<<

Published OnlineFirst November 12, 2018; DOI: 10.1158/0008-5472.CAN-18-1958 Review Research

Tolerance of Chromosomal Instability in Cancer: Mechanisms and Therapeutic Opportunities Eva Gronroos1 and Carlos Lopez-García 1,2

Abstract

Chromosomal instability (CIN) is the result of ongoing additional mechanisms have been identified in CIN-surveil- changes in the number () and structure of chromo- lance, resulting in a more complex network of pathways acting somes. CIN is induced by missegregation in independently or in cooperation with . Tolerance might also and leads to karyotypic diversity within the cancer cell be achieved by modifying aspects of the cancer cell physiology population, thereby adding to intratumor heterogeneity. in order to attenuate CIN or by adaptation to the consequences Regardless of the overall pro-oncogenic function of CIN, its of aneuploid . In this review, we summarize the onset is typically detrimental for cell fitness and thus tumors current knowledge about p53-dependent and -independent must develop CIN-tolerance mechanisms in order to propagate. mechanisms of CIN-tolerance in cancer, the adaptations There is overwhelming genetic and functional evidence linking observed in CIN cells buffering CIN levels, its consequences mutations in the tumor suppressor TP53 with CIN-tolerance. for cellular homeostasis, and the potential of exploiting However, the pathways leading to p53 activation following these adaptations in order to design new cancer therapies. chromosome missegregation remain controversial. Recently, Cancer Res; 78(23); 1–7. 2018 AACR.

Background missegregation events and generate sufficient karyotypic diver- sity within the population to confront the challenges that arise Most cancer types show some extent of chromosomal insta- during tumor evolution. In this review, we will focus on the bility (CIN). This hallmark of cancer, which can be defined as the current knowledge of the effects of CIN on cancer cell fitness and continual spatiotemporal alterations of the number and structure the mechanisms whereby cells override the deleterious effects of of , fuels intercellular heterogeneity and is a major chromosome segregation errors by inactivating CIN surveillance driving force of tumor evolution and adaptation to challenges pathways or adapting their physiology to the newly acquired arising from the tumor microenvironment. Numerical CIN chromosome imbalances. (nCIN) originates from missegregation of whole chromosomes in , resulting in two daughter cells with a differing from the natural karyotype of the species, also known as How Much CIN Can a Cancer Cell Tolerate? aneuploidy. However, segregation errors can also result in struc- During the course of CIN research, several controversies tural CIN (sCIN), giving rise to structural variants of chromo- about the role of CIN in cancer have been raised. CIN is very somes. Although both types of CIN originate from different frequent in cancer and it would be easy to assume that CIN mitotic (1–7) and premitotic alterations (8), they often co-occur operates as an oncogenic event with tumor promoting capa- and are interrelated (8–13). bilities. However, this conclusion is premature. Although extensive and detailed research has been carried out Induction of segregation errors in vivo by genetic inactivation of on the origin of segregation errors in cancer, the processes that components of the spindle assembly checkpoint (SAC) or spindle allow the propagation of CIN in tumors has not been subject to attachment proteins (21) has shown variable tumorigenesis the same level of research. Perhaps the most notable observa- when these mice are crossed onto cancer predisposed models. tion about CIN and its role in cancer is its apparently paradox- Induction of CIN shows oncogenic and tumor suppressive ical negative impact on cell fitness in vitro and in vivo (14–20). capacity depending on the models of carcinogenesis and the Thus, mechanisms of tolerance (denominated CIN-tolerance) CIN drivers considered. Explanations to address these differ- remain necessary in order to allow cancer cells to survive ences converge into a "just-right" model whereby only optimal levels of CIN can promote tumorigenesis. Very similar conclusions canbedrawnwhenhumancancers 1Translational Cancer Therapeutics Laboratory, Francis Crick Institute, London, are analyzed. Stratification of patients with breast cancer United Kingdom. 2National Heart and Lung Institute, Airway Disease Group, according to levels of CIN measured by centromeric FISH Faculty of Medicine, Imperial College London, London, United Kingdom. analysis (cell-to-cell variation) or computational surrogates of Corresponding Author: Carlos Lopez-García, Imperial College London, Guy CIN (weighted instability index) showed that tumors Scadding Building, Dovehouse Street, London SW3 6LY, UK. Phone: 44 (0) 20 with intermediate levels CIN (mid-CIN) had worse prognostic 7594 7846; Orchid: 0000-0001-9848-8216; E-mail: features than samples with high and low-CIN, confirming the [email protected] requirement of an intermediate level of CIN for cancer cell doi: 10.1158/0008-5472.CAN-18-1958 fitness (22, 23). A similar trend has also been shown in a pan- 2018 American Association for Cancer Research. cancer analysis (24). Although empirical evidence clearly point

www.aacrjournals.org OF1

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 12, 2018; DOI: 10.1158/0008-5472.CAN-18-1958

