Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

Chromosomal Instability as a Driver of Tumor Heterogeneity and Evolution

Samuel F. Bakhoum1,2 and Dan Avi Landau2,3,4

1Department of Radiation Oncology, Memorial Sloan Kettering Center, New York, New York 10065 2Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065 3Division of Hematology and Medical Oncology and the Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York 10021 4Core member of the New York Genome Center, New York, New York 10013 Correspondence: [email protected]; [email protected]

Large-scale, massively parallel sequencing of human cancer samples has revealed tremen- dous genetic heterogeneity within individual tumors. Indeed, tumors are composed of an admixture of diverse subpopulations—subclones—that vary in space and time. Here, we discuss a principal driver of clonal diversification in cancer known as chromosomal instability (CIN), which complements other modes of genetic diversification creating the multilayered genomic instability often seen in human cancer. Cancer cells have evolved to fine-tune missegregation rates to balance the acquisition of heterogeneity while preserving favorable genotypes, a dependence that can be exploited for a therapeutic benefit. We discuss how whole-genome doubling events accelerate clonal evolution in a subset of tumors by providing a viable path toward favorable near-triploid and present evidence for CIN-induced clonal speciation that can overcome the dependence on truncal initiating events.

ancer originates from a single cell that has order in which these mutations occur, and the Cacquired a number of central genetic lesions specific types of alterations required to produce allowing it to rescind normal multicellular de- a malignant tumor (Bozic et al. 2010; Vogelstein

www.perspectivesinmedicine.org velopment and expand, in an uncontrolled et al. 2013; Tomasetti et al. 2014; Landau et al. fashion at the expense of the host (Hanahan 2015). and Weinberg 2011). Genetic alterations that After acquiring truncal mutations, most tu- allow the initial expansion are typically defined mors undergo a second phase of intratumoral as truncal and have been the focus of much diversification. Massively parallel sequencing of investigation, as they represent attractive thera- primary, metastatic, and relapsed tumors re- peutic targets harbored by the entirety of the vealed frequent cancer subclones, which harbor malignant population (Barber et al. 2015). genomic alterations that are not detected in the These studies have shed light on the number remainder of the tumor cell population. Nota- of mutations necessary for tumorigenesis, the bly, these subclones often show evidence sup-

Editors: Charles Swanton, Alberto Bardelli, Kornelia Polyak, Sohrab Shah, and Trevor A. Graham Additional Perspectives on Cancer Evolution available at www.perspectivesinmedicine.org Copyright # 2017 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a029611 Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611

1 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

S.F. Bakhoum and D.A. Landau

portive of positive selection in an active evolu- , has long been recognized as a hall- tionary process. In many tumors, subclonal mark of tumorigenesis (Boveri 1903). Karyo- mutations show marked enrichment of driver typic heterogeneity in tumor cells draws its or- events, as well as convergent evolutionary pro- igin from “chromosomal instability” (otherwise cesses whereby multiple subclones indepen- known as CIN), a hallmark that distinguishes dently acquire mutations that activate similar most from its normal tissue counter- oncogenic pathways (Gerlinger et al. 2012; Yates part. CIN does not only generate abnormal an- and Campbell 2012; Landau et al. 2013; 2015; euploid karyotypes, but continually expands Brastianos et al. 2015; Murugaesu et al. 2015; phenotypic heterogeneity as tumor cell popula- Stachler et al. 2015; Yates et al. 2015). tions undergo consecutive cell divisions (Fig. Fundamentally, subclonal heterogeneity 1A) (Lengauer et al. 1998, 1997). stems from the ability of malignant cells to ac- is present in 80% of human quire novel mutations, a defining feature of solid (Ame et al. 2008) the majority many human tumors. Ongoing genomic diver- of which often exhibit CIN (Storchova and sification has been associated with poor prog- Kuffer 2008). CIN can be present in two major nosis and increased probability of tumor relapse forms: The first is numerical CIN whereby copy after therapy (Landau et al. 2013, 2015; Andor numbers of entire vary within et al. 2016; McGranahan et al. 2016). One of and among tumor cells. The second form is the central drivers of intratumoral diversifica- structuralCIN, whichis defined bycopynumber tion is copy number instability of particular variations of subchromosomal regions (Roschke loci, large chromosome segments, or entire and Rozenblum 2014). Although both types of chromosomes (Holland and Cleveland 2009; CIN arise through distinct cellular mechanisms Navin et al. 2011; Wang et al. 2014). Indeed, and occur at different rates, they are nonetheless cancer cells often show substantial diversity related and the presence of numerical CIN can with respect to chromosomal content (karyo- induce, or accelerate, structural CIN and vice types) (Gilgenkrantz and Rivera 2003). Aneu- versa (Crasta et al. 2012; Bakhoum et al. , which represents a “state” of abnormal 2014a). For the purpose of this review, we will

AB Lagging chromosome

Microtubules www.perspectivesinmedicine.org

Spindle pole

Kinetochore

Figure 1. Chromosomal instability (CIN) in cancer. (A) Cancer cells experience frequent errors in the segrega- tion of entire chromosomes, leading to karyotypic heterogeneity. Depending on the oncogenic and tumor suppressive effect of genes encoded by individual chromosomes, individual karyotypes will lead to distinct proliferative states and subclonal heterogeneity shaping the overall fitness of the population. Cell size depicts the relative cell fitness. Chromosomal instability (CIN) can enable rapid expansion of cells with variable fitness. (B) Lagging chromosomes during are a hallmark of CIN in cancer. They result from the erroneous attach- ments of chromosomes to spindle microtubules at the . Most of the cellular defects that lead to CIN in cancer converge onto this process and produce lagging chromosomes.

