Accepted Article

Received Date : 30-Jan-2014 Revised Date : 20-Feb-2014 Accepted Date : 20-Feb-2014 Article type

: Key Symposium

The challenge of intratumour heterogeneity in precision medicine

Running head: Tumour heterogeneity and precision medicine

Joan Seoane1,2,3 & Leticia De Mattos-Arruda1,2

From the

1

Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital,

Barcelona, Spain; 2Universitat Autònoma de Barcelona, Barcelona, Spain; and 3Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain

Abstract Cells within tumours have diverse genomes and epigenomes, and interact differentially with their surrounding microenvironment generating intratumour heterogeneity, which has critical implications for treating cancer patients. Understanding the cellular and microenvironment composition and characteristics in individual tumours is critical to stratify the patient This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1111/joim.12240 This article is protected by copyright. All rights reserved.

Accepted Article

population that is likely to benefit from specific treatment regimens. Here, we will review the current understanding of intratumour heterogeneity at the genomic, epigenomic and microenvironmental levels. We will also discuss the clinical implications and the challenges posed by intratumour heterogeneity, and evaluate non-invasive methods such as circulating biomarkers to characterise the cellular diversity of tumours. Comprehensive assessment of the molecular features of patients based on tumour specimen characterisation (including intratumour spatial and temporal variations), ancillary non-invasive methods (such as circulating biomarkers and molecular imaging approaches), and the correct design of clinical trials are required to guide administration of targeted therapy and to control therapeutic resistance. Finding the means to accurately determine and effectively control tumour heterogeneity and translate these achievements into patient benefit are major goals in modern oncology.

Keywords: cancer, genomics, intratumour heterogeneity, precision medicine, targeted therapy.

Introduction Progress in cancer management has frequently resulted from the interaction between clinical and laboratory research [1, 2]. The molecular characterisation of tumours provides a basis for personalising cancer treatment resulting in increased clinical benefit, reduced toxicity and overcoming therapeutic resistance [3].

This article is protected by copyright. All rights reserved.

Accepted Article

The genomic characterisation of cancers has contributed to our understanding of the molecular landscape of tumours and has provided a rationale for the use of matched targeted therapies based on specific molecular targets. Improved treatment for specific tumour subtypes has highlighted the potential of precision medicine [3]; for example, the use of trastuzumab in HER2-positive breast cancer [4], vemurafenib for advanced BRAFV600-mutant melanoma [5], the PARP inhibitor olaparib in patients whose tumours bear BRCA1/2 mutations [6] and crizotinib in non-small-cell lung cancer with anaplastic lymphoma kinase translocations [7].

However, at present, precision medicine is not entirely successful. There is growing recognition that the heterogenous nature of cancer, and in particular the intrinsic heterogeneity of individual tumours as well as between primary and metastatic tumours, presents an important challenge in cancer therapeutics. In addition to the fact that all tumours show a unique combination of genomic alterations (intertumour heterogeneity), they are composed of cells with different molecular characteristics (intratumour heterogeneity). This results in diversity between and within tumours [8, 9].

Here, we review the current knowledge of the diverse patterns of intratumour heterogeneity. Cells within tumours can comprise distinct genomic alterations. In addition, cells with the same genomic landscape can show different states of differentiation and, hence, epigenetic status. Moreover, tumour cells can be influenced by a complex and diverse environment generated by stromal cell heterogeneity. These three different levels of heterogeneity are crucial for explaining the biology of cancer and have critical therapeutic implications.

This article is protected by copyright. All rights reserved.

Accepted Article

Understanding intratumoural heterogeneity is essential for designing effective therapeutic strategies in the context of precision medicine.

Tumour heterogeneity Although tumour heterogeneity is a well- and long-established concept, recognition of its various challenges to cancer therapy has increased in recent years [10–20] (Table 1). The study of cancer genomics has been boosted by the development and use of massively parallel (or next-generation) sequencing technologies [21]. Global research initiatives such as the 1000 Genomes Project [22], the Cancer Genome Atlas [23–25] and the International Cancer Genome Consortium [26], and publicly available catalogues such as the Catalogue of Somatic Mutations in Cancer) [27] and Genomics of Drug Sensitivity in Cancer [28], have added to improved delineation of cancer genomics. Together, these demonstrate variability among individual tumours and how diverse genomic aberrations of tumours sharing a similar histology is crucial for predicting how patients will respond to targeted therapies [8, 9] (Fig. 1).

In addition to intertumour heterogeneity, another layer of complexity is created by the intermingling of diverse subpopulations of cancer cells with normal cells, which leads to intratumour heterogeneity that can progress and become modified over time. Intratumour heterogeneity influences the dynamic tumour landscape and plays a key role in shaping responses to specific therapies [29, 30].

This article is protected by copyright. All rights reserved.

Accepted Article

Intratumour genomic heterogeneity The findings of massively parallel sequencing studies have demonstrated that the compendium of genomic aberrations in cancer is complex and diverse and only a limited number of genes are frequently and recurrently mutated in a substantial proportion of cancer cases [31–33]. Genomic analyses of human cancers have provided evidence of spatial and temporal intratumour genomic heterogeneity [11, 14, 34–36], and have shown that tumours are composed of mosaics of cells [12, 36] with subclones harbouring both private (or unique) and ubiquitous (i.e. common to all tumour cells) genomic alterations. For example, Gerlinger et al. investigated intratumour genomic heterogeneity in renal cancer through an analysis of genomic alterations in multiple tumour regions [11]. Using exome sequencing, chromosome aberration and DNA ploidy analyses confirmed the heterogeneity of different regions within the same tumour mass and of the primary tumour versus matched metastases. Genomic aberrations were found in all cell subclones throughout individual tumour specimens, whereas other aberrations were heterogeneously distributed [11]. In this context, it was shown that performing single biopsies of primary tumours or metastatic deposits, as is usual clinical practice, is unlikely to reveal the complete profile of genomic alterations in any tumour [37].

