A population genetics perspective on the determinants of intra-tumor heterogeneity

https://doi.org/10.1016/j.bbcan.2017.03.001Get rights and content

Highlights

  • Population genetics principles provide insight into the forces that influence intra-tumor heterogeneity.

  • Distinct modes of evolution are operative during different stages of tumor progression and in different tumor types.

  • Spatial information obtained via multi-region sequencing can help to resolve tumor evolutionary dynamics.

  • A quantitative understanding of tumor dynamics may inform the detection and treatment of cancer.

Abstract

Cancer results from the acquisition of somatic alterations in a microevolutionary process that typically occurs over many years, much of which is occult. Understanding the evolutionary dynamics that are operative at different stages of progression in individual tumors might inform the earlier detection, diagnosis, and treatment of cancer. Although these processes cannot be directly observed, the resultant spatiotemporal patterns of genetic variation amongst tumor cells encode their evolutionary histories. Such intra-tumor heterogeneity is pervasive not only at the genomic level, but also at the transcriptomic, phenotypic, and cellular levels. Given the implications for precision medicine, the accurate quantification of heterogeneity within and between tumors has become a major focus of current research. In this review, we provide a population genetics perspective on the determinants of intra-tumor heterogeneity and approaches to quantify genetic diversity. We summarize evidence for different modes of evolution based on recent cancer genome sequencing studies and discuss emerging evolutionary strategies to therapeutically exploit tumor heterogeneity. This article is part of a Special Issue entitled: Evolutionary principles - heterogeneity in cancer?, edited by Dr. Robert A. Gatenby.

Introduction

Cancer results from the acquisition of alterations during somatic cell division in a microevolutionary process that typically occurs over decades, much of which is occult and with extended clinically latent intervals. For example, more than half of all detectable somatic mutations in colorectal cancers occur prior to transformation [1], [2]. A set of initiating (epi)genetic events (so called drivers) provide a selective fitness advantage (resulting in higher proliferation or lower apoptosis for example) in the target cell relative to other pre-malignant cells, resulting in clonal expansion. These initiating events in the founding tumor cell are often specific to certain tissue types or cells of origin as is the case for APC in colon and other gastrointestinal tumors [3], [4], [5] or DNMT3 in leukemias [6]. Hence, the fitness benefit that the mutation confers may depend on the microenvironment or cellular context in which it occurs. However, a single mutation is seldom sufficient to evoke a fully malignant phenotype and the pre-malignant clones likely maintain functionality until additional ‘hits’ accrue, eventually leading to transformation and uncontrolled cell growth (Fig. 1).

Given that tumor cells continuously accrue mutations and are subject to ever changing microenvironments, genetic and phenotypic heterogeneity is expected. Indeed, according to the clonal evolution theory, heterogeneity is attributed to continued genetic and heritable epigenetic alterations and selection in diverse microenvironments within tumors [7], [8], [9]. Hence mutation rates, genetic drift, population structure, microenvironment and selection impact the extent of tumor heterogeneity. As tumors progress, expanding into an environment that is resource limited, cells that are better able to replicate, utilize energy, and migrate will have a competitive advantage and these ‘hallmarks’ are noted in most advanced cancers [10]. At this stage, the initiating genetic lesions may no longer ensure cell survival. As such, it is essential to develop an understanding of the phenotypic consequences of putative oncogenes and tumor suppressors in a context-dependent fashion.

Although intra-tumor heterogeneity (ITH) has been appreciated for decades [11], the advent of high-throughput technologies has enabled the characterization of (epi)genomic, transcriptomic, phenotypic, and cellular heterogeneity at enhanced resolution [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25]. The presence of pervasive ITH poses significant challenges for precision medicine. For example, sampling bias due to solid tumor spatial structure within lesions can obscure the interpretation of genomic profiles for patient stratification and therapeutic decision-making. Elevated ITH may also be associated with disease progression [26] and poor prognosis [27], [28]. Given the numerous clinical implications, the accurate quantification of heterogeneity within and between tumors from the same patient is a major focus of current research. However, to date, these analyses have largely been descriptive in nature.