2 Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

CIN and Subclonal Evolution

focus primarily on the contribution of numeri- attachments to microtubules emanating from cal CIN to intratumoral heterogeneity. However, opposite spindle poles (Cimini et al. 2003, we will also discuss how numerical and struc- 2002), which increases their potential for mis- tural CIN are related and how the multilayered segregation or encapsulation in micronuclei CIN phenotype can play different roles in tumor (Crasta et al. 2012). evolution under various selection pressures. The process of chromosome segregation is Numerical CIN is primarily defined by the vulnerable in the face of many cellular defects “rate” at which chromosomes missegregate that are known to occur in cancer. Activation of throughout consecutive cell divisions (Len- oncogenic pathways, loss of key tumor suppres- gauer et al. 1998). The frequency of chromo- sor pathways, and a large number of therapeutic some missegregation rates, directly measured interventions can either facilitate or induce CIN in cancer-derived cell lines, are on the order of (Orr and Compton 2013; Lee et al. 2016). This 1.3 Â 1023 per chromosome copy per cell di- vulnerability stands in sharp contrast to the lack vision, which is 50–100-fold higher than in of aberrant karyotypes observed in normal hu- normal nontransformed cells (Thompson and man cells (Knouse et al. 2014). Although not yet Compton 2008; Bakhoum et al. 2009b; Laugh- fully understood, work in mice and cancer-de- ney et al. 2015). This translates into a chromo- rived cell lines revealed that the tumor suppres- some missegregation event every 3–10 cell divi- sors and p21 play a key role in safeguarding sions after accounting for cellular ploidy. In full- against aneuploidy. Mice without functional fledged malignancies, chromosome missegrega- p53 generate tumors with aneuploid karyotypes tion events may have a positive or negative effect (Shao et al. 2000). Furthermore, abrogation of on cellular fitness depending on the altered dos- p53 or p21 allows tolerance of aneuploidy and age of hundreds to thousands of genes carried the propagation of instability (Thompson and by the missegregated chromosomes (Davoli Compton 2010). Accordingly, mutations in the et al. 2013; Laughney et al. 2015); making CIN p53 pathway are more prevalent in advanced a powerful tool to rapidly shape the phenotypic tumors that typically contain significant chro- landscape of tumor cell populations. mosome copy number variations (Rivlin et al. 2011). More recently, the Hippo tumor suppres- sor signaling pathway was shown to be activated THE ORIGINS OF CIN in response to cytokinesis failure, thereby acting A number of cellular mechanisms have been as a selective safeguard against tetraploidy but found to lead to CIN in cancer (reviewed in not aneuploidy (Ganem et al. 2014). Thompson et al. 2010), including weakened sis- www.perspectivesinmedicine.org ter chromatid cohesion (Zhang et al. 2008), de- THE CONSEQUENCES OF ANEUPLOIDY regulated centrosome duplication (Ganem et al. 2009; Silkworth et al. 2009), defects in microtu- Even in a potentially permissible cellular envi- bule attachments to chromosomes at the kinet- ronment lacking p53, there is strong evidence to ochores (Cimini et al. 2001; Bakhoum et al. show that induction of aneuploidy in diploid 2009a,b), premitotic replication stress (Burrell cells is associated with reduced fitness and delays et al. 2013), and mitotic DNA damage (Crasta tumor formation (Rowald et al. 2016; Sheltzer et al. 2012; Bakhoum et al. 2014a). Irrespective et al. 2016). Workinyeast and human cells shows of the underlying cause, these mechanisms con- that aneuploidy induces metabolic and proteo- verge onto the process of chromosome segrega- toxic stress, which translate into lower prolifer- tion during mitosis, and lead to lagging chro- ation rates (Torres et al. 2007, 2010; Williams mosomes during , which is considered et al. 2008; Sheltzerand Amon 2011; Oromendia a hallmark of chromosomally unstable cells et al. 2012; Sheltzer et al. 2012; Sheltzer 2013; (Fig. 1B) (Cimini et al. 2001; Thompson and Du¨rrbaum et al. 2014). Given the increased met- Compton 2011; Bakhoum et al. 2014b). Chro- abolic demand and oxidative stress imparted by mosomes lag as a result of their simultaneous aneuploidy, this has led some to postulate that

Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 3 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

S.F. Bakhoum and D.A. Landau

aneuploidy may serve as one of the triggers for any given chromosome has 623 unique karyo- the Warburg effect often seen in cancer, whereby typic combinations. Our understanding of how cells shift away from oxidative phosphorylation CIN enables tumor subclones to sample this vast toward aerobic glycolysis (Vander Heiden et al. aneuploid fitness landscape is only beginning to 2009; Siegel and Amon 2012). Inducing aneu- emerge. Laughney et al. (2015) explored this by ploidy in vivo through depletion of Cenp-E—a allowing clonal populations to sample the an- mitotic centromere protein involved in chro- euploid fitness landscape in silico, and found mosome segregation—reduces the incidence of that they strikingly converged onto a favorable hepatocellular carcinomas (Weaver et al. 2007) near-triploid state that maximized the numbers and computational evidence suggests that the of chromosomes that are more likely to harbor percent reduction in normal cell fitness in re- and minimize the copies of chromo- sponse to aneuploidy could range anywhere somes that are more likely to harbor tumor from 6% to 30% (Gusev et al. 2001; Valind suppressor genes (Fig. 2). Importantly, both et al. 2013). diploid-derived and tetraploid-derived clones Although aneuploidy leads to a global re- achieved this optimal karyotype state, yet at a duction in cellular fitness, it also enhances the much lower fitness cost for the latter. This was ability of malignant populations to sample the due to the ability of tetraploid-derived clones to fitness landscape for superior evolutionary tra- buffer chromosome loss and avoid lethal nullis- jectories. There are many examples in which the omy. This suggests that a tetraploid interme- induction of aneuploidy and CIN leads to tu- diate arising from whole genome doubling is a morigenesis. For instance, induction of CIN desirable path toward the commonly observed through overexpression of mad2, a spindle as- favorable near-triploid state. sembly checkpoint protein, leads to spontane- Consistent with this framework, copy num- ous tumor formation (Schvartzman et al. 2011). ber analysis across cancer revealed a propensity Likewise, depletion of Cenp-E leads to the for- mation of lymphomas and lung tumors, and overexpression of hec1, a protein involved in chromosome segregation, also leads to spontaneous tumor and adenoma formation (Dı´az-Rodrı´guez et al. 2008). It is important to Near-triploid karyotype note, however, that in many of these cases, sec- ondary alterations were required for tumorigen- esis, such as the deletion of p53, p21,orp19Arf www.perspectivesinmedicine.org (Fujiwara et al. 2005; Weaver et al. 2007). Thus, Diploid aneuploidy can have both oncogenic and tumor founder

suppressive phenotypes in a context-dependent Clonal fitness Whole-genome manner. The determinants of this dichotomous doubling phenotype are still poorly understood; some have postulated that the degree of aneuploidy is an important determinant of its effect on tumor- igenesis (Silk et al. 2013); however, this concept requires additional experimental validation. Time

Figure 2. Experimental and computational work re- WHOLE-GENOME DOUBLING DURING veals a favorable near-triploid karyotype, which fa- TUMOR EVOLUTION vors clonal fitness. Diploid-derived and tetraploid- derived populations alike can achieve this karyotype, Chromosomally unstable clones have a wide ar- albeit at a much lower fitness cost for the latter, sug- ray of karyotypes available for them to sample. gesting that whole-genome doubling provides a more For example, a cell containing up to six copies of cost-effective path toward a highly favorable state.