Additionally, recent evidence shows that intratumoural heterogeneity is dynamic and versatile [14, 18, 38, 39]. For example, Nik-Zainal et al. studied the evolution of cancer genomic clones and quantified the extent and dynamics of subclonal variation in breast cancer [18]. Moreover, while reconstructing tumour phylogeny through a multisampling scheme devised for individual glioblastoma patients, Sottoriva et al. constructed a model that inferred tumour progression in time and space and showed that copy number aberrations in

This article is protected by copyright. All rights reserved.

Accepted Article

EGFR and CDKN2A/B/p14ARF were early events, and aberrations in PDGFRA and PTEN occurred later during cancer progression [14].

Evolutionary studies have focused on analyses of the relationship between primary tumours and metastatic deposits, and within and between metastatic deposits [10, 12, 14, 36], as well as subclone evolution within a given tumour mass [15, 18, 40]. Increasing evidence demonstrates that genome evolution reflects the Darwinian model and branched evolutionary tumour growth contributes to intratumour genomic heterogeneity [10, 37, 38]. In this regard, the fittest clones [11, 41] are selected during the metastatic process via external selective pressures (e.g. administration of systemic therapeutics) [10] or via pressures from the tumour microenvironment (e.g. hypoxia, growth factors) [30] (Fig. 2).

The functional impact of intratumour genomic heterogeneity on cancer remains to be fully understood. Further studies are required to assess how intratumour genomic diversity influences the response and resistance to therapies during tumour relapse and the generation of metastasis.

Heterogeneity in the cellular state of differentiation Diverse states of differentiation and, hence, diverse epigenomic states can be found in cancer cells that share the same genomic alterations [42]. This implies that tumour deposits with homogeneous genomic characteristics contain functionally heterogeneous cancer cells with

This article is protected by copyright. All rights reserved.

Accepted Article

diverse phenotypes, including different capacities to proliferate, migrate and invade and, most importantly, with different sensitivities to therapeutic agents.

With regard to the concept of heterogeneity in the cellular state of differentiation, recent evidence from transplantation and lineage tracing experiments shows that only a fraction of cells within a tumour have tumour-initiating capabilities [43, 44]. These cells, termed cancerinitiating cells (CICs) or cancer stem cells, are more tumorigenic than their surrounding cells. CICs are usually poorly differentiated (with stem cell features), have a different epigenetic status compared to the rest of the cells and are considered to be tumour drivers [44, 45]. CICs are responsible for the re-initiation of tumours, mediating relapse and metastatic dissemination; moreover, these cells seem to be more resistant to DNA-damaging therapeutic agents [46, 47] (Fig. 2).

The intratumoural heterogeneity generated through the different degrees of cellular differentiation, including CICs and non-CICs, is due to epigenetic regulation of cells guided by signals from the microenvironment [42, 46–48]. Of note, CICs are not randomly spread through the tumour mass; these cells tend to be found in specific locations or niches, depending on the local microenvironment [49, 50]. Such niches are composed of non-tumour cells (i.e. inflammatory cells, endothelial cells and fibroblasts) and the extracellular matrix, which enable direct cell-to-cell interactions and secrete factors that maintain the stem cells in a quiescent state, regulating their self-renewal capacity and pluripotency. CICs can be located near vessels in a perivascular niche or in a hypoxic niche, suggesting close communication between CICs and the tumour microenvironment. Several signalling pathways, including the Wnt, Notch, Hedgehog and transforming growth factor-beta pathways, have been implicated This article is protected by copyright. All rights reserved.

Accepted Article

in the regulation of CIC self-renewal, and also on differentiation or dedifferentiation of nonCICs into CICs [45, 51, 52].

Of note, CICs and non-CICs differ in their sensitivity to treatment [47] and, therefore, unravelling the mechanism(s) underlying their epigenetic heterogeneity will be crucial for cancer treatment.

Intratumour microenvironment heterogeneity Tumour cells are in intimate contact with a heterogeneous microenvironment which regulates their biology and sensitivity to treatment [53]. The tumour niche is created by a dynamic microenvironment that supports and interacts with tumour cells through a complex signal cross-talk. The non-tumoural component of cancer, the tumour stroma, is composed primarily of the extracellular matrix, fibroblasts, blood vessels and immune cells [54]. These stromal elements can vary substantially within the topography of primary tumours and/or metastatic deposits, both in terms of the number of cell types and the proportion of lineage relationships [55], generating intratumour heterogeneity. Of importance, the normal cells of the stroma coevolve with tumour cells in a dynamic way; thus, tumour deposits should be evaluated in the context of an evolving tumour microenvironment which has definite implications for the response and resistance to therapeutic agents [54].

The tumour stroma comprises a modified extracellular matrix [56] with a large number of fibroblasts with specific characteristics [57]. The so-called cancer-associated fibroblasts

This article is protected by copyright. All rights reserved.