While human tumor evolution cannot be directly observed, spatiotemporal patterns of genetic variation amongst tumor cells are stably inherited during cell division, and hence surreptitiously encode their ancestries. As such, the resultant patterns of ITH can be ‘read’ via multi-region sequencing (MRS) of spatially and/or temporally separated tumor regions or single-cell sequencing (SCS) and used to reconstruct the evolutionary relationships amongst the distinct cell populations (clones) that comprise a tumor, as has now been performed in a variety of tumor types [13], [16], [17], [18], [19], [21], [22], [25], [29], [30], [31], [32], [33], [34]. Intriguingly, these and other cancer sequencing studies suggest that distinct modes of evolution are operative during tumor progression and in different tumor types. However, this has yet to be systematically evaluated. More generally, while the elements of somatic evolution have been defined [7], [8], [35] and include mutation, genetic drift, migration, population structure, and selection, the evolutionary dynamics that are operative at different stages of progression in individual tumors are poorly characterized. A quantitative understanding of tumor dynamics has the potential to define cancers' evolutionary playbook and might hold clues to as to how to better prevent, detect, and treat cancers. For example, the earlier the initial clonal expansion is detected, the less diverse and likely less fit the cell population, and the better the prognosis. Hence, knowledge of the initiating events and their relative temporal ordering may inform strategies for earlier intervention. Likewise, computational and mathematical modeling can provide insights into mechanisms of progression and enable the inference of patient-specific tumor dynamics. Such information may ultimately inform the design of rational treatment strategies.

In this review, we provide a population genetic perspective on the origins of tumor heterogeneity and summarize approaches to quantify genetic diversity, drawing from established theory for studying genetic variation within and among natural populations in light of the forces of mutation, genetic drift, selection, and demography. We summarize evidence for different modes of clonal evolution based on recent cancer genome sequencing studies and the potential clinical relevance of the resultant heterogeneity. Finally, we discuss emerging evolutionary strategies to therapeutically exploit tumor heterogeneity and steer clonal dynamics.

Section snippets

Genomic heterogeneity within and between tumors

Conventionally, the distinct stages of tumor initiation and subsequent growth have been assumed to proceed in a ‘linear’ sequential fashion in which successively more fit ‘driver’ events arise within a clone, resulting in the replacement or clonal succession of less fit clones through selective sweeps [8], [36]. While the sequential model may accurately describe tumor initiation, it is not known whether this applies to most solid tumors. Sequencing of established primary tumors has revealed

Practical challenges in quantifying ITH in tumor samples

The quantification of ITH from current NGS data is challenging for multiple reasons. From a practical perspective, the power to detect subclonal somatic single nucleotide variants (SSNVs) is limited in current bulk tumor WES studies, which often aim to achieve 80–100 × coverage (with WGS often lower) [84]. This is particularly problematic when coupled with the low purity of many tumor samples, which contain a heterogeneous mixture of non-cancerous and cancerous cells. Ideally purity is estimated

Evolutionary forces determining the extent of genetic heterogeneity

Mutation, genetic drift, migration and selection are the fundamental forces that collectively shape genetic diversity within a population [185]. Clonal evolution, the process of mutation accumulation and adaptation in somatic cells, likely involves each of these forces, although their collective impact on ITH is seldom considered. Here we discuss these factors in the context of classical population genetic theory and recent cancer genomic data.

Understanding, predicting and preventing tumor progression

Although tumor growth cannot be directly observed, patterns of genetic diversity present in a lesion reflect the evolutionary forces that gave rise to it. It is therefore not surprising that ITH since a tumor's future behavior depends on its evolutionary course. In theory, the earlier the initial clonal expansion is detected, the less diverse and less fit the cell population (since natural selection operates on diversity), and the better the prognosis. Even relatively early tumors exhibit high

Conclusions

In this review, we have outlined some of the recent advances in characterizing ITH, as well as technical and methodological challenges in quantifying and interpreting these patterns. Although much focus to date has been on genotypic heterogeneity, emergent techniques may facilitate the characterization of heterogeneity in situ [281]. In parallel, functional heterogeneity amongst tumor cells can be assessed in patient-derived xenograft [282], [283] or organoid [284], [285] models. Moreover,

Transparency document

Transparency document.

Acknowledgements

This work was funded by awards from the NIH (R01CA182514), Susan G. Komen Foundation (IIR13260750), and the Breast Cancer Research Foundation (BCRF-16-032) to C.C. Z.H. is supported by an Innovative Genomics Initiative (IGI) Postdoctoral Fellowship.

References (286)

  • K. Rowald

    Negative selection and chromosome instability induced by Mad2 overexpression delay breast cancer but facilitate oncogene-independent outgrowth

    Cell Rep.

    (2016)
  • E.J. Fox et al.

    Lethal mutagenesis: targeting the mutator phenotype in cancer

    Semin. Cancer Biol.

    (2010)
  • B.A. Weaver

    Aneuploidy acts both oncogenically and as a tumor suppressor

    Cancer Cell

    (2007)
  • J.L. Tsao

    Genetic reconstruction of individual colorectal tumor histories

    Proc. Natl. Acad. Sci. U. S. A.

    (2000)
  • C. Tomasetti et al.

