencing, recently approved by the U.S. Food and Drug
Administration (FDA), is now providing the tools to accu-
rately defi ne the genomic landscape of an individual patient’s
cancer and measure heterogeneity. This approach offers the
potential to rationally target an individual patient’s cancer.
One simple concept of heterogeneity is the existence of
subclones of resistant cells, such as BCR–ABL acute myel-
ogenous leukemia (AML) with a subpopulation of cells with
T315I mutations, or BRAF -mutant melanoma with a sub-
population of cells with NRAS mutations. In this model,
ABL or BRAF inhibition kills the sensitive cells, allowing the
outgrowth of resistant cells. The solution, then, is to inhibit
the resistant cells upfront with a T315I inhibitor or the
combination of a BRAF and MAP–ERK kinase (MEK) inhibi-
tor that kills BRAF / NRAS –mutant cells. However, this is a
simple case of oncogene addiction with the straightforward
approach of turning off the signaling of an oncogene. In real-
ity, cancer can have complex mechanisms of drug resistance,
including antiapoptotic signals and alterations to cell-cycle
checkpoints or mechanisms of DNA repair. So, how can we
deal with such biologic complexity that overlies the inher-
ently heterogeneous nature of human cancer? The answer
may come from providing the road map of an individual
patient’s cancers using sophisticated genomic and epigenetic
interrogation of a cancer coupled with functional knowledge
of how the heterogeneity can be targeted. Is it then simply a
case of adding together drugs that each alone or in synergy
kill the major subpopulations of cells? The study of Zhao and
colleagues (5) in this issue of Cancer Discovery indicates it may
not be that simple.
Zhao and colleagues (5) address the impact of intratumoral
heterogeneity on treatment response using computational
modeling and functional validation in Eμ-Myc;p19
Arf / mouse
lymphoma cells, a well-characterized model of human MYC-
driven lymphoma. This current study builds on previous
work by this laboratory, which determined the responses
of homogenous short hairpin RNA (shRNA)–expressing
Eμ-Myc;p19
Arf / cell populations to combination therapies
(6, 7 ). By using this dataset of drug–genotype interactions,
Zhao and colleagues (5) devised a mathematical algorithm
to predict the response of known mutant-cell populations to
two-drug combination therapies. To phenocopy intratumoral
heterogeneity, three-component tumor populations were gen-
erated by combining two shRNA-expressing subpopulations
Research.
on February 6, 2014. © 2014 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from
FEBRUARY 2014!CANCER DISCOVERY|147
VIEWS
with parental cells. Interestingly, although most preclini-
cal drug development studies use sensitivity as readout of
drug effi cacy, here, prevention of subclonal resistance was
the desired outcome. Computational simulation of different
tumor compositions was undertaken and optimal combina-
tion therapy was systematically predicted. Through these
analyses, Zhao and colleagues (5) discovered a key nding:
that the optimal treatment for a heterogenous tumor may
not necessarily incorporate drugs that most effi ciently kill
each population separately.
In one such case, homogenous cell populations expressing
either Chk2 - or Bok -targeted shRNAs (sh Chk2 or sh Bok ) were
not predicted to have optimal sensitivity to SAHA treatment
alone or in combination with vincristine (Vin). However,
when resistance was taken into consideration, a heterogene-
ous tumor composed of both subpopulations was predicted
to optimally respond to Vin/SAHA treatment. Therefore, the
overall composition of a tumor may dictate the best treat-
ment option, and this may not necessarily follow an intuitive
rationale if considering drug sensitivity alone.
On the basis of these fi ndings, functional validation was
performed in vitro . As predicted computationally, treatment
of heterogeneous sh Chk2 /sh Bok cell populations with Vin/
SAHA resulted in effi cient killing of sh Bok cells while pre-
venting sh Chk2 population outgrowth. In contrast, treat-
ment with an alternate strategy systematically predicted
to be least effective [irinotecan/chlorambucil (IRT/CBL)]
resulted in strong selection and outgrowth of a resistant
sh Chk2 population. The ef cacy of Vin/SAHA treatment was
maintained even when an additional level of genetic com-
plexity was introduced into the system through inclusion
of a cell population expressing both sh Chk2 and sh Bok . A s
expected, drug combinations predicted to optimally target
each cell subpopulation individually were not as effi cient in
the context of population heterogeneity.