Accepted Article

(CAFs) are not evenly distributed within the extracellular matrix and play a key role in tumorigenesis [58]. In addition, CAFs and tumour cells can generate an intricate and dense extracellular matrix, which contributes to increased interstitial fluid pressure that hampers the penetration of drugs through tissue [59, 60]. This limits the diffusion of therapeutic compounds to tumour regions, and thus might represent an obstacle to clinical treatment.

Tumours are nourished by a dynamic vascular network, which controls tumour growth [61]. The tumour vasculature is variable with distinct characteristics even within the same tumour mass [30]. The vascular heterogeneity within tumours may result from the formation of new vessels (angiogenesis) or modification of existing vessels within the tissue [57]. In some parts of a tumour, abnormalities in blood vessels and differences in hydrostatic pressure impair blood flow, delivery of nutrients and clearance of metabolic products leading to acidic and hypoxic tumour regions [62]. This generates a variable milieu for tumour cells ranging from hypoxic to well-irrigated and non-hypoxic regions.

On the other hand, the inflammatory and immune components of the stroma can also vary in different locations of the tumour mass [30]. Tumour-infiltrating immune cells include varied cell types with specific roles both as pro- and anti-tumour cells; in addition, their geographic location can vary within individual tumours. This intratumoural spatial diversity of immune response and inflammatory cells controls the tumour biology, and its understanding can be critical for designing therapies based on the cancer immune checkpoint [29, 63].

This article is protected by copyright. All rights reserved.

Accepted Article

Tackling tumour heterogeneity in the clinic Cancer heterogeneity challenges the potential benefit of precision medicine. Consequently, genomic and non-genomic biomarker analyses of single biopsies may differ according to the area of the sampled tumour [11]. This multiregional separation of molecular aberrations can lead to sampling bias [37, 64], potentially impairing the interpretation of the molecular characterisation of tumours and having an impact on the selection of treatment.

Accordingly, approaches that provide global assessment of the catalogue of somatic genomic and epigenomic aberrations and tumour microenvironment-based biomarkers are clearly important for accurately selecting targeted therapies for individual patients. Recent studies in the metastatic setting have provided evidence of the emergence of a small number of subclones within primary tumours [34, 35, 65, 66]. These subclones could clarify how treatment-resistant subpopulations of tumour cells can be monitored and targeted over time. Pilot initiatives (i.e. molecularly driven clinical trials) [9, 67] and also circulating biomarkers [i.e. liquid biopsies, circulating tumour cells (CTCs) or cell-free tumour DNA (ctDNA)] [67, 68] may play a key role in this context. Pilot initiatives The purpose of using biomarkers to provide information for clinical decision-making is to enable individualised cancer therapy. The fact that there are different levels of heterogeneity (i.e. inter- and intratumour heterogeneity) is one the major problems for the administration of targeted therapies and has influenced the design of translational pilot initiatives and other innovative clinical trials.

This article is protected by copyright. All rights reserved.

Accepted Article

To date, most pilot initiatives have focused on individual patients through identifying groups of patients who share similar targetable biomarkers, and personalising appropriate targeted therapies to these individuals (Table 2) [45, 67, 69–77]. An example is the WinTHER study, which is a Phase II clinical trial launched by the Worldwide Innovative Networking Consortium in personalized cancer medicine (WIN Consortium) to assess the potential of selecting targeted therapies for ‘actionable’ targets found in the analysis of patients’ tumours [74, 78]. By contrast, few other initiatives (e.g. MOSCATO 01 study, NCT01566019) that included subgroups of patients enriched for specific genomic alterations have used the stratification approach, which focuses on molecular biomarkers rather than individual patients.

In addition, single subject clinical trials (i.e. n-of-1 trial design) investigate the efficacy of a given therapy or the side effect profiles of different therapeutic interventions in an individualised manner, primarily for patients harbouring rare molecular aberrations [9, 67]. Clinical trials using other designs are also being conducted, such as the ‘basket trials’ in which one study (the basket) includes one or more targets (e.g. NCT01219699 study enrolling patients whose tumours have a PIK3CA oncogene mutation to receive an alpha phosphatidylinositol 3-kinase (PI3K) inhibitor) and allows patients with multiple diseases to be

enrolled [11].

These studies to empirically test the concept of precision medicine are transforming the way tumour tissue is assessed for molecular aberrations, the design of clinical trials and the measurement of treatment efficacy. To date, these studies have usually been based on ‘biased‘ tumour tissue sampling, which relies on spatial variability in the expression of key This article is protected by copyright. All rights reserved.

Accepted Article

molecular biomarkers and, in some instances, on archival tumour specimens. This approach, which is rapidly becoming outdated, does not appropriately reflect tumour heterogeneity or dynamics.

Recent clinical and translational programmes have attempted to investigate intra- or intertumour heterogeneity (Table 3). These initiatives together with other non-invasive methods based on tumour molecular imaging [79, 80], which might identify and quantify actionable molecular biomarkers over time, are also leading the way to patient selection and longitudinal monitoring of response to therapy.

Circulating biomarkers The use of circulating biomarkers, in particular CTCs and ctDNA, is a promising approach that may illuminate the challenges posed by intratumour heterogeneity and sampling bias of single biopsies [64, 81–83]. CTCs and plasma-derived ctDNA have been widely investigated as potential non-invasive surrogates for tumour tissue biopsies [64]. The genomic characterisation of these blood-borne biomarkers has introduced a new method for investigating the metastatic process and the mechanisms of therapeutic resistance as well as for disease monitoring [65, 66, 82–85]. The ability to comprehensively identify tumour clones and subclones from cancer patients is likely to be instrumental in delineating the molecular and clinical heterogeneity of cancer and may facilitate the precise delivery of targeted therapy.