4 Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

CIN and Subclonal Evolution

for a near-triploid state (Storchova and Kuffer to the unicellular-like cancer genome. Consis- 2008; Laughney et al. 2015). Interestingly, tently, powerful mechanisms have evolved to Storchova and Kuffer found that intratumor prevent whole-genome doubling (Ganem and karyotypic heterogeneity was maximal in tu- Pellman 2007; Mable 2013); however, once cells mors with near-triploid karyotypes, which are able to overcome these safeguards whole- may signify that higher rates of CIN allow a genome doubling generates a more plastic ge- more rapid convergence onto this favorable nome that empowers the evolutionary process state, or that the near-triploid state allows and confers higher adaptive potential to the more facile clonal diversification (Storchova malignant population. The exact contribution and Kuffer 2008). Second, genomic analysis of whole-genome doubling during early and also supports the presence of a tetraploid inter- late tumor evolution remains to be established. mediate occurring early during tumor evolu- Although tetraploidization may make it harder tion followed by gradual chromosome loss in for cells to fully inactivate key tumor suppressor at least a third of tumor samples examined (Car- genes, it is likely that the ratio of oncogenes-to- ter et al. 2012; Zack et al. 2013). Accordingly, tumor suppressor genes is the major determi- experimental work in yeast, cancer cell lines, nant of tumor fitness. Moreover, the presence of and xenograft mouse models shows that induc- multiple copies of each chromosome allows the ing whole-genome doubling promotes tumori- cells to shuffle their chromosome numbers with genesis and propagates further genomic insta- a lower risk of lethal nullisomy, where all copies bility (Fujiwara et al. 2005; Dewhurst et al. 2014; of an essential gene are lost. In summary, the Selmecki et al. 2015). recurrent patterns of chromosome copy num- Given the facilitating impact of genome du- ber changes seen across human cancer, as well as plication on cancer, it is not surprising that the observation of convergent evolution toward many factors safeguard against the proliferation near triploid karyotypes, show that chromo- of tetraploid cells either before transformation some missegregation is not a mere bystander or in the early stages of tumorigenesis (Ganem during tumor evolution, but actively contrib- and Pellman 2007). Just like aneuploidy induc- utes to malignant speciation. tion, polyploidization induces a p53-mediated cell-cycle arrest (Carter 1967; Fujiwara et al. LINKING NUMERICAL CIN TO STRUCTURAL 2005). This arrest is likely instigated by DNA CIN AND CHROMOTHRIPSIS damage and other osmotic and proteotoxic stressors that are mediated by p38/MAP kinase Numerical CIN does not exist in isolation. (Castedo et al. 2006; Srsen et al. 2006). More Numerical chromosomal aberrations are often www.perspectivesinmedicine.org recently, Ganem et al. (2014) identified the coinherited with complex structural damage Hippo tumor suppressor pathway as a tetra- that alters the juxtaposition of genomic loci ploid-specific checkpoint that responds to cy- (e.g., translocations, inversions, duplications, tokinesis failure. Activation of this pathway is etc.). Although they occur through distinct mediated in part by the presence of extra cen- mechanisms and at different rates, structural trosomes that alter small G-protein signaling, and numerical CIN are interrelated (Sheltzer which activate LATS2 kinase. LATS2 in turn in- et al. 2011; Crasta et al. 2012; Burrell et al. hibits the transcriptional regulators YAP and 2013; Bakhoum et al. 2014a). Replication TAZ through a p53-dependent mechanism. In- stress and structural chromosomal damage terestingly, this pathway was selectively activated can lead to defective mitotic chromosome seg- in response to whole-genome doubling and not regation as both acentric chromosomes and in response to whole-chromosome missegrega- dicentric chromosomes cannot form proper tion, highlighting the specificity of this pathway bi-oriented attachments to the mitotic spindle toward tetraploidy. (Murnane 2012; Burrell et al. 2013). Addition- Thus, whole-genome doubling is a key step ally, activation of the DNA damage response in the “evolution” of the multicellular genome during mitosis, either through extrinsic fac-

Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 5 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

S.F. Bakhoum and D.A. Landau

tors or uncorrected premitotic defects, per- fers the means to dramatically alter the genome turbs microtubule attachments to chromo- in the span of a single cell division. Chromo- somes and leads to whole-chromosome segre- thripsis can also occur as a result of erroneous gation errors and CIN (Bakhoum et al. 2014a; segregation of dicentric chromosomes resulting Orthwein et al. 2014). from fusion (Maciejowski et al. 2015). Conversely, chromosome missegregation It is likely that most chromothripsis events lead during mitosis can also lead to abrupt structural to unviability—as cells lose chromosome frag- chromosomal aberrations. Lagging chromo- ments containing essential genes (Zhang et al. somes are repeatedly encapsulated in micronu- 2015)—although rarely chromothripsis leads to clei, which are defective in DNA replication and amplification or fusion events that significantly repair and have dysfunctional micronuclear en- enhance cellular fitness. Indeed, in their original velopes (Crasta et al. 2012). Such micronuclei report of chromothripsis, Stephens et al. (2011) undergo catastrophic micronuclear envelope found that this process is associated with the collapse associated with invasion of the endo- formation of -harboring double-mi- plasmic reticulum tubules into their enclosed nute chromosomes that existed in hundreds of chromatin (Hatch et al. 2013). Through a yet copies per cell. unidentified mechanism, exposure of this dou- The rapid, punctuated genomic changes ble-stranded DNA to the cytosol leads to dra- that result from such genomic “catastrophes,” matic chromosome pulverization (Fig. 3) stands in stark contrast to the progressive geno- (Crasta et al. 2012). By performing single-cell mic alteration caused by numerical changes in sequencing, Zhang et al. (2015) were able to chromosome copy numbers. The interconnec- show that pulverized chromosomes can be re- tedness between structural and numerical CIN paired through nonhomologous end joining, offers tumor cell populations the ability to al- leading to massive chromosome rearrange- ternate between these two rates of genomic ments, a process known as “chromothripsis” transformation during the process of cancer (Stephens et al. 2011). This mitotic pathway of- evolution.

Aneuploid karyotype

Micronucleus formation www.perspectivesinmedicine.org

Lagging chromosomes Chromosome pulverization

Mitotic DNA damage

Figure 3. Diagram depicting the link between numerical and structural chromosomal instability (CIN). In addition to aneuploid karyotypes, chromosome segregation during mitosis also leads to the formation of chromosome-containing micronuclei. This in turn leads to pulverization of their enclosed chromosomes and persistence of DNA damage into the subsequent mitosis. Activation of a partial mitotic DNA damage response in turn leads to whole chromosome segregation forming a cycle linking numerical and structural CIN.