The authors further validated their model in vivo . s h Chk2
a n d s h Bok Eμ-Myc;p19
Arf / lymphoma cells were cotransplanted
into immunocompetent mice, followed by systemic treatment
with different drug combinations. As predicted, the combina-
tion of Vin/SAHA successfully prevented selective outgrowth
of any subpopulation. Signifi cantly, tumor-free survival was
also improved compared with IRT/CBL treatment. By experi-
mentally determining the responses of homogeneous tumors
to drug combinations in vivo , t h e a u t h o r s t h e n e x p a n d e d
their analyses to computationally predict the in vivo r e s p o n s e
of three-component heterogeneous tumors with all possible
subpopulation proportions (i.e., 0%–100%). Interestingly, sys-
tematic modeling predicted that even the presence of a low-
abundance cell variant (0.1% of the overall population) is
suffi cient to affect the overall tumor response to treatments.
These studies highlight the potential power of compu-
tational approaches in modeling and predicting biologic
behavior. Over the last decade, advances in systems biology
have greatly enhanced our understanding of complex bio-
logic systems, in particular human cancers. Intratumoral
heterogeneity presents a signifi cant challenge to the devel-
opment of effective anticancer therapies. In the endeavor to
develop better treatments, systems approaches are likely to
play a fundamental role. Toward this end, large-scale molecu-
lar analysis and drug screening of human cancer cell lines
have been undertaken, such as the human Cancer Cell Line
Encyclopedia and Genomics of Drug Sensitivity studies ( 8, 9 ).
Zhao and colleagues (5) provide functional validation of
such systematic approaches, providing a strong foundation
for future predictive computing in preclinical drug develop-
ment and clinical decision-making. In particular, this study
highlights the importance of considering resistance as a sig-
nifi cant contributing factor in overall drug effi cacy, especially
in the context of intratumoral heterogeneity. Currently, very
few drugs that show promise in preclinical testing, mostly on
homogenous populations of cancer cell lines, actually trans-
late to clinical effi cacy ( 10 ).
H o w e v e r , t h e a p p l i c a t i o n o f s y s t e m s b i o l o g y t o c a n c e r t r e a t -
ment remains in its relative infancy, and there are major hurdles
still to be overcome. Computational modeling requires intimate
knowledge of subpopulations present within a heterogeneous
tumor. We are only beginning to understand the complexity
of some human tumors and the level of genetic and epigenetic
heterogeneity present therein, which are extremely diffi cult to
model experimentally. One particularly challenging concept
is that cancer is not a static entity and that many tumors can
potentially undergo continual genetic evolution, allowing adap-
tation to new selective pressures such as anticancer treatment.
It is unclear how systematic modeling will be able to cope with
genomic instability and seemingly random genetic alterations
occurring within cancer cells, not to mention the added com-
plication contributed by nongenetic factors affecting reversible
tumor cell behavior, including microenvironmental infl uences
such as stromal components and even exosomes.
N o t w i t h s t a n d i n g t h e b i o l o g i c c o m p l e x i t i e s o f m o d e l i n g
cancer overall, an additional technical challenge in clinical
translation is considering to what extent a single biopsy will be
sensitive enough to identify all subpopulations present within
a tumor. This may be particularly diffi cult in solid cancers.
Liquid biopsies, including circulating tumor cells and cell-free
tumor DNA, could be part of the answer as they will allow
continued monitoring of cancer progression without the need
for more-invasive tissue biopsy. As we build large compendia
and knowledge on drug-to-genotype relationships, focus must
therefore also be given to better detection strategies.
But even in the face of such challenges, the potential power
of computational modeling cannot be underestimated when
considering the future of personalized cancer treatment.
Knowledge of a patient’s tumor composition may allow for
systematic prediction of therapy response, based on pre-
established drug–genotype matrices, that permits clinicians
to make rational decisions on treatment strategies ( Fig. 1 ).