This article is protected by copyright. All rights reserved.

Accepted Article

CTCs and ctDNA are shed into the circulation from primary tumours and/or their metastases; therefore they constitute a potential source of tumour material from all disease sites, and offer a real-time, easily obtainable, cost-effective and minimally invasive tool for the development of molecular biomarkers [64, 68]. It should be noted, however, that CTCs may represent heterogeneous subpopulations of cancer cells [81, 86, 87] so that plasma-derived ctDNA might provide a better readout for bridging the gap between sample bias and the administration of targeted therapies. To this end, genomic characterisation of plasma-derived ctDNA may offer a way to confront the emergence of treatment-resistant clones, to track the evolution of the intratumour heterogeneity of cancer and to predict responses and resistance to treatment.

Conclusions and future perspectives Tumour diversity poses a challenge for managing the treatment of cancer patients. Decoding heterogeneity has important implications that will probably refine our understanding of cancer biology, its genomic, epigenomic and functional diversity and the mechanisms that lead to therapeutic resistance [10, 37, 88–90]. Nevertheless, the extent of tumour heterogeneity, and its multifaceted nature, remains poorly understood.

In addition to tumour heterogeneity, the design of studies appropriate for the era of precision medicine may be impeded by other barriers. These include the complexity of identifying and validating predictive molecular biomarkers, technical limitations of molecular tests, slow progress in unravelling the biology of some types of cancer, mechanisms of resistance per se,

This article is protected by copyright. All rights reserved.

Accepted Article

the high failure rate and cost of molecularly targeted agents and reimbursement and regulatory issues.

Current biomarker assessment programmes do not thoroughly address the issues of heterogeneity at the single-cell level (within and/or between patient tumours) or the dynamic environment of a cancer (i.e. changes in genomic, epigenomic and functional cancer characteristics). In addition, biomarker analysis does not consider minor genetic subclones or predict future tumour development. There is evidence to suggest that ’non-standard personalised strategies‘ using mathematic model-based genetic evolutionary dynamics and single-cell heterogeneity that personalise cancer therapeutics might lead to superior patient outcomes compared with the current model [91]. A comprehensive assessment of the molecular characteristics of patients and their therapeutic needs (Fig. 3), based on (i) tumour specimen characterisation, which includes intratumour spatial and temporal variations, (ii) ancillary non-invasive methods, such as circulating biomarkers and molecular imaging approaches, (iii) availability of clinical trials or compassionate use of specific therapeutic agents and (iv) functional characterisation of tumour-derived biomarkers based on in vivo cancer experimental models, would ideally guide targeted therapy administration and manage therapeutic resistance. Finding the means to accurately determine and effectively control tumour heterogeneity and translate these achievements into patient benefit are major goals in modern oncology. Precision medicine will not become a reality until heterogeneity-related issues are resolved.

Conflict of interest statement The authors have no conflicts of interest to declare. This article is protected by copyright. All rights reserved.

Accepted Article

Acknowledgments The authors acknowledge finantial support from Fundación Rafael del Pino and la Asociación Española Contra el Cáncer. References

1

Baselga J. Bringing precision medicine to the clinic: from genomic profiling to the

power of clinical observation. Ann Oncol 2013; 24: 1956-7.

2

Gazdar AF, Minna JD. Precision medicine for cancer patients: lessons learned and the

path forward. J Natl Cancer Inst 2013; 105: 1262-3.

3

Garraway LA, Verweij J, Ballman KV. Precision oncology: an overview. J Clin

Oncol 2013; 31: 1803-5.

4

Ross JS, Slodkowska EA, Symmans WF, Pusztai L, Ravdin PM, Hortobagyi GN. The

HER-2 receptor and breast cancer: ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist 2009; 14: 320-68.

5

Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in

melanoma with BRAF V600E mutation. N Engl J Med 2011; 364: 2507-16.

6

Fong PC, Boss DS, Yap TA, et al. Inhibition of poly(ADP-ribose) polymerase in

tumors from BRCA mutation carriers. N Engl J Med 2009; 361: 123-34.

7

Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in

non-small-cell lung cancer. N Engl J Med 2010; 363: 1693-703.

8

Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of

genetic heterogeneity in cancer evolution. Nature 2013; 501: 338-45.

This article is protected by copyright. All rights reserved.

Accepted Article

9

Bedard PL, Hansen AR, Ratain MJ, Siu LL. Tumour heterogeneity in the clinic.

Nature 2013; 501: 355-64.

10

Turner NC, Reis-Filho JS. Genetic heterogeneity and cancer drug resistance. Lancet

Oncol 2012; 13: e178-85.

11

Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and branched

evolution revealed by multiregion sequencing. N Engl J Med 2012; 366: 883-92.

12

Shah SP, Roth A, Goya R, et al. The clonal and mutational evolution spectrum of

primary triple-negative breast cancers. Nature 2012.

13

Yancovitz M, Litterman A, Yoon J, et al. Intra- and inter-tumor heterogeneity of

BRAF(V600E) mutations in primary and metastatic melanoma. PLoS One 2012; 7: e29336.

14

Sottoriva A, Spiteri I, Piccirillo SG, et al. Intratumor heterogeneity in human

glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci U S A 2013; 110: 4009-14.