    Half or more of the somatic mutations in cancers of self-renewing tissues originate prior to tumor initiation

    Proc. Natl. Acad. Sci. U. S. A.

    (2013)
  • S. Nakatsuru

    Somatic mutation of the APC gene in gastric cancer: frequent mutations in very well differentiated adenocarcinoma and signet-ring cell carcinoma

    Hum. Mol. Genet.

    (1992)
  • A. Horii

    Frequent somatic mutations of the APC gene in human pancreatic cancer

    Cancer Res.

    (1992)
  • L.I. Shlush

    Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia

    Nature

    (2014)
  • P.C. Nowell

    The clonal evolution of tumor cell populations

    Science

    (1976)
  • M. Greaves et al.

    Clonal evolution in cancer

    Nature

    (2012)
  • M. Greaves

    Evolutionary determinants of cancer

    Cancer Discov.

    (2015)
  • G.H. Heppner

    Tumor heterogeneity

    Cancer Res.

    (1984)
  • K. Anderson

    Genetic variegation of clonal architecture and propagating cells in leukaemia

    Nature

    (2011)
  • M. Gerlinger

    Intratumor heterogeneity and branched evolution revealed by multiregion sequencing

    N. Engl. J. Med.

    (2012)
  • S.P. Shah

    The clonal and mutational evolution spectrum of primary triple-negative breast cancers

    Nature

    (2012)
  • A. Marusyk et al.

    Intra-tumour heterogeneity: a looking glass for cancer?

    Nat. Rev. Cancer

    (2012)
  • A. Sottoriva

    Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics

    Proc. Natl. Acad. Sci. U. S. A.

    (2013)
  • A. Bashashati

    Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling

    J. Pathol.

    (2013)
  • M. Gerlinger

    Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing

    Nat. Genet.

    (2014)
  • B.E. Johnson

    Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma

    Science

    (2014)
  • A. Sottoriva

    A Big Bang model of human colorectal tumor growth

    Nat. Genet.

    (2015)
  • H. Kim

    Whole-genome and multisector exome sequencing of primary and post-treatment glioblastoma reveals patterns of tumor evolution

    Genome Res.

    (2015)
  • P.C. Boutros

    Spatial genomic heterogeneity within localized, multifocal prostate cancer

    Nat. Genet.

    (2015)
  • M.R. Corces

    Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution

    Nat. Genet.

    (2016)
  • A. McPherson

    Divergent modes of clonal spread and intraperitoneal mixing in high-grade serous ovarian cancer

    Nat. Genet.

    (2016)
  • C.C. Maley

    Genetic clonal diversity predicts progression to esophageal adenocarcinoma

    Nat. Genet.

    (2006)
  • E.A. Mroz

    Intra-tumor genetic heterogeneity and mortality in head and neck cancer: analysis of data from the Cancer Genome Atlas

    PLoS Med.

    (2015)
  • D. Wangsa

    Phylogenetic analysis of multiple FISH markers in oral tongue squamous cell carcinoma suggests that a diverse distribution of copy number changes is associated with poor prognosis

    Int. J. Cancer

    (2016)
  • J. Zhang

    Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing

    Science

    (2014)
  • E.C. de Bruin

    Spatial and temporal diversity in genomic instability processes defines lung cancer evolution

    Science

    (2014)
  • R. Gao

    Punctuated copy number evolution and clonal stasis in triple-negative breast cancer

    Nat. Genet.

    (2016)
  • Z. Weng

    Cell-lineage heterogeneity and driver mutation recurrence in pre-invasive breast neoplasia

    Genome Med.

    (2015)
  • P.K. Brastianos

    Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets

    Cancer Discov.

    (2015)
  • J. Cairns

    Mutation selection and the natural history of cancer

    Nature

    (1975)
  • L.R. Yates et al.

    Evolution of the cancer genome

    Nat. Rev. Genet.

    (2012)
  • A. Sottoriva

    Single-molecule genomic data delineate patient-specific tumor profiles and cancer stem cell organization

    Cancer Res.

    (2013)
  • S.L. Cooke

    Intra-tumour genetic heterogeneity and poor chemoradiotherapy response in cervical cancer

    Br. J. Cancer

    (2011)
  • E.A. Mroz

    High intratumor genetic heterogeneity is related to worse outcome in patients with head and neck squamous cell carcinoma

    Cancer

    (2013)
  • R.A. Burrell

    The causes and consequences of genetic heterogeneity in cancer evolution

    Nature

    (2013)
  • C. Swanton

    Intratumor Heterogeneity: evolution through space and time

    Cancer Res.

    (2012)
  • Cited by (0)

    This article is part of a Special Issue entitled: Evolutionary principles - heterogeneity in cancer?, edited by Dr. Robert A. Gatenby.

    View full text