This may even extend to multiple rounds of treatment to
sequentially target individual subpopulations that may arise
de novo . Currently, next-generation sequencing provides the
tool to form a detailed picture of the genetic composition of
a patient’s cancer. Although it does not directly give infor-
mation of the host microenvironment or subpopulations
of cancer-initiating cells or cancer stem cells, it does allow
description of heterogeneity and the frequency of genetic
subpopulations. If these data are integrated with a broad
and deep knowledge base of the ability of drugs to synergize
to target heterogeneous cell populations, then the goal of
a personalized cancer medicine in a signifi cant proportion
of cancers will be within our grasp. To reach this goal, we
Research.
on February 6, 2014. © 2014 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from
148|CANCER DISCOVERY"FEBRUARY 2014 www.aacrjournals.org
VIEWS
Figure 1. The proposed future of personalized cancer medicine. Bioinformatic analysis of a biopsy taken from a patient tumor will reveal the presence
of distinct cell subpopulations. Consultation of an established drug–genotype matrix will allow treating clinicians to computationally determine optimal
combination therapies specifi c for that patient.
Patient tumor
Drug 1
Drug 2
Drug 3
Drug 4
Possible tumor cell subpopulations
Drug response
Sensitive Resistant
must continue to invest in preclinical and clinical functional
datasets of combination drug therapies powered by compu-
tational biologic models to provide the data systems needed
for personalized cancer medicine.
Disclosure of Potential Confl icts of Interest
No potential confl icts of interest were disclosed.
Published online February 5, 2014.
REFERENCES
1. Nowell PC . The clonal evolution of tumor cell populations . Science
1976 ; 194 : 23 8 .
2. Vogelstein B , Fearon ER , Hamilton SR , Kern SE , Preisinger AC , Lep-
pert M , etal. Genetic alterations during colorectal-tumor develop-
ment . N Engl J Med 1988 ; 319 : 525 32 .
3. Burrell RA , McGranahan N , Bartek J , Swanton C . The causes and
consequences of genetic heterogeneity in cancer evolution . Nature
2013 ; 501 : 338 45 .
4. DeVita VT Jr, Young RC , Canellos GP . Combination versus single
agent chemotherapy: a review of the basis for selection of drug treat-
ment of cancer . Cancer 1975 ; 35 : 98 110 .
5 . Z h a o B , P r i t c h a r d J R , L a u f f e n b u r g e r D A , H e m a n n M T . A d d r e s s -
ing genetic tumor heterogeneity through computationally predictive
combination therapy. Cancer Discov 2014;4:166–74 .
6 . J i a n g H , P r i t c h a r d J R , W i l l i a m s R T , L a u f f e n b u r g e r D A , H e m a n n M T .
A mammalian functional-genetic approach to characterizing cancer
therapeutics. Nat Chem Biol 2011;7:92–100.
7. Pritchard JR, Bruno PM, Gilbert LA, Capron KL, Luaffenburger DA,
Hemann MT. Defi ning principles of combination drug mechanisms
of action. Proc Natl Acad Sci U S A 2013;110:E170–9.
8. Barretina J , Caponigro G , Stransky N , Venkatesan K , Margolin
A A , K i m S , e t al. The Cancer Cell Line Encyclopedia enables pre-
dictive modelling of anticancer drug sensitivity . Nature 2012 ; 483 :
603 7 .
9. Yang W , Soares J , Greninger P , Edelman EJ , Lightfoot H , Forbes S ,
etal. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for
therapeutic biomarker discovery in cancer cells . Nucleic Acids Res
2013 ; 41 : D955 61 .
10. Hutchinson L , Kirk R . High drug attrition rates—where are we going
wrong? Nat Rev Clin Oncol 2011 ; 8 : 189 90 .
Research.
on February 6, 2014. © 2014 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from
... Tumor heterogeneity is associated with cancer progression, recurrence, and responses to drug treatments. 1 Determining the cell composition of tumors is the key to stratifying treatments for cancer patients and developing personalized therapies. 2 Pharmacogenomics, the science of uncovering the genetic de-terminants of drug responses, 3 is a potential solution for stratification and customization of cancer treatments. However, largescale drug response datasets are mainly derived from cancer cell lines, with limited screening data from patient-derived xenograft (PDX) models. ...