15

Wu X, Northcott PA, Dubuc A, et al. Clonal selection drives genetic divergence of

metastatic medulloblastoma. Nature 2012; 482: 529-33.

16

Taniguchi K, Okami J, Kodama K, Higashiyama M, Kato K. Intratumor heterogeneity

of epidermal growth factor receptor mutations in lung cancer and its correlation to the response to gefitinib. CancerSci 2008; 99: 929-35.

17

Bashashati A, Ha G, Tone A, et al. Distinct evolutionary trajectories of primary high-

grade serous ovarian cancers revealed through spatial mutational profiling. J Pathol 2013; 231: 21-34.

This article is protected by copyright. All rights reserved.

Accepted Article

18

Nik-Zainal S, Van Loo P, Wedge DC, et al. The life history of 21 breast cancers. Cell

2012; 149: 994-1007.

19

Yachida S, Jones S, Bozic I, et al. Distant metastasis occurs late during the genetic

evolution of pancreatic cancer. Nature 2010; 467: 1114-7.

20

De Mattos-Arruda L, Bidard FC, Won HH, et al. Establishing the origin of metastatic

deposits in the setting of multiple primary malignancies: The role of massively parallel sequencing. Mol Oncol 2014;8:150-8.

21

Meyerson M, Gabriel S, Getz G. Advances in understanding cancer genomes through

second-generation sequencing. Nat Rev Genet 2010; 11: 685-96.

22

Mills RE, Walter K, Stewart C, et al. Mapping copy number variation by population-

scale genome sequencing. Nature 2011; 470: 59-65.

23

Comprehensive genomic characterization defines human glioblastoma genes and core

pathways. Nature 2008; 455: 1061-8.

24

Integrated genomic analyses of ovarian carcinoma. Nature 2011; 474: 609-15.

25

Comprehensive molecular portraits of human breast tumours. Nature 2012; 490: 61-

70.

26

Hudson TJ, Anderson W, Artez A, et al. International network of cancer genome

projects. Nature 2010; 464: 993-8.

27

Forbes SA, Bhamra G, Bamford S, et al. The Catalogue of Somatic Mutations in

Cancer (COSMIC). Curr Protoc Hum Genet 2008; Chapter 10: Unit 10 1.

This article is protected by copyright. All rights reserved.

Accepted Article

28

McDermott U, Sharma SV, Dowell L, et al. Identification of genotype-correlated

sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling. Proc Natl Acad Sci U S A 2007; 104: 19936-41.

29

Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human

tumours: impact on clinical outcome. Nat Rev Cancer 2012; 12: 298-306.

30

Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on

therapeutic response. Nature 2013; 501: 346-54.

31

Collisson EA, Cho RJ, Gray JW. What are we learning from the cancer genome? Nat

Rev Clin Oncol 2012; 9: 621-30.

32

Banerji S, Cibulskis K, Rangel-Escareno C, et al. Sequence analysis of mutations and

translocations across breast cancer subtypes. Nature 2012; 486: 405-9.

33

Curtis C, Shah SP, Chin SF, et al. The genomic and transcriptomic architecture of

2,000 breast tumours reveals novel subgroups. Nature 2012; 486: 346-52.

34

Shah SP, Roth A, Goya R, et al. The clonal and mutational evolution spectrum of

primary triple-negative breast cancers. Nature 2012; 486: 395-9.

35

Ding L, Ellis MJ, Li S, et al. Genome remodelling in a basal-like breast cancer

metastasis and xenograft. Nature 2010; 464: 999-1005.

36

Navin N, Kendall J, Troge J, et al. Tumour evolution inferred by single-cell

sequencing. Nature 2011; 472: 90-4.

37

Swanton C. Intratumor heterogeneity: evolution through space and time. Cancer Res

2012; 72: 4875-82.

This article is protected by copyright. All rights reserved.

Accepted Article

38

Newburger DE, Kashef-Haghighi D, Weng Z, et al. Genome evolution during

progression to breast cancer. Genome Res 2013; 23: 1097-108.

39

Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in

human cancer. Nature 2013; 500: 415-21.

40

Maley CC, Galipeau PC, Finley JC, et al. Genetic clonal diversity predicts

progression to esophageal adenocarcinoma. Nat Genet 2006; 38: 468-73.

41

Fidler IJ. Tumor heterogeneity and the biology of cancer invasion and metastasis.

Cancer Res 1978; 38: 2651-60.

42

Magee JA, Piskounova E, Morrison SJ. Cancer stem cells: impact, heterogeneity, and

uncertainty. Cancer Cell 2012; 21: 283-96.

43

Wicha MS, Liu S, Dontu G. Cancer stem cells: an old idea--a paradigm shift. Cancer

Res 2006; 66: 1883-90; discussion 95-6.

44

Kakarala M, Wicha MS. Implications of the cancer stem-cell hypothesis for breast

cancer prevention and therapy. J Clin Oncol 2008; 26: 2813-20.

45

Anido J, Saez-Borderias A, Gonzalez-Junca A, et al. TGF-beta Receptor Inhibitors

Target the CD44(high)/Id1(high) Glioma-Initiating Cell Population in Human Glioblastoma. Cancer Cell 2010; 18: 655-68.

46

Dick JE. Stem cell concepts renew cancer research. Blood 2008; 112: 4793-807.