6 Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

CIN and Subclonal Evolution

THE DYNAMICS OF CIN IN TUMOR To quantitatively model the evolutionary CLONAL EVOLUTION AND THERAPEUTIC impact of CIN, Laughney et al. (2015) simulated RESPONSE the process of clonal evolution under chromo- somally unstable conditions. Cell viability was How distinct chromosome copy numbers alter determined by the net dosage of pro- and anti- tumor cell fitness remains a poorly understood proliferative genes they contained, dictated by process. Chromosome (or arm)-level amplifica- the respective chromosome copy numbers tions and deletions alter transcript and protein (Fig. 1A). Interestingly, the dependence of clon- levels of hundreds to thousands of genes (Torres al fitness on chromosome missegregation rates et al. 2007; Williams et al. 2008) and have often was nonlinear and parabolic—fitness rapidly been associated with altered prognosis and dif- declined when missegregation rates were either ferent tumor behavior. too high or too low. At very low rates, popula- The identification of genes on these chro- tions lacked the ability to generate phenotypic mosome segments responsible for altered prog- heterogeneity induced by karyotypic diversity. nosis has been challenging. A recent attempt by As such, clonal populations were less capable of Roy et al. (2016) used a multitiered genomic sampling fitter karyotypic states. On the other dissection strategy and patient data to identify a hand, excessively high rates of chromosome handful of genes responsible for poor progno- missegregation prevented populations from sis in low-grade gliomas harboring loss of the preserving highly fit karyotypic states, and also 9p chromosome arm. The investigators found resulted in frequent nullisomy—the lethal loss that reductions in messenger RNA levels in re- of all copies of any given chromosome. Thus, sponse to 9p loss was due to haploinsufficiency tumor populations thrive when they balance the in subset of genetic loci. In another subset, ability to diversify with the ability to preserve there was evidence for epigenetic inactivation favorable genotypes. Strikingly, When missegre- on the remaining copies of the 9p chromosome gation frequencies were experimentally mea- arms leading to significant down-regulation of sured in cancer-derived cell lines they largely transcript levels. Interestingly, many genes on clustered around the range predicted to be op- this chromosome arm were not significantly timal for population fitness (1.9 Â 1023 affected suggesting a dosage compensation chromosome per copy per cell division), sug- mechanism. gesting a strong evolutionary pressure to opti- In a separate study, Davoli et al. (2013) mize CIN rates in human cancer (Fig. 4) mapped tumor suppressor genes and oncogenes (Laughney et al. 2015). on human chromosomes inferred from the mu- The concept of an optimal range of chromo- www.perspectivesinmedicine.org tational patterns of these genes in human tu- some missegregation rates has important ther- mors. By analyzing the potency and density of apeutic implications (Silk et al. 2013; Laughney oncogenes and tumor suppressor genes, they et al. 2015). It predicts that tumors with mod- found that human chromosomes are not equiv- erate CIN (within the optimal range) would alent with respect to the net cumulative onco- show the highest likelihood of stable plasticity genic and tumor suppressive effect of the genes promoting therapeutic resistance. On the other they encode, suggesting that cumulative hap- hand, tumors would be vulnerable to either a loinsufficiency and triplosensitivity of pro- decrease or an increase of CIN rates outside of and antiproliferative genes shape tumor karyo- the optimal range that supports clonal evolu- types and in part explains chromosome-level tion. This is indeed supported by a number of amplifications and deletions in cancer. This clinical observations and experimental evidence work provides aframework to explain how chro- (Bakhoum et al. 2011; Birkbak et al. 2011; Roy- mosome copy number changes that affect hun- lance et al. 2011; Jamal-Hanjani et al. 2015; An- dreds of genes may still output a coherent fitness dor et al. 2016). Among -de- effect, and therefore subjected to evolutionary rived cell lines, chromosomally unstable cells selection. are more resistant to targeted kinase inhibition

Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 7 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

S.F. Bakhoum and D.A. Landau

Optimal for Optimal for targeted cytotoxic therapy therapy Optimal viability

Inability Genomic to adapt catastrophe

CIN Low High

Figure 4. Relative tumor clonal fitness as a function of chromosomal instability (CIN). Excessively low CIN would suppress genomic heterogeneity leading to reduction in tumor adaptability, whereas excessively high CIN would lead to genomic collapse, DNA damage, and frequent nullisomy events incompatible with viability.

as compared with their chromosomally stable mosome missegregation are associated with a counterparts (Lee et al. 2011). In patients with higher likelihood of pathologic response after breast cancer, the presence of moderate CIN is treatment with radiation therapy and concur- associated with inferior prognosis and resis- rent 5-fluorouracil (Zaki et al. 2014). Consistent tance to taxanes whereas extreme CIN is associ- with this notion, in the pancancer analysis per- ated with improved survival and reduced tumor formed by Andor et al. (2016), the negative im- relapse after primary therapy (Swanton et al. pact of CIN on survival was abolished in the 2009; Roylance et al. 2011; Jamal-Hanjani subset of patients who received DNA-damaging et al. 2015). Furthermore, in a pancancer geno- therapies (Fig. 4). mic analysis of intratumor heterogeneity, pa- Thus, elevating chromosome missegrega- tients that had tumors in the middle quartiles tion rates in inherently chromosomally unstable of chromosome copy number variations were at cancers has been proposed as a potential anti- www.perspectivesinmedicine.org a significantly greater risk for death compared neoplastic strategy. Experimental evidence with those with tumors in the lowest or highest shows that cells are rendered more sensitive to quartiles of chromosome copy number varia- agents that increase chromosome missegrega- tions (Andor et al. 2016). tion when they already experience preexisting This notion offers a largely untapped oppor- CIN (Janssen et al. 2009; Bakhoum et al. tunity to target chromosomally unstable tu- 2015). The recent development of specific in- mors, by increasing CIN beyond the tolerable hibitors that are capable of inducing massive threshold. Support for this strategy stems from chromosome missegregation is encouraging observations that CIN is a predictor of favorable and has shown substantial preclinical promise response to therapies that induce chromosome (Mason et al. 2014). These agents can be ratio- missegregation, particularlyconventional DNA- nally combined with traditional therapies, such damaging cytotoxic therapies. For instance, as DNA-damaging agents, to achieve greater CIN correlates with sensitivity to platinum- therapeutic benefits. Collectively, a better un- based therapies in ovarian cancer (Swanton derstanding of the pretreatment CIN status as et al. 2009), and in those with locally advanced well as the reciprocal effect of therapy on chro- rectal cancer, considerablyelevated rates of chro- mosome stability will enable us to design strat-

8 Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

CIN and Subclonal Evolution

egies that aim at displacing cells from their ac- pects of CIN in cancer. These include identifying quired favorable copy number states. more comprehensively the cellular mechanisms that underlie CIN and enable tolerance for a chromosomally unstable state. Second, because CIN AND THE EMERGENCE OF SUBCLONAL CIN is defined by a rate of chromosome mis- THERAPEUTIC RESISTANCE segregation, methods interrogating this process in cancer should be refined to not only measure The ability for CIN to enable phenotypic diver- the extent of chromosome-level copy number sity among tumor cells can accelerate the emer- alterations in cancer but also the rate at which gence of subclones with specific genotypes that chromosome number changes throughout con- confer therapeutic resistance. For instance, secutive cell divisions. Widely used methods treatment with EGFR inhibitors rapidly selects such as comparative genomic hybridization for glioblastoma-derived cells with amplifica- and DNA sequencing of bulk tumor samples tion of the number of copies of chromosome significantly mask the extent of karyotypic 7, which harbors the EGFR gene (Chen et al. cell-to-cell variation, which may now be ex- 2015). tensively probed through single-cell analyses In addition to directly altering the copy (Laughney et al. 2015; Bakker et al. 2016). Third, number of the target gene, CIN has the potential our efforts to understand chromosome copy to induce resistance by circumventing driver number changes would need to be coupled alterations and relieving tumors from their de- with quantitative models that aim to predict pendence on oncogenic events that underlie the behavior of tumor cell populations derived their genesis. This was experimentally tested by from our knowledge of chromosome segrega- Sotillo et al. (2010) where they induced CIN in tion at the single-cell level. Ultimately, these Kras-drivenlungtumorsthroughtransientover- models hold the promise of providing robust expression of Mad2. Subsequent withdrawal of predictive tools enabling precision therapeutic Kras led to tumor regression; however, transient manipulation of the rapidly evolving cancer Mad2 overexpression promoted tumor relapse genome. after Kras withdrawal. These Kras-independent tumors were characterized by markedly elevated rates of aneuploidy and CIN. In these tumors, the investigators found up-regulation of pro- ACKNOWLEDGMENTS proliferative pathways as evidenced by elevated The authors thank Ashley Laughney Bakhoum, levels of phosphorylated forms of ERK, Stat3, Luciano Martelotto (both at Memorial Sloan