Article
Full-text available
Large-scale cancer drug sensitivity data have become available for a collection of cancer cell lines, but only limited drug response data from patients are available. Bridging the gap in pharmacogenomics knowledge between in vitro and in vivo datasets remains challenging. In this study, we trained a deep learning model, Scaden-CA, for deconvoluting tumor data into proportions of cancer-type-specific cell lines. Then, we developed a drug response prediction method using the deconvoluted proportions and the drug sensitivity data from cell lines. The Scaden-CA model showed excellent performance in terms of concordance correlation coefficients (>0.9 for model testing) and the correctly deconvoluted rate (>70% across most cancers) for model validation using Cancer Cell Line Encyclopedia (CCLE) bulk RNA data. We applied the model to tumors in The Cancer Genome Atlas (TCGA) dataset and examined associations between predicted cell viability and mutation status or gene expression levels to understand underlying mechanisms of potential value for drug repurposing.
... Nanostructure-mediated PDT and PTT in combination with ICIs has been actively investigated for cancer treatment [19,[26][27][28][29]. However, some solutions should be required for following issues: cancer heterogeneity and resistancerelated insufficient therapeutic outcomes, low cancer accumulation efficiency, and unwanted movement to normal organs and tissues [30,31]. ...
Article
Alum-crosslinked hyaluronic acid-dopamine (HD) hydrogel containing indocyanine green (ICG) with anti-programmed cell death-1 (PD-1) antibody (Ab) administration was developed for immunophoto therapy of cancer. Alum modulates the rheological characteristics of hydrogel for enabling syringe injection, shear-thinning feature, and slower biodegradation. In addition, alum in HD-based hydrogel provided CD8+ T cell-mediated immune responses for cancer therapy. ICG in the hydrogel under near-infrared (NIR) light exposure may induce hyperthermia and generate singlet oxygen for selective cancer cell killing. HD/alum/ICG hydrogel injection with NIR laser irradiation elevated PD-1 level in CD8+ T cells. Administration of PD-1 Ab aiming at highly expressed PD-1 in T cells may amplify the anticancer efficacies of HD/alum/ICG hydrogel along with NIR laser. HD/alum/ICG hydrogel with NIR light may have both CD8+ T cell-linked immune responses and ICG-related photodynamic/photothermal effects. Additional injection of immune checkpoint inhibitor can ultimately suppress primary and distant tumor growth by combination with those therapeutic actions.
... Hence, treating cancer requires a multidisciplinary approach and involves various therapeutic strategies. Thereby, challenges in cancer therapy include the problem of heterogeneity of cancers, the resistance to medicines, and the side effects of treatment, especially in chemotherapy and radiation therapy but also in targeted therapies 6,7 . ...
Article
Full-text available
Based on prior research findings with pentacyclic triterpenoids, it was hypothesized that (un)-substituted benzylamides would exhibit enhanced cytotoxic activity compared to parent abietic acid. Conversely, none of these compounds was cytotoxic, but (homo)-piperazinyl amides demonstrated significant cytotoxic activity across multiple cell lines, even at concentrations as low single-digit micromolar levels. Additional staining experiments revealed that the most potent compound (with an EC50 value of 2.8 μM for HT29 colon carcinoma cells) primarily induced apoptosis rather than necrosis.
... Heterogeneous tumors create three major obstacles to treatment. First, cells can have different susceptibilities to treatment, meaning that even a targeted treatment with high efficacy can fail to kill or inhibit a subset of cancer cells 18,19 . Second, this differential survival and proliferation can promote the continued evolution of tumor resistance during drug therapy 16,20 . ...