47

Shibata M, Shen MM. The roots of cancer: stem cells and the basis for tumor

heterogeneity. BioEssays : news and reviews in molecular, cellular and developmental biology 2013; 35: 253-60.

This article is protected by copyright. All rights reserved.

Accepted Article

48

Shackleton M, Quintana E, Fearon ER, Morrison SJ. Heterogeneity in cancer: cancer

stem cells versus clonal evolution. Cell 2009; 138: 822-9.

49

Ohlstein B, Kai T, Decotto E, Spradling A. The stem cell niche: theme and variations.

Curr Opin Cell Biol 2004; 16: 693-9.

50

Takakura N. Formation and regulation of the cancer stem cell niche. Cancer science

2012; 103: 1177-81.

51

Penuelas S, Anido J, Prieto-Sanchez RM, et al. TGF-beta increases glioma-initiating

cell self-renewal through the induction of LIF in human glioblastoma. Cancer Cell 2009; 15: 315-27.

52

Oshimori N, Fuchs E. The harmonies played by TGF-beta in stem cell biology. Cell

Stem Cell 2012; 11: 751-64.

53

Tlsty TD, Coussens LM. Tumor stroma and regulation of cancer development. Annu

Rev Pathol 2006; 1: 119-50.

54

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144:

646-74.

55

Freeman MR, Li Q, Chung LW. Can stroma reaction predict cancer lethality? Clin

Cancer Res 2013; 19: 4905-7.

56

Aznavoorian S, Stracke ML, Krutzsch H, Schiffmann E, Liotta LA. Signal

transduction for chemotaxis and haptotaxis by matrix molecules in tumor cells. J Cell Biol 1990; 110: 1427-38.

This article is protected by copyright. All rights reserved.

Accepted Article

57

Tredan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor

microenvironment. J Natl Cancer Inst 2007; 99: 1441-54.

58

Orimo A, Gupta PB, Sgroi DC, et al. Stromal fibroblasts present in invasive human

breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 2005; 121: 335-48.

59

Brekken C, Bruland OS, de Lange Davies C. Interstitial fluid pressure in human

osteosarcoma xenografts: significance of implantation site and the response to intratumoral injection of hyaluronidase. Anticancer Res 2000; 20: 3503-12.

60

Netti PA, Berk DA, Swartz MA, Grodzinsky AJ, Jain RK. Role of extracellular

matrix assembly in interstitial transport in solid tumors. Cancer Res 2000; 60: 2497-503.

61

Ferrara N, Kerbel RS. Angiogenesis as a therapeutic target. Nature 2005; 438: 967-

74.

62

Tatum JL, Kelloff GJ, Gillies RJ, et al. Hypoxia: importance in tumor biology,

noninvasive measurement by imaging, and value of its measurement in the management of cancer therapy. Int J Radiat Biol 2006; 82: 699-757.

63

Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in

patients with metastatic melanoma. N Engl J Med 2010; 363: 711-23.

64

De Mattos-Arruda L, Cortes J, Santarpia L, Vivancos A, Tabernero J, Reis-Filho JS,

Seoane J. Circulating tumour cells and cell-free DNA as tools for managing breast cancer. Nat Rev Clin Oncol 2013; 10: 377-89.

This article is protected by copyright. All rights reserved.

Accepted Article

65

Diaz LA, Jr., Williams RT, Wu J, et al. The molecular evolution of acquired

resistance to targeted EGFR blockade in colorectal cancers. Nature 2012; 486: 537-40.

66

Misale S, Yaeger R, Hobor S, et al. Emergence of KRAS mutations and acquired

resistance to anti-EGFR therapy in colorectal cancer. Nature 2012; 486: 532-6.

67

De Mattos-Arruda L, Rodon J. Pilot Studies for Personalized Cancer Medicine:

Focusing on the Patient for Treatment Selection. Oncologist 2013.

68

Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring

cancer-genetics in the blood. Nat Rev Clin Oncol 2013; 10: 472-84.

69

Bertotti A, Migliardi G, Galimi F, et al. A molecularly annotated platform of patient-

derived xenografts ("xenopatients") identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov 2011; 1: 508-23.

70

Hidalgo M, Bruckheimer E, Rajeshkumar NV, et al. A pilot clinical study of

treatment guided by personalized tumorgrafts in patients with advanced cancer. Mol Cancer Ther 2011; 10: 1311-6.

71

Von Hoff DD, Stephenson JJ, Jr., Rosen P, et al. Pilot study using molecular profiling

of patients' tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol 2010; 28: 4877-83.

72

Roychowdhury S, Iyer MK, Robinson DR, et al. Personalized oncology through

integrative high-throughput sequencing: a pilot study. Sci Transl Med 2011; 3: 111ra21.

This article is protected by copyright. All rights reserved.

Accepted Article

73

Tran B, Brown AM, Bedard PL, et al. Feasibility of real time next generation

sequencing of cancer genes linked to drug response: results from a clinical trial. Int J Cancer 2013; 132: 1547-55.

74

Mendelsohn J, Tursz T, Schilsky RL, Lazar V. WIN Consortium--challenges and

advances. Nat Rev Clin Oncol 2011; 8: 133-4.

75

Kim ES, Herbst RS, Wistuba, II, et al. The BATTLE trial: personalizing therapy for

lung cancer. Cancer Discov 2011; 1: 44-53.

76

Barker AD, Sigman CC, Kelloff GJ, Hylton NM, Berry DA, Esserman LJ. I-SPY 2:

an adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy. Clin Pharmacol Ther 2009; 86: 97-100.