www.perspectivesinmedicine.org and AKT. Other studies revealed that CIN can Kettering Cancer Center), and Jason Sheltzer circumvent the loss-of-function of otherwise (Cold Spring Harbor Laboratory) for comments essential genes in yeast (Rancati et al. 2008; Liu and feedback. D.A.L. is supported by the et al. 2015). Collectively, these findings support Burroughs Wellcome Fund Career Award for the notion that CIN represents an adaptive Medical Scientists, Stand Up to Cancer Innova- cellular tool to overcome inhibition of essential tive Research Grant, the Kimmel Scholar’s genes or truncal genetic alterations that occur Award, the ASH Scholar Award, Lym- early during tumor evolution. phoma Society translational research program, and by the National Institutes of Health (NIH) Big Data to Knowledge initiative (BD2K, CONCLUDING REMARKS 1K01ES025431-01). S.F.B. is supported by The Understanding the mechanisms and dynamics Department of Defense Breast Cancer Research of tumor genomic diversification is critical to Breakthrough Award No. W81XWH-16-1-0315, our ability to overcome the challenge of drug the Elsa U. Pardee Foundation, and the NCI resistance. To target CIN therapeutically, we re- MSKCC Cancer Center Core Grant P30 quire a deeper understanding of several key as- CA008748.

Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 9 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

S.F. Bakhoum and D.A. Landau

REFERENCES Burrell RA, McClelland SE, Endesfelder D, Groth P, Weller M-C, Shaikh N, Domingo E, Kanu N, Dewhurst SM, Ame JC, Cimini D, Fouquerel E, Gauthier LR, Biard D, Gronroos E, et al. 2013. Replication stress links structural Boussin FD, Dantzer F, de Murcia G, Schreiber V. 2008. and numerical cancer chromosomal instability. Nature Merotelic kinetochore orientation, aneuploidy, and can- 494: 492–496. cer. Biochim Biophys Acta 1786: 32–40. Carter SB. 1967. Effects of cytochalasins on mammalian Andor N, Graham TA, Jansen M, Xia LC, Aktipis CA, Pet- cells. Nature 213: 261–264. ritsch C, Ji HP,Maley CC. 2016. Pan-cancer analysis of the Carter SLS, Cibulskis KK, Helman EE, McKenna AA, Shen extent and consequences of intratumor heterogeneity. HH, Zack TT, Laird PWP, Onofrio RCR, Winckler WW, Nat Med 22: 105–113. Weir BAB, et al. 2012. Absolute quantification of somatic Bakhoum SF, Genovese G, Compton DA. 2009a. Deviant DNA alterations in human cancer. Nat Biotechnol 30: kinetochore microtubule dynamics underlie chromo- 413–421. somal instability. Curr Biol 19: 1937–1942. Castedo M, Coquelle A, Vivet S, Vitale I, Kauffmann A, Bakhoum SF, Thompson SL, Manning AL, Compton DA. Dessen P, Pequignot MO, Casares N, Valent A, Mouha- 2009b. Genome stability is ensured by temporal control mad S, et al. 2006. Apoptosis regulation in tetraploid of kinetochore-microtubule dynamics. Nat Cell Biol 11: cancer cells. EMBO J 25: 2584–2595. 27–35. Chen G, Mulla WA, Kucharavy A, Tsai H-J, Rubinstein B, Bakhoum SF, Danilova OV,Kaur P,Levy NB, Compton DA. Conkright J, McCroskey S, Bradford WD, WeemsL, Haug 2011. Chromosomal instability substantiates poor prog- JS, et al. 2015. Targeting the adaptability of heteroge- nosis in patients with diffuse large B-cell lymphoma. Clin neous aneuploids. Cell 160: 771–784. Cancer Res 17: 7704–7711. Cimini D, Howell B, Maddox P, Khodjakov A, Degrassi F, Bakhoum SF, Kabeche L, Murnane JP, Zaki BI, Compton Salmon ED. 2001. Merotelic kinetochore orientation is a DA. 2014a. DNA-damage response during mitosis induc- major mechanism of aneuploidy in mitotic mammalian es whole-chromosome missegregation. Cancer Discov 4: tissue cells. J Cell Biol 153: 517–527. 1281–1289. Cimini D, Fioravanti D, Salmon ED, Degrassi F. 2002. Mer- Bakhoum SF, Silkworth WT, Nardi IK, Nicholson JM, otelic kinetochore orientation versus chromosome Compton DA, Cimini D. 2014b. The mitotic origin of mono-orientation in the origin of lagging chromosomes chromosomal instability. Curr Biol 24: R148–R149. in human primary cells. J Cell Sci 115: 507–515. Bakhoum SF, Kabeche L, Wood MD, Laucius CD, Qu D, Cimini DD, Ben B Moree, Canman JCJ, Salmon EDE. 2003. Laughney AM, Reynolds GE, Louie RJ, Phillips J, Chan Merotelic kinetochore orientation occurs frequently dur- DA, et al. 2015. Numerical chromosomal instability me- ing early mitosis in mammalian tissue cells and error diates susceptibility to radiation treatment. Nat Commun correction is achieved by two different mechanisms. 6: 5990. J Cell Sci 116: 4213–4225. Bakker B, Taudt A, Belderbos ME, Porubsky D, Spierings Crasta KK, Ganem NJ, Dagher RR, Lantermann AB, Ivanova DCJ, de Jong TV, Halsema N, Kazemier HG, Hoekstra- EVE, Pan YY, Nezi LL, Protopopov AA, Chowdhury DD, Wakker K, Bradley A, et al. 2016. Single-cell sequencing Pellman DD. 2012. DNA breaks and chromosome pul- reveals karyotype heterogeneity in murine and human verization from errors in mitosis. Nature 482: 53–58. malignancies. Genome Biol 17: 115. Davoli T, Xu AW, Mengwasser KE, Sack LM, Yoon JC, Park Barber LJ, Davies MN, Gerlinger M. 2015. Dissecting cancer PJ, Elledge SJ. 2013. Cumulative haploinsufficiency and evolution at the macro-heterogeneity and micro-hetero- triplosensitivity drive aneuploidy patterns and shape the geneity scale. Curr Opin Genet Dev 30: 1–6. cancer genome. Cell 155: 948–962. www.perspectivesinmedicine.org Birkbak NJ, Eklund AC, Li Q, McClelland SE, Endesfelder D, Dewhurst SM, McGranahan N, Burrell RA, Rowan AJ, Tan P, Tan IB, Richardson AL, Szallasi Z, Swanton C. Gronroos E, Endesfelder D, Joshi T, Mouradov D, Gibbs 2011. Paradoxical relationship between chromosomal P,Ward RL, et al. 2014. Tolerance of whole-genome dou- instability and survival outcome in cancer. Cancer Res bling propagates chromosomal instability and accelerates 71: 3447–3452. cancer genome evolution. Cancer Discov 4: 175–185. Boveri T. 1903. U¨ ber mehrpolige mitosen als mittel zur ´ ´ analyse des zellkerns [On multipolar mitosis as a means Dıaz-Rodrıguez E, Sotillo R, Schvartzman JM, Benezra R. of analysis of the cell nucleus]. Verh Phys Med Ges Wurz- 2008. Hec1 overexpression hyperactivates the mitotic burg 35: 67–90. checkpoint and induces tumor formation in vivo. Proc Natl Acad Sci 105: 16719–16724. Bozic I, Antal T, Ohtsuki H, Carter H, Kim D, Chen S, Karchin R, Kinzler KW, Vogelstein B, Nowak MA. 2010. Du¨rrbaum M, Kuznetsova AY,Passerini V,Stingele S, Stoehr Accumulation of driver and passenger mutations during G, Storchova Z. 2014. Unique features of the transcrip- tumor progression. Proc Natl Acad Sci 107: 18545– tional response to model aneuploidy in human cells. 18550. BMC Genomics 15: 139. Brastianos PK, Carter SL, Santagata S, Cahill DP, Taylor- Fujiwara T, Bandi M, Nitta M, Ivanova EV, Bronson RT, Weiner A, Jones RT, Van Allen EM, Lawrence MS, Pellman D. 2005. Cytokinesis failure generating tetra- Horowitz PM, Cibulskis K, et al. 2015. Genomic char- ploids promotes tumorigenesis in p53-null cells. Nature acterization of brain metastases reveals branched evolu- 437: 1043–1047. tion and potential therapeutic targets. Cancer Discov 5: Ganem NJ, Pellman D. 2007. Limiting the proliferation of 1164–1177. polyploid cells. Cell 131: 437–440.