Article
Full-text available
The interplay of positive and negative interactions between drug-sensitive and resistant cells influences the effectiveness of treatment in heterogeneous cancer cell populations. Here, we study interactions between estrogen receptor-positive breast cancer cell lineages that are sensitive and resistant to ribociclib-induced cyclin-dependent kinase 4 and 6 (CDK4/6) inhibition. In mono- and coculture, we find that sensitive cells grow and compete more effectively in the absence of treatment. During treatment with ribociclib, sensitive cells survive and proliferate better when grown together with resistant cells than when grown in monoculture, termed facilitation in ecology. Molecular, protein, and genomic analyses show that resistant cells increase metabolism and production of estradiol, a highly active estrogen metabolite, and increase estrogen signaling in sensitive cells to promote facilitation in coculture. Adding estradiol in monoculture provides sensitive cells with increased resistance to therapy and cancels facilitation in coculture. Under partial inhibition of estrogen signaling through low-dose endocrine therapy, estradiol supplied by resistant cells facilitates sensitive cell growth. However, a more complete blockade of estrogen signaling, through higher-dose endocrine therapy, diminished the facilitative growth of sensitive cells. Mathematical modeling quantifies the strength of competition and facilitation during CDK4/6 inhibition and predicts that blocking facilitation has the potential to control both resistant and sensitive cancer cell populations and inhibit the emergence of a refractory population during cell cycle therapy.
... Abundant evidence has shown that ITH is linked to tumor progression, immune evasion, therapeutic resistance, and unfavorable clinical outcomes [1]. Drug resistance can occur via different mechanisms related to genetic, epigenetic, and phenotypic heterogeneity [2]. Moreover, different cells in a tumor may have different oncogenic drivers, making it difficult to implement effective targeted therapies [3]. ...
Article
Background: Intratumor heterogeneity (ITH) plays a crucial role in tumor progression, relapse, immune evasion, and drug resistance. Existing ITH quantification methods based on a single molecular level are inadequate to capture ITH evolving from genotype to phenotype. Methods: We designed a set of information entropy (IE)-based algorithms for quantifying ITH at the genome (somatic copy number alterations and mutations), mRNA, microRNA (miRNA), long non-coding RNA (lncRNA), protein, and epigenome level, respectively. We evaluated the performance of these algorithms by analyzing the correlations between their ITH scores and ITH-associated molecular and clinical features in 33 TCGA cancer types. Moreover, we evaluated the correlations between the ITH measures at different molecular levels by Spearman correlation and clustering analysis. Results: The IE-based ITH measures had significant correlations with unfavorable prognosis, tumor progression, genomic instability, antitumor immunosuppression, and drug resistance. The mRNA ITH showed stronger correlations with the miRNA, lncRNA, and epigenome ITH than with the genome ITH, supporting the regulatory relationships of miRNA, lncRNA, and DNA methylation towards mRNA. The protein-level ITH displayed stronger correlations with the transcriptome-level ITH than with the genome-level ITH, supporting the central dogma of molecular biology. Clustering analysis based on the ITH scores identified four subtypes of pan-cancer showing significantly different prognosis. Finally, the ITH integrating the seven ITH measures displayed more prominent properties of ITH than that at a single level. Conclusions: Here we regard each gene, mRNA, miRNA, lncRNA, protein, and methylation region (or CpG island) as the basic element of their respective molecular systems. Our analysis suggests that it is the variation of the molecular system's perturbation rather than the molecular system's perturbation itself that correlates with cancer development and outcomes. In fact, even though all or most elements display dramatic perturbations simultaneously to result in a large-scale perturbation of the molecular systems in cancer patients, they are likely to have favorable outcomes if the elements' perturbations are homogeneous or harmonious to yield the low ITH. Therefore, to assess the ITH, the perspective of molecular system biology is preferred to that of single molecule biology. To translate this discovery into clinical practice, a system's test of multiple molecules instead of a single molecule may improve personalized diagnosis and treatment of cancer patients.
... Cancer heterogeneity has been recognized as an important clinical determinant of patient outcomes, such as response or resistance to anti-cancer therapies [11,12]. Heterogeneity is prevalent in cancer, both between and within individuals. ...
Article
Full-text available
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide. HCC is diagnosed in its advanced stage when limited treatment options are available. Substantial morphologic, genetic and epigenetic heterogeneity has been reported in HCC, which poses a challenge for the development of a targeted therapy. In this review, we discuss the role and involvement of several microRNAs (miRs) in the heterogeneity and metastasis of hepatocellular carcinoma with a special emphasis on their possible role as a diagnostic and prognostic tool in the risk prediction, early detection, and treatment of hepatocellular carcinoma.