77

Tsimberidou AM, Iskander NG, Hong DS, et al. Personalized medicine in a phase I

clinical trials program: the MD Anderson Cancer Center initiative. Clin Cancer Res 2012; 18: 6373-83.

78

Tursz T, Andre F, Lazar V, Lacroix L, Soria JC. Implications of personalized

medicine--perspective from a cancer center. Nat Rev Clin Oncol 2011; 8: 177-83.

79

Tolmachev V, Stone-Elander S, Orlova A. Radiolabelled receptor-tyrosine-kinase

targeting drugs for patient stratification and monitoring of therapy response: prospects and pitfalls. Lancet Oncol 2010; 11: 992-1000.

80

van Kruchten M, de Vries EG, Brown M, et al. PET imaging of oestrogen receptors in

patients with breast cancer. Lancet Oncol 2013; 14: e465-75.

This article is protected by copyright. All rights reserved.

Accepted Article

81

Powell AA, Talasaz AH, Zhang H, et al. Single cell profiling of circulating tumor

cells: transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS One 2012; 7: e33788.

82

Forshew T, Murtaza M, Parkinson C, et al. Noninvasive identification and monitoring

of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 2012; 4: 136ra68.

83

Dawson SJ, Rosenfeld N, Caldas C. Circulating tumor DNA to monitor metastatic

breast cancer. N Engl J Med 2013; 369: 93-4.

84

Murtaza M, Dawson SJ, Tsui DW, et al. Non-invasive analysis of acquired resistance

to cancer therapy by sequencing of plasma DNA. Nature 2013; 497: 108-12.

85

Zhang L, Ridgway LD, Wetzel MD, et al. The identification and characterization of

breast cancer CTCs competent for brain metastasis. Sci Transl Med 2013; 5: 180ra48.

86

Hayes DF, Paoletti C. Circulating tumour cells: insights into tumour heterogeneity. J

Intern Med 2013; 274: 137-43.

87

Gasch C, Bauernhofer T, Pichler M, et al. Heterogeneity of epidermal growth factor

receptor status and mutations of KRAS/PIK3CA in circulating tumor cells of patients with colorectal cancer. Clin Chem 2013; 59: 252-60.

88

Yap TA, Gerlinger M, Futreal PA, Pusztai L, Swanton C. Intratumor heterogeneity:

seeing the wood for the trees. Sci Transl Med 2012; 4: 127ps10.

89

Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for

cancer? Nat Rev Cancer 2012; 12: 323-34.

This article is protected by copyright. All rights reserved.

Accepted Article

90

Stephens PJ, Tarpey PS, Davies H, et al. The landscape of cancer genes and

mutational processes in breast cancer. Nature 2012; 486: 400-4.

91

Beckman RA, Schemmann GS, Yeang CH. Impact of genetic dynamics and single-

cell heterogeneity on development of nonstandard personalized medicine strategies for cancer. Proc Natl Acad Sci U S A 2012; 109: 14586-91.

Correspondence: Prof. Joan Seoane, Translational Research Program, Vall d’Hebron Institut d’Oncologia, Pg Vall d’Hebron 119-129 08035, Barcelona, Spain. Tel: +34 93 4894167; Fax: +34 93 489 40 15; E-mail: [email protected]

This article is protected by copyright. All rights reserved.

Accepted Article

Figure legends Fig. 1 The different patterns of tumour heterogeneity. (A) Intertumour heterogeneity: heterogeneity between tumours in different patients or within the same patient with different tumour deposits. (B) Intratumour heterogeneity: diversity within a tumour. (C) Intratumour genomic heterogeneity: cells with distinct genomic alterations within a tumour. (D) Intratumour epigenomic heterogeneity: cells with diverse states of differentiation based on different epigenomic states. (E) Intratumour microenvironment heterogeneity: differences in tumour stroma (extracellular matrix, vasculature and immune cells) within the same tumour. Fig. 2 Functional heterogeneity and the impact of treatment in the selection of the fittest cells/clones. (A) Genomic selection: the treatment affects a cellular clone with a specific genomic alteration. (B) Epigenomic selection: the treatment affects a cellular compartment with a specific state of differentiation/epigenetic status [e.g. non-cancer-initiating cells (CICs) vs. CICs]. (C) Microenvironment selection: the treatment affects cells present in a specific stromal niche. Fig. 3 Projected strategy of data integration to connect tumour characterisation and matched targeted therapy in the context of precision medicine. Clinical trial allocation would be based not only on the characterisation of tumours, which involves genomic/non-genomic aspects of tumour tissue, the use of surrogate tools (e.g. circulating blood-based biomarkers and molecular imaging) and functional studies, but would consider inter- and intratumour heterogeneity as core parameters for the specific choice of the most appropriate drug for each patient.

This article is protected by copyright. All rights reserved.