10 Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

CIN and Subclonal Evolution

Ganem NJ, Godinho SA, Pellman D. 2009. A mechanism Lengauer C, Kinzler KW, Vogelstein B. 1998. Genetic insta- linking extra centrosomes to chromosomal instability. bilities in human cancers. Nature 396: 643–649. Nature 460: 278–282. Liu G, Yong MYJ, Yurieva M, Srinivasan KG, Liu J, Lim JSY, Ganem NJ, Cornils H, Chiu SY, O’Rourke KP, Arnaud J, Poidinger M, Wright GD, Zolezzi F, Choi H, et al. 2015. Yimlamai D, The´ry M, Camargo FD, Pellman D. 2014. Gene essentiality is a quantitative property linked to cel- Cytokinesis failure triggers Hippo tumor suppressor lular evolvability. Cell 163: 1388–1399. pathway activation. Cell 158: 833–848. Mable BK. 2013. Polyploids and hybrids in changing envi- Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, ronments: Winners or losers in the struggle for adapta- Gronroos E, Martinez P,Matthews N, Stewart A, Tarpey P, tion? Heredity (Edinb) 110: 95–96. et al. 2012. Intratumor heterogeneity and branched evo- Maciejowski J, Li Y,Bosco N, Campbell PJ, de Lange T.2015. lution revealed by multiregion sequencing. N Engl J Med Chromothripsis and kataegis induced by telomere crisis. 366: 883–892. Cell 163: 1641–1654. Gilgenkrantz S, Rivera EM. 2003. The history of cytogenet- Mason JM, Lin DC-C, Wei X, Che Y, Yao Y, Kiarash R, ics. Portraits of some pioneers. Ann Genet 46: 433–442. Cescon DW, Fletcher GC, Awrey DE, Bray MR, et al. Gusev Y,Kagansky V,Dooley WC. 2001. Long-term dynam- 2014. Functional characterization of CFI-400945, a ics of chromosomal instability in cancer: A transition Polo-like kinase 4 inhibitor, as a potential anticancer probability model. Math Comput Model 33: 1253–1273. agent. Cancer Cell 26: 163–176. Hanahan D, Weinberg RA. 2011. Hallmarks of cancer: The McGranahan N, Furness AJS, Rosenthal R, Ramskov S, next generation. Cell 144: 646–674. Lyngaa R, Saini SK, Jamal-Hanjani M, Wilson GA, Birk- Hatch EM, Fischer AH, Deerinck TJ, Hetzer MW. 2013. bak NJ, Hiley CT, et al. 2016. Clonal neoantigens elicit T Catastrophic nuclear envelope collapse in cancer cell mi- cell immunoreactivity and sensitivity to immune check- cronuclei. Cell 154: 47–60. point blockade. Science 351: 1463–1469. Holland AJ, Cleveland DW.2009. Boveri revisited: Chromo- Murnane JP. 2012. Telomere dysfunction and chromosome somal instability, aneuploidy and tumorigenesis. Nat Rev instability. Mutat Res 730: 28–36. Mol Cell Biol 10: 478–487. Murugaesu N, Wilson GA, Birkbak NJ, Watkins TBK, Jamal-Hanjani M, A’Hern R, Birkbak NJ, Gorman P,Gron- McGranahan N, Kumar S, Abbassi-Ghadi N, Salm M, roos E, Ngang S, Nicola P, Rahman L, Thanopoulou E, Mitter R, Horswell S, et al. 2015. Tracking the genomic Kelly G, et al. 2015. Extreme chromosomal instability evolution of esophageal adenocarcinoma through neo- forecasts improved outcome in ER-negative breast can- adjuvant chemotherapy. Cancer Discov 5: 821–831. cer: A prospective validation cohort study from the TACT Navin N, Kendall J, Troge J, Andrews P,Rodgers L, McIndoo trial. Ann Oncol 26: 1340–1346. J, Cook K, Stepansky A, Levy D, Esposito D, et al. 2011. Janssen A, Kops GJPL, Medema RH. 2009. Elevating the Tumour evolution inferred by single-cell sequencing. frequency of chromosome mis-segregation as a strategy Nature 472: 90–94. to kill tumor cells. Proc Natl Acad Sci 106: 19108–19113. Oromendia AB, Dodgson SE, Amon A. 2012. Aneuploidy Knouse KA, Wu J, Whittaker CA, Amon A. 2014. Single cell causes proteotoxic stress in yeast. Genes Dev 26: 2696– sequencing reveals low levels of aneuploidy across mam- 2708. malian tissues. Proc Natl Acad Sci 111: 13409–13414. Orr B, Compton DA. 2013. A double-edged sword: How Landau DA, Carter SL, Stojanov P, McKenna A, Stevenson oncogenes and tumor suppressor genes can contribute K, Lawrence MS, Sougnez C, Stewart C, Sivachenko A, to chromosomal instability. Front Oncol 3: 164. Wang L, et al. 2013. Evolution and impact of subclonal Orthwein A, Fradet-Turcotte A, Noordermeer SM, Canny mutations in chronic lymphocytic leukemia. Cell 152: MD, Brun CM, Strecker J, Escribano-Diaz C, Durocher