Article
Breast tumors often display an astonishing degree of spatial and temporal heterogeneity, which are associated with cancer progression, drug resistance, and relapse. Triple-negative breast cancer (TNBC) is a particularly aggressive and heterogeneous subtype for which targeted therapies are scarce. Consequently, patients with TNBC have a poorer overall prognosis compared to other breast cancer patients. Within heterogeneous tumors, individual clonal subpopulations may exhibit differences in their rates of growth and degrees of invasiveness. We hypothesized that such phenotypic heterogeneity at the single-cell level may accelerate tumor progression by enhancing the overall growth and invasion of the entire tumor. To test this hypothesis, we isolated and characterized clonal subpopulations with distinct morphologies and biomarker expression from the inherently heterogeneous 4T1 mouse mammary carcinoma cell line. We then leveraged a 3D microfluidic tumor model to reverse-engineer intratumoral heterogeneity and thus investigate how interactions between phenotypically distinct subpopulations affect tumor growth and invasion. We found that the growth and invasion of multiclonal tumors were largely dictated by the presence of cells with epithelial and mesenchymal traits, respectively. The latter accelerated overall tumor invasion, even when these cells comprised less than 1% of the initial population. Consistently, tumor progression was delayed by selectively targeting the mesenchymal subpopulation. This work reveals that highly invasive cells can dominate tumor phenotype and that specifically targeting these cells can slow the progression of heterogeneous tumors, which may help inform therapeutic approaches.
Article
Full-text available
The advent of iPSCs has brought about a significant transformation in stem cell research, opening up promising avenues for advancing cancer treatment. The formation of cancer is a multifaceted process influenced by genetic, epigenetic, and environmental factors. iPSCs offer a distinctive platform for investigating the origin of cancer, paving the way for novel approaches to cancer treatment, drug testing, and tailored medical interventions. This review article will provide an overview of the science behind iPSCs, the current limitations and challenges in iPSC-based cancer therapy, the ethical and social implications, and the comparative analysis with other stem cell types for cancer treatment. The article will also discuss the applications of iPSCs in tumorigenesis, the future of iPSCs in tumorigenesis research, and highlight successful case studies utilizing iPSCs in tumorigenesis research. The conclusion will summarize the advancements made in iPSC-based tumorigenesis research and the importance of continued investment in iPSC research to unlock the full potential of these cells.
Article
An immunosuppressive tumor microenvironment and tumor heterogeneity have led to the resilience of metastatic castrate resistant prostate cancer (mCRPC) to current treatments. To address these challenges, we developed and evaluated a new drug paradigm, Radio-IMmunostimulant (RIMS), in a syngeneic model of murine prostate cancer. RIMS-1 was generated using a convergent synthesis employing solid phase peptide and solution chemistries. The prostate-specific membrane antigen (PSMA) inhibitory constant for natLu-RIMS-1 was determined, and radiolabeling with 177Lu generated 177Lu-RIMS-1. The TLR 7/8 agonist payload release from natLu-RIMS-1 was determined using a cathepsin B assay. The biodistribution of 177Lu-RIMS-1 was evaluated in a bilateral xenograft model in NCru nude mice bearing PSMA(+) (PC3-PiP) and PSMA(-) (PC3-Flu) tumors at 2, 24, and 72 h. The therapeutic effect of 177Lu-RIMS-1 was evaluated in C57BL/6J mice bearing RM1-PGLS (PSMA-positive, green fluorescent protein-positive, and luciferase-positive) tumors and compared to that of 177Lu-PSMA-617 at the same total administered radioactivity of 57 MBq and molar activity of 5.18 MBq/nmol. natLu-RIMS-1 and vehicle were evaluated as the controls. Immuno-positron emission tomography (PET) using 89Zr-DFO-anti-CD3 was used to visualize T-cell distribution during treatment. 177Lu-RIMS-1 was quantitatively radiolabeled at >99% radiochemical purity and maintained a high affinity toward PSMA (Ki = 3.77 ± 0.5 nM). Cathepsin B efficiently released the entire immunostimulant payload in 17.6 h. 177Lu-RIMS-1 displayed a sustained uptake in PSMA(+) tumor tissue up to 72 h (2.65 ± 1.03% ID/g) and was not statistically different (P = 0.1936) compared to 177Lu-PSMA-617 (3.65 ± 0.59% ID/g). All animals treated with 177Lu-RIMS-1 displayed tumor growth suppression and provided a median survival of 30 days (P = 0.0007) while 177Lu-PSMA-617 provided a median survival of 15 days, which was not statistically significant (P = 0.3548) compared to the vehicle group (14 days). ImmunoPET analysis revealed 2-fold more tumor infiltrating T-cells in 177Lu-RIMS-1-treated animals compared to 177Lu-PSMA-617-treated animals; 177Lu-RIMS-1 improves therapeutic outcomes in a syngeneic model of mouse prostate cancer and elicits greater T-cell infiltration to the tumor compared to 177Lu-PSMA-617. These results support further investigation of the RIMS paradigm as the first example of a single molecular entity combining radiotherapy and immunostimulation.