Accepted Article

Table 1 Selected examples of studies to assess tumour heterogeneity Tumour Description Type of assessment type Melanoma Intra- and intertumour heterogeneity of Intra- and intertumour heterogeneity the BRAFV600E mutation using BRAF mutant-specific PCR and conventional sequencing Renal Branched evolutionary growth through Intra- and intertumour multiregion exome sequencing heterogeneity Intratumour heterogeneity Glioblasto Integrated genomic analysis (copy ma number and gene expression) after multiple intratumour sample collection reveals multiple glioblastoma subtypes within the same tumour and inferred genomic evolution Medullobl Metastases from an individual are quite Intertumour heterogeneity astoma similar to each other but differ from the matched primary tumour Intratumour heterogeneity NSCLC Multiregion sampling from each tumour showed heterogeneous populations of both EGFR-mutated and non-mutated cancer cells, which was associated with reduced response to gefitinib Ovarian Intratumour variation in mutation, copy Intratumour heterogeneity number and gene expression profiles through the analysis of spatially and temporally separated high-grade serous ovarian tumour specimens. Key driver gene alterations (e.g. PIK3CA, CTNNB1 and NF1) present in a subset of samples; only TP53 somatic mutation present in all samples Intratumour heterogeneity Breast Triple-negative breast tumours vary widely in their clonal frequencies (e.g. TP53, PIK3CA and PTEN somatic mutations were not clonally dominant in all tumours) Breast The subclonal architecture of 21 breast Intratumour heterogeneity cancers was mapped by massively parallel sequencing analyses Pancreatic Whole-exome sequencing of pancreatic Intra- and intertumour cancer specimens showed the clonal heterogeneity relationships among primary and metastatic tumours. Timing of the genetic evolution (i.e. distant metastasis occurs late for these cancers) was inferred. This article is protected by copyright. All rights reserved.

Reference [13]

[11] [14]

[15]

[16]

[17]

[12]

[18]

[19]

Accepted Article

[20] Synchrono Massively parallel sequencing can help Intra- and intertumour define the origin of metastatic deposits heterogeneity us in patients with multiple primary tumours tumours and reveal genetic heterogeneity within and between lesions from the same patient EGFR, epidermal growth factor receptor, NSCLC, non-small-cell lung cancer; PCR, polymerase chain reaction.

Table 2 Selected pilot studies focusing on individual patients

Pilot study design

Matched actionable molecular alteration with targeted therapy

Treated patients

Reference

D

D

[71] [72]

D

D

D

D

D

D

D

D

D

D

D

D

D

D

D

D

Personalised Bisgrove trial MI-ONCOSEQ COMPACT WinTher

[73] [74]

Stratified MOSCATO 01 BATTLE I-SPY 2

The M. D. Anderson Cancer Center Initiative ‘Basket trials’

[75] [76] [77] -

Both personalised and stratified

Patient-derived xenografts

[45, 69, 70]

BATTLE, Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination; COMPACT, Canadian multicenter clinical trial; MI-ONCOSEQ, Michigan Oncology Sequencing Project; MOSCATO 01, The MOlecular Screening for CAncer Treatment and Optimisation; I-SPY 2; Investigation of Serial studies to Predict Your therapeutic response with imaging and molecular analysis; WinTHER, Worldwide Innovative Networking.

This article is protected by copyright. All rights reserved.

Accepted Article

Table 3 Selected studies that access tumour heterogeneity

Study NCT ID Tumour type Intratumour and inter-patient heterogeneity BEAUT Non-metastatic breast Y N/A cancer

Whole genome sequencing

TRACER NCT018 x 88601

Whole-genome and -exome sequencing

Xenograft models Circulating biomarker and functional imaging

MiSeq/Sequenom

Circulating biomarkers

Non-metastatic NSCLC Metastatic breast, colorectal or gynaecological cancer

NCT017 MATCH 03585 EPREDIC ISRCTN T/SPREDIC 2297960 Metastatic renal cell 4 cancer T Inter-patient heterogeneity

Main platform used

Ancillary approaches used

Whole exome sequencing, transcriptome

BATTLE -FL

NCT012 63782

Front-line treatment of NSCLC (stage IIIB or IV)

BATTLE -2

NCT012 48247

Metastatic NSCLC

NSCLC-based molecular biomarkers (PCR/FISH) NSCLC-based molecular biomarkers (PCR/FISH)

I-SPY 2

NCT010 42379

CanSeq

N/A

Breast cancer (stage III, neoadjuvant) NSCLC, colorectal, breast and prostate cancers

TargetPrint HER2, Mammaprint OncoMap/whole exome sequencing

-

-

Blood and breast magnetic resonance imaging -

BATTLE, Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination; BATTLE-FL: Front-Line Biomarker-Integrated Treatment Study in Non Small Cell Lung Cancer; BEAUTY, Breast Cancer Genome-Guided Therapy; FISH, fluorescence in situ hybridisation. HER2, Human Epidermal growth factor Receptor 2, I-SPY 2 TRIAL: Neoadjuvant and Personalized Adaptive Novel Agents to Treat Breast Cancer; ISRCTN, International Standard Randomised Controlled Trial Number; NCT ID, National

This article is protected by copyright. All rights reserved.

Accepted Article

Clinical Trials Identifier (ClinicalTrials.gov); MATCH, Feasibility Study of Genomic Profiling Methods and Timing in Tumor Samples; N/A, non available, NSCLC, non-smallcell lung cancer; PCR, polymerase chain reaction; PREDICT, Study of Preoperative Everolimus in Metastatic Renal Cancer; TRACERx, TRAcking Non-small Cell Lung Cancer Evolution Through Therapy.

This article is protected by copyright. All rights reserved.

Accepted Article This article is protected by copyright. All rights reserved.

Accepted Article This article is protected by copyright. All rights reserved.

The challenge of intratumour heterogeneity in precision medicine.

Cells within tumours have diverse genomes and epigenomes and interact differentially with their surrounding microenvironment generating intratumour he...
308KB Sizes 3 Downloads 3 Views