www.perspectivesinmedicine.org 714–726. D. 2014. Mitosis inhibits DNA double-strand break Landau DA, Tausch E, Taylor-Weiner AN, Stewart C, Reiter repair to guard against telomere fusions. Science 344: JG, Bahlo J, Kluth S, Bozic I, Lawrence M, Bo¨ttcher S, et 189–193. al. 2015. Mutations driving CLL and their evolution in Rancati G, Pavelka N, Fleharty B, Noll A, Trimble R, Walton progression and relapse. Nature 526: 525–530. K, Perera A, Staehling-Hampton K, Seidel CW, Li R. Laughney AM, Elizalde S, Genovese G, Bakhoum SF. 2015. 2008. Aneuploidy underlies rapid adaptive evolution of Dynamics of tumor heterogeneity derived from clonal yeast cells deprived of a conserved cytokinesis motor. Cell karyotypic evolution. Cell Rep 12: 809–820. 135: 879–893. Lee AJX, Endesfelder D, Rowan AJ, Walther A, Birkbak NJ, Rivlin N, Brosh R, Oren M, Rotter V.2011. Mutations in the Futreal PA, Downward J, Szallasi Z, Tomlinson IPM, p53 : Important milestones at the Howell M, et al. 2011. Chromosomal instability confers various steps of tumorigenesis. Genes Cancer 2: 466–474. intrinsic multidrug resistance. Cancer Res 71: 1858– Roschke AV,Rozenblum E. 2014. Multi-layered cancer chro- 1870. mosomal instability phenotype. Front Oncol 3: 302. Lee H-S, Lee NCO, Kouprina N, Kim J-H, Kagansky A, Bates Rowald K, Mantovan M, Passos J, Buccitelli C, Mardin BR, S, Trepel JB, Pommier Y, Sackett D, Larionov V. 2016. Korbel JO, Jechlinger M, Sotillo R. 2016. Negative selec- Effects of anticancer drugs on chromosome instability tion and chromosome instability induced by Mad2 over- and new clinical implications for tumor-suppressing expression delay breast cancer but facilitate oncogene- therapies. Cancer Res 76: 902–911. independent outgrowth. Cell Rep 15: 2679–2691. Lengauer C, Kinzler KW, Vogelstein B. 1997. Genetic insta- Roy DM, Walsh LA, Desrichard A, Huse JT, Wu W, Gao J, bility in colorectal cancers. Nature 386: 623–627. Bose P, Lee W, Chan TA. 2016. Integrated genomics for

Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 11 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

S.F. Bakhoum and D.A. Landau

pinpointing survival loci within arm-level somatic copy Stephens PJ, Greenman CD, Fu B, YangF,Bignell GR, Mudie number alterations. Cancer Cell 29: 737–750. LJ, Pleasance ED, Lau KW, Beare D, Stebbings LA, et al. Roylance R, Endesfelder D, Gorman P,Burrell RA, Sander J, 2011. Massive genomic rearrangement acquired in a sin- Tomlinson I, Hanby AM, Speirs V, Richardson AL, Birk- gle catastrophic event during cancer development. Cell bak NJ, et al. 2011. Relationship of extreme chromosomal 144: 27–40. instability with long-term survival in a retrospective Storchova Z, Kuffer C. 2008. The consequences of tetraploi- analysis of primary breast cancer. Cancer Epidemiol Bio- dy and aneuploidy. J Cell Sci 121: 3859–3866. markers Prev 20: 2183–2194. Swanton C, Nicke B, Schuett M, Eklund AC, Ng C, Li Q, Schvartzman J-M, Duijf PHG, Sotillo R, Coker C, Benezra Hardcastle T, Lee A, Roy R, East P, et al. 2009. Chromo- R. 2011. Mad2 is a critical mediator of the chromosome somal instability determines taxane response. Proc Natl instability observed upon Rb and p53 pathway inhibi- Acad Sci 106: 8671–8676. tion. Cancer Cell 19: 701–714. Thompson SL, Compton DA. 2008. Examining the link Selmecki AM, Maruvka YE, Richmond PA, Guillet M, Shor- between chromosomal instability and aneuploidy in hu- esh N, Sorenson AL, De S, Kishony R, Michor F, Dowell man cells. J Cell Biol 180: 665–672. R, et al. 2015. can drive rapid adaptation in Thompson SL, Compton DA. 2010. Proliferation of aneu- yeast. Nature 519: 349–352. ploid human cells is limited by a p53-dependent mech- Shao C, Deng L, Henegariu O, Liang L, Stambrook PJ, anism. J Cell Biol 188: 369–381. Tischfield JA. 2000. Chromosome instability contributes Thompson SL, Compton DA. 2011. Chromosome missegre- to loss of heterozygosity in mice lacking p53. Proc Natl gation in human cells arises through specific types of Acad Sci 97: 7405–7410. kinetochore–microtubule attachment errors. Proc Natl Sheltzer JM. 2013. A transcriptional and metabolic signature Acad Sci 108: 17974–17978. of primary aneuploidy is present in chromosomally un- Thompson SL, Bakhoum SF, Compton DA. 2010. Mecha- stable cancer cells and informs clinical prognosis. Cancer nisms of chromosomal instability. Curr Biol 20: R285– Res 73: 6401–6412. R295. Sheltzer JM, Amon A. 2011. The aneuploidy paradox: Costs Tomasetti C, Marchionni L, Nowak MA, Parmigiani G, Vo- and benefits of an incorrect karyotype. Trends Genet 27: gelstein B. 2014. Only three driver gene mutations are 446–453. required for the development of lung and colorectal can- Sheltzer JM, Blank HM, Pfau SJ, Tange Y, George BM, cers. Proc Natl Acad Sci 112: 118–123. Humpton TJ, Brito IL, Hiraoka Y, Niwa O, Amon A. Torres EM, Sokolsky T, Tucker CM, Chan LY, Boselli M, 2011. Aneuploidy drives genomic instability in yeast. Sci- Dunham MJ, Amon A. 2007. Effects of aneuploidy on ence 333: 1026–1030. cellular physiology and cell division in haploid yeast. Sci- Sheltzer JM, Torres EM, Dunham MJ, Amon A. 2012. Tran- ence 317: 916–924. scriptional consequences of aneuploidy. Proc Natl Acad Torres EM, Dephoure N, Panneerselvam A, Tucker CM, Sci 109: 12644–12649. Whittaker CA, Gygi SP, Dunham MJ, Amon A. 2010. Sheltzer JM, Ko JH, Burgos N, Chung ES. 2016. Single- Identification of aneuploidy-tolerating mutations. Cell chromosome aneuploidy commonly functions as a tu- 143: 71–83. mor suppressor. bioRxiv 040162; doi: 10.1101/040162. Valind A, Jin Y, Gisselsson D. 2013. Elevated tolerance to Siegel JJ, Amon A. 2012. New insights into the troubles of aneuploidy in cancer cells: Estimating the fitness aneuploidy. Annu Rev Cell Dev Biol 28: 189–214. effects of chromosome number alterations by in silico Silk AD, Zasadil LM, Holland AJ, Vitre B, Cleveland DW, modelling of somatic genome evolution. PLoS ONE 8: e70445.