Article
Full-text available
Recent studies have revealed extensive genetic diversity both between and within tumours. This heterogeneity affects key cancer pathways, driving phenotypic variation, and poses a significant challenge to personalized cancer medicine. A major cause of genetic heterogeneity in cancer is genomic instability. This instability leads to an increased mutation rate and can shape the evolution of the cancer genome through a plethora of mechanisms. By understanding these mechanisms we can gain insight into the common pathways of tumour evolution that could support the development of future therapeutic strategies.
Article
Full-text available
Alterations in cancer genomes strongly influence clinical responses to treatment and in many instances are potent biomarkers for response to drugs. The Genomics of Drug Sensitivity in Cancer (GDSC) database (www.cancerRxgene.org) is the largest public resource for information on drug sensitivity in cancer cells and molecular markers of drug response. Data are freely available without restriction. GDSC currently contains drug sensitivity data for almost 75 000 experiments, describing response to 138 anticancer drugs across almost 700 cancer cell lines. To identify molecular markers of drug response, cell line drug sensitivity data are integrated with large genomic datasets obtained from the Catalogue of Somatic Mutations in Cancer database, including information on somatic mutations in cancer genes, gene amplification and deletion, tissue type and transcriptional data. Analysis of GDSC data is through a web portal focused on identifying molecular biomarkers of drug sensitivity based on queries of specific anticancer drugs or cancer genes. Graphical representations of the data are used throughout with links to related resources and all datasets are fully downloadable. GDSC provides a unique resource incorporating large drug sensitivity and genomic datasets to facilitate the discovery of new therapeutic biomarkers for cancer therapies.
Article
Full-text available
The systematic translation of cancer genomic data into knowledge of tumour biology and therapeutic possibilities remains challenging. Such efforts should be greatly aided by robust preclinical model systems that reflect the genomic diversity of human cancers and for which detailed genetic and pharmacological annotation is available. Here we describe the Cancer Cell Line Encyclopedia (CCLE): a compilation of gene expression, chromosomal copy number and massively parallel sequencing data from 947 human cancer cell lines. When coupled with pharmacological profiles for 24 anticancer drugs across 479 of the cell lines, this collection allowed identification of genetic, lineage, and gene-expression-based predictors of drug sensitivity. In addition to known predictors, we found that plasma cell lineage correlated with sensitivity to IGF1 receptor inhibitors; AHR expression was associated with MEK inhibitor efficacy in NRAS-mutant lines; and SLFN11 expression predicted sensitivity to topoisomerase inhibitors. Together, our results indicate that large, annotated cell-line collections may help to enable preclinical stratification schemata for anticancer agents. The generation of genetic predictions of drug response in the preclinical setting and their incorporation into cancer clinical trial design could speed the emergence of 'personalized' therapeutic regimens.