Gronroos and Lopez-García

at low- and high-CIN as good prognostic markers, a more mulation when cells are treated with SAC inhibitors (40). More detailed analysis is necessary to elucidate the cancer types that recently, in a model that used cold-shock to induce missegrega- are more sensitive to CIN and to design new therapeutic tion, it was observed that p53 accumulation was dependent on strategies to target cancer types that do not tolerate extreme H3.3 Ser31 phosphorylation on lagging chromosomes levels of CIN. (41). Finally, evidence drawn from SAC inactivation with Mps1 inhibitors showed that p53 accumulation was dependent on Tolerance of Chromosome Missegregation activation of caspase-2 (Fig. 1; ref. 42). Thus, it seems that multiple mechanisms to stabilize p53 after CIN exist and, in Cancer perhaps, different types of segregation errors, reflecting the The p53/MDM2 pathway various processes that can induce them, trigger p53 accumu- The most frequently mutated gene in cancer is TP53, encoding lation through different pathways. Taken into account that p53, a protein that prevents the propagation of genomically some of these mechanisms could occur concomitantly, the link unstable cells. The relationship between TP53 loss-of-function between p53 activation and CIN remains an open field that and CIN-tolerance in cancer derives primarily from the strong requires further insight. correlations found between TP53 mutations and aneuploidy in p53 also restricts the proliferation of tetraploid cells. Genome- multiple tumor types. In endometrial, gastric, and colorectal doubling (also known as tetraploidization) is common in CIN cancer, TP53-mutant tumors present more unstable and complex tumors and this process has been proposed as an intermediate karyotypes than TP53-proficient tumors (25–29). Notwithstand- stage in the development of CIN with high rates of missegregation ing this abundant genetic evidence, it is not possible to assign a and enhanced CIN tolerance (14, 43). Evidence drawn from CIN-tolerance function to TP53 mutations in all tumor types: for genome wide loss-of-function screens has documented a clear instance, microsatellite-unstable has a high connection between tolerance of tetraploidization and the fraction of TP53 mutations (10%–20%) while remaining dip- Hippo pathway (Fig. 1). Cytokinesis failure triggers activation loid, possibly reflecting the pleiotropic effects of p53. of this pathway with downstream phosphorylation of LATS2 Experimental cell biology and mouse models, additionally that specifically binds and inactivates MDM2, resulting in p53 support the role of p53 in CIN tolerance derived from cancer stabilization (44). genetics. Diploid cell lines treated with segregation error inducing drugs or bearing mutations in components of the SAC become Beyond p53: Other Mechanisms Driving aneuploid only in a TP53-mutant background (20, 30, 31). Also, ablation of TP53 in certain cancer predisposed mouse models CIN Tolerance promotes CIN and overall aneuploidy (32, 33). More recently, Although inactivation of TP53 seems to dominate the muta- genome-editing of human derived colon organoids revealed that tional landscape associated with CIN tolerance, TP53 wild-type when TP53 is mutated, organoids spontaneously develop CIN tumors are frequently aneuploid and therefore other mechanisms and a more aggressive phenotype (34). of tolerance are likely to take place. p53 is primarily regulated by a posttranslational mechanism BCL9L is one example of a CIN-tolerance gene that functions that involves MDM2, an ubiquitin-E3-ligase targeting p53 for either independently of, or in cooperation with p53. BCL9L is a proteasomal degradation. Although MDM2 is rarely mutated in Wnt signaling component that facilitates b-catenin transcriptional cancer, transgenic MDM2 expression in mouse models induces activity. Loss of BCL9L function in colorectal cancer reduces CIN (35) and MDM2 gene amplification and overexpression are the expression of caspase-2, an "orphan" caspase that can cleave common in some solid tumors and hematological malignancies substrates controlling cell viability such as the p53 regulator (36). However, the general role of MDM2 in tumorigenesis MDM2 and the pro-apoptotic protein BID (Fig. 1; ref. 42). Impor- and CIN is more complicated as this protein can also mediate tantly, this role of BCL9L as a CIN tolerance gene might shed light the degradation of other targets such as the tumor suppressor on the "just-right" theory of Wnt mediated tumorigenesis (45). Rb (37–39). An attenuation of Wnt signaling levels in cancer cells (e.g., by BCL9L dysfunction or selection of certain APC mutant alleles) Pathways leading to p53 activation could modulate the expression of caspase-2 to levels compatible Although the role of p53 in CIN-surveillance has been solidly with CIN. demonstrated, the mechanisms whereby p53 senses chromo- Dysfunction of caspase-2 has previously been shown to some missegregation require a better explanation. The p38 drive karyotypic alterations in various cancer predisposed stress kinase has been reported as necessary for p53-mediated models (46, 47) and to prevent p53 stabilization (46, 48, arrest in experimental models with cytoskeletal or centrosome 49). However, the connection between caspase-2 and BCL9L disruption (Fig. 1; ref. 20). Additionally, DNA damage follow- reinforced the relevance of caspase-2 for CIN tolerance in ing chromosome missegregation has also been proposed as the human tumors. Caspase-2 is activated by cleavage of the event triggering p53 stabilization. Janssen and colleagues (11) proenzyme generating the catalytically active p19 peptide. demonstrated that missegregated chromosomes are subject to Oliver and colleagues (48) proposed a p53-dependent mech- mechanical stress and DNA damage, leading to ATM-dependent anism whereby p53 transcriptionally activates PIDD, a protein p53 stabilization (Fig. 1). Other authors have postulated that mediating caspase-2 dimerization and self-cleavage. Thomp- micronuclei (extranuclear chromatin formed by missegregated son and colleagues (50) proposed a different mechanism chromosomes) are more prone to DNA damage than nuclear involving ATM dependent phosphorylation of PIDD that is DNA (10). But the dependency of p53-mediated arrest on DNA subsequently recruited by BubR1 to the thereby damage does not seem to apply in every experimental context. preventing caspase-2 activation and apoptosis. Andersen and For instance, inhibition of ATM does not prevent p53 accu- colleagues (51) also showed that phosphorylation of caspase-2