www.perspectivesinmedicine.org Weaver BA. 2013. Chromosome missegregation rate pre- dicts whether aneuploidy will promote or suppress tu- Vander Heiden MG, Cantley LC, Thompson CB. 2009. Un- mors. Proc Natl Acad Sci 110: E4134–E4141. derstanding the Warburg effect: The metabolic require- Silkworth WT, Nardi IK, Scholl LM, Cimini D. 2009. Mul- ments of cell proliferation. Science 324: 1029–1033. tipolar spindle pole coalescence is a major source of ki- Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz netochore mis-attachment and chromosome mis-segre- LA, Kinzler KW. 2013. Cancer genome landscapes. Sci- gation in cancer cells. PLoS ONE 4: e6564. ence 339: 1546–1558. Sotillo R, Schvartzman J-M, Socci ND, Benezra R. 2010. Wang Y, Waters J, Leung ML, Unruh A, Roh W, Shi X, Chen Mad2-induced chromosome instability leads to lung tu- K, Scheet P, Vattathil S, Liang H, et al. 2014. Clonal evo- mour relapse after oncogene withdrawal. Nature 464: lution in breast cancer revealed by single nucleus genome 436–440. sequencing. Nature 1–15. Srsen V, Gnadt N, Dammermann A, Merdes A. 2006. Weaver BAA, Silk AD, Montagna C, Verdier-Pinard P,Cleve- Inhibition of centrosome protein assembly leads to land DW. 2007. Aneuploidy acts both oncogenically and p53-dependent exit from the . J Cell Biol 174: as a tumor suppressor. Cancer Cell 11: 25–36. 625–630. Williams BR, Prabhu VR, Hunter KE, Glazier CM, Whitta- Stachler MD, Taylor-WeinerA, Peng S, McKenna A, Agoston ker CA, Housman DE, Amon A. 2008. Aneuploidy affects AT, Odze RD, Davison JM, Nason KS, Loda M, proliferation and spontaneous immortalization in mam- Leshchiner I, et al. 2015. Paired exome analysis of Bar- malian cells. Science 322: 703–709. rett’s esophagus and adenocarcinoma. Nat Genet 47: Yates LR, Campbell PJ. 2012. Evolution of the cancer ge- 1047–1055. nome. Nat Rev Genet 13: 795–806.

12 Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

CIN and Subclonal Evolution

Yates LR, Gerstung M, Knappskog S, Desmedt C, Gundem superior response of rectal adenocarcinoma to chemo- G, Van Loo P,Aas T,Alexandrov LB, Larsimont D, Davies radiation therapy. Cancer 120: 1733–1742. H, et al. 2015. Subclonal diversification of primary breast Zhang N, Ge G, Meyer R, Sethi S, Basu D, Pradhan S, Zhao cancer revealed by multiregion sequencing. Nat Med 21: YJ, Li XN, Cai WW, El-Naggar AK, et al. 2008. 751–759. Overexpression of Separase induces aneuploidy and Zack TI, Schumacher SE, Carter SL, Cherniack AD, Saksena mammary tumorigenesis. Proc Natl Acad Sci 105: G, Tabak B, Lawrence MS, Zhang C-Z, Wala J, Mermel 13033–13038. CH, et al. 2013. Pan-cancer patterns of somatic copy Zhang CZ, Spektor A, Cornils H, Francis JM, Jackson EK, number alteration. Nat Genet 45: 1134–1140. Liu S, Meyerson M, Pellman D. 2015. Chromothripsis Zaki BI, Suriawinata AA, Eastman AR, Garner KM, from DNA damage in micronuclei. Nature 522: 179– Bakhoum SF. 2014. Chromosomal instability portends 184. www.perspectivesinmedicine.org

Cite this article as Cold Spring Harb Perspect Med 2017;7:a029611 13 Downloaded from http://perspectivesinmedicine.cshlp.org/ at New York Genome Center on September 8, 2017 - Published by Cold Spring Harbor Laboratory Press

Chromosomal Instability as a Driver of Tumor Heterogeneity and Evolution

Samuel F. Bakhoum and Dan Avi Landau

Cold Spring Harb Perspect Med 2017; doi: 10.1101/cshperspect.a029611 originally published online February 17, 2017

Subject Collection Cancer Evolution

Homeostasis Back and Forth: An Ecoevolutionary The Role of Aneuploidy in Cancer Evolution Perspective of Cancer Laurent Sansregret and Charles Swanton David Basanta and Alexander R.A. Anderson Principles of Reconstructing the Subclonal Treatment-Induced Mutagenesis and Selective Architecture of Cancers Pressures Sculpt Cancer Evolution Stefan C. Dentro, David C. Wedge and Peter Van Subramanian Venkatesan, Charles Swanton, Barry Loo S. Taylor, et al. The Evolution and Ecology of Resistance in Big Bang Tumor Growth and Clonal Evolution Cancer Therapy Ruping Sun, Zheng Hu and Christina Curtis Robert Gatenby and Joel Brown Phylogenetic Quantification of Intratumor Tumor Microenvironment and Differential Heterogeneity Responses to Therapy Thomas B.K. Watkins and Roland F. Schwarz Eishu Hirata and Erik Sahai Observing Clonal Dynamics Across The ''Achilles Heel'' of Cancer and Its Implications Spatiotemporal Axes: A Prelude to Quantitative for the Development of Novel Immunotherapeutic Fitness Models for Cancer Strategies Andrew W. McPherson, Fong Chun Chan and Kroopa Joshi, Benjamin M Chain, Karl S Peggs, et Sohrab P. Shah al. Chromosomal Instability as a Driver of Tumor Evolution of Premalignant Disease Heterogeneity and Evolution Kit Curtius, Nicholas A. Wright and Trevor A. Samuel F. Bakhoum and Dan Avi Landau Graham Order Matters: The Order of Somatic Mutations Coevolution of Leukemia and Host Immune Cells Influences Cancer Evolution in Chronic Lymphocytic Leukemia David G. Kent and Anthony R. Green Noelia Purroy and Catherine J. Wu The Cellular Origin and Evolution of Breast Natural Selection in Cancer Biology: From Cancer Molecular Snowflakes to Trait Hallmarks Mei Zhang, Adrian V. Lee and Jeffrey M. Rosen Angelo Fortunato, Amy Boddy, Diego Mallo, et al.

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright © 2017 Cold Spring Harbor Laboratory Press; all rights reserved