Article
The systematic translation of cancer genomic data into knowledge of tumour biology and therapeutic possibilities remains challenging. Such efforts should be greatly aided by robust preclinical model systems that reflect the genomic diversity of human cancers and for which detailed genetic and pharmacological annotation is available¹. Here we describe the Cancer Cell Line Encyclopedia (CCLE): a compilation of gene expression, chromosomal copy number and massively parallel sequencing data from 947 human cancer cell lines. When coupled with pharmacological profiles for 24 anticancer drugs across 479 of the cell lines, this collection allowed identification of genetic, lineage, and gene-expression-based predictors of drug sensitivity. In addition to known predictors, we found that plasma cell lineage correlated with sensitivity to IGF1 receptor inhibitors; AHR expression was associated with MEK inhibitor efficacy in NRAS-mutant lines; and SLFN11 expression predicted sensitivity to topoisomerase inhibitors. Together, our results indicate that large, annotated cell-line collections may help to enable preclinical stratification schemata for anticancer agents. The generation of genetic predictions of drug response in the preclinical setting and their incorporation into cancer clinical trial design could speed the emergence of ‘personalized’ therapeutic regimens².
Article
Unlabelled: Recent tumor sequencing data suggest an urgent need to develop a methodology to directly address intratumoral heterogeneity in the design of anticancer treatment regimens. We use RNA interference to model heterogeneous tumors, and demonstrate successful validation of computational predictions for how optimized drug combinations can yield superior effects on these tumors both in vitro and in vivo. Importantly, we discover here that for many such tumors knowledge of the predominant subpopulation is insufficient for determining the best drug combination. Surprisingly, in some cases, the optimal drug combination does not include drugs that would treat any particular subpopulation most effectively, challenging straightforward intuition. We confirm examples of such a case with survival studies in a murine preclinical lymphoma model. Altogether, our approach provides new insights about design principles for combination therapy in the context of intratumoral diversity, data that should inform the development of drug regimens superior for complex tumors. Significance: This study provides the first example of how combination drug regimens, using existing chemotherapies, can be rationally designed to maximize tumor cell death, while minimizing the outgrowth of clonal subpopulations.
Article
Combination chemotherapies have been a mainstay in the treatment of disseminated malignancies for almost 60 y, yet even successful regimens fail to cure many patients. Although their single-drug components are well studied, the mechanisms by which drugs work together in clinical combination regimens are poorly understood. Here, we combine RNAi-based functional signatures with complementary informatics tools to examine drug combinations. This approach seeks to bring to combination therapy what the knowledge of biochemical targets has brought to single-drug therapy and creates a statistical and experimental definition of "combination drug mechanisms of action." We show that certain synergistic drug combinations may act as a more potent version of a single drug. Conversely, unlike these highly synergistic combinations, most drugs average extant single-drug variations in therapeutic response. When combined to form multidrug regimens, averaging combinations form averaging regimens that homogenize genetic variation in mouse models of cancer and in clinical genomics datasets. We suggest surprisingly simple and predictable combination mechanisms of action that are independent of biochemical mechanism and have implications for biomarker discovery as well as for the development of regimens with defined genetic dependencies.
Article
In a period of a little over 20 years, chemotherapy of cancer has evolved from a period of empiricism with little impact on the cancer problem to become part of a sound medical discipline with firm scientific underpinning playing an increasingly important role in the control of cancer. This progress has come from an increasing knowledge of cancer biology and pharmacology and the application of this knowledge to improved design of clinical trials, with due consideration to the intricacies of the natural history of each disease in question. Now that the chemotherapeutic tools are sharpened, their use in combinations with other modalities in the previously unfamiliar setting of the patient with early stages of the disease promises to lead to an even more exciting chapter in clinical cancer research in the next decade.
Article
Drug attrition rates for cancer are much higher than in other therapeutic areas. Only 5% of agents that have anticancer activity in preclinical development are licensed after demonstrating sufficient efficacy in phase III testing, which is much lower than, for example, 20% for cardiovascular disease. To compound this issue, many new cancer agents are being withdrawn, suspended or discontinued. Figure 1 illustrates that this trend is extremely prevalent for VEGF inhibitors although less so for drugs targeting Aurora B kinase and some targeted therapies. The reasons for this high attrition rate are complex; however, several articles in this issue provide insights into why this is occurring. In essence, the preclinical strategies to evaluate novel agents are suboptimal, and identifying the correct target using appropriate preclinical models will be critical to prevent further drug failures.