OF2 Cancer Res; 78(23) December 1, 2018 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 12, 2018; DOI: 10.1158/0008-5472.CAN-18-1958

CIN Tolerance in Cancer

Segregation error

Caspase-2 DNA damage p38 MAPK activation

BID MDM2 ATM cleavage cleavage phosphorylation

Apoptosis p21 Arrest Figure 1. Pathways involved in CIN-surveillance found p53 deregulated in cancer. The majority of pathways result in stabilization of p53 (blue), whereas some pathways operate independently of, or in cooperation with, p53 (red). MDM2 inhibition

Low Wild-type LATS2 Cyclin D1/D2 PIK3CA

Genome doubling

p53-dependent pathways p53-independent pathways

© 2018 American Association for Cancer Research

on serine 340 prevents caspase-2 activation during mitosis. As agation of aneuploid cells. For this reason, other mechanisms with p53 stabilization, modes of caspase-2 activation seem to of CIN tolerance have been proposed based on adaptations of be dependent on the experimental systems used and additional the cellular physiology that partially compensate mitotic altera- research is needed to fully elucidate the mechanisms of caspase-2 tions and the impact of chromosomal imbalances on cellular activation. homeostasis (Fig. 2). p53-independent mechanisms can also induce tolerance of tetraploidization. Two independent reports found that overex- CIN stabilization pression of cyclin D1 and D2, two cyclins frequently deregulated The proposed "just-right" level of CIN described above might in cancer, counteracted p53 activity by sequestering p21 and be the explanation for the intermediate levels of CIN found in thereby preventing p21-mediated arrest (Fig. 1; refs. 52, 53). patients with reduced survival and enhanced disease progres- Berenjeno and colleagues have also shown that oncogenic sion. This observation has motivated efforts to elucidate PIK3CA mutations mediate tolerance of tetraploidization in CIN modulatory mechanisms selected for in tumors. Loss-of- TP53-wild-type cells, although it is unclear whether this effect function mutations in the anaphase promoting complex/ involves a deficient p53 stabilization or a blockade of down- cyclosome complex (APC/C) have been recently found to stream effectors (54). counteract the effects of various mitotic alterations on mitotic fidelity (40, 55). The APC/C complex remains inhibited by the SAC until chromosomes achieve the appropriate biorientation Adaptation to CIN in Cancer Cells in metaphase. Hence, inactivation of the APC/C complex Although the mechanisms described above appear indis- results in a prolonged mitosis, enabling cells to correct mitotic pensable in many cancer types, newly acquired alterations, enhancing mitotic fidelity, and limiting CIN might still be detrimental for cell fitness and restrict the prop- (Fig. 2). Of note, a pan-cancer analysis showed a higher

www.aacrjournals.org Cancer Res; 78(23) December 1, 2018 OF3

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 12, 2018; DOI: 10.1158/0008-5472.CAN-18-1958

Gronroos and Lopez-García

APC/C Mitotic delay dysfunction (error correction)

Figure 2. Adaptations of the cellular physiology and homeostasis to attenuate the p62 effects of CIN. Top, APC/C dysfunction (autophagy) allows segregation error correction and prevents excessive CIN. Middle, Gene Protein aneuploid cells restrict the effects of Aneuploidy chromosome imbalances on the tolerance proteome by activating autophagy- Copy related protein p62. Bottom, transition Proteotoxic Protein number to near-triploid karyotypes enhances stress homeostasis fi gain cancer cell tness.

2N Near-triploid 3N transition 4N

© 2018 American Association for Cancer Research

frequency of mutations in APC/C subunits in aneuploid suggested that aneuploid cells mitigate the detrimental effects tumors, indicating that a longer mitosis is necessary to avoid of certain copy number changes by modulating the expression excessive genomic chaos (40). This "buffering" of CIN by of those genes or activating the degradation of the surplus regulating mitosis and enhancing error correction could be protein, to achieve levels compatible with cell survival. In selected in tumors as a mechanism that promotes the expan- particular, Hose and colleagues (62) documented a poor stoi- sion of cancer cells with intermediate and viable levels of CIN. chiometric correlation between mRNA and copy number in Of note, these studies predict that APC/C dysfunction could be aneuploid yeast strains. Chromosome imbalances also generate a mechanism of resistance to Mps1 inhibitors (BOS172722, changes in the stoichiometry of the cellular protein content, BAY1217389, BAY1161909, and CFI402257), a group of novel known as proteotoxic stress, that compromises the propagation anticancer drugs that induce CIN by inhibiting the SAC of CIN cells. The buffering of this process has been proposed as (56, 57), resulting in excessive CIN and cell death. a mechanism of propagation of aneuploidy. Seminal work by Inactivation of APC/C is not the only mechanism for CIN Torres and colleagues (63) reported that inactivation in the buffering. For instance, some cancer types have gene encoding the deubiquitinating enzyme Ubp6 increased attenuation in early phases of the disease, with the proliferation rates of aneuploid yeast and attenuated pro- reactivation at later stages thereby preventing excessive CIN tein changes caused by aneuploidy. (58–61). The relevance of these observations for mammalian cells was later confirmed with artificial introduction polysomy in Correcting the Consequences of CIN: near-diploid cancer cells. Transcriptomic analysis of viable Dosage Compensation and Protein polysomic strains showed a good correlation between copy number changes and mRNA abundance, whereas correlation of Homeostasis copy number with protein abundance showed protein levels CIN drives phenotypic evolution by altering dosage and similar to the parental near-diploid cells (64). Pathway analysis expression of genes located on imbalanced chromosomes of uncovered activation of the p62-dependent autophagic path- aneuploid cells. However, it is likely that subsets of gene dosage way, which conceivably contributedtocounteracttheeffect imbalances are advantageous for the aneuploid cell, whereas of copy number changes in protein homeostasis (Fig. 2). others impair cell fitness or are simply neutral. It has been Similarly, recent analysis of aneuploid cancer cells has shown

OF4 Cancer Res; 78(23) December 1, 2018 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 12, 2018; DOI: 10.1158/0008-5472.CAN-18-1958

CIN Tolerance in Cancer

that these cells modulate their phospho-proteome and the and NCT02792465, clinicaltrials.gov). However, the success of this microRNAome to adapt their physiology to the adverse effects strategy depends heavily on stratifying patients according to levels of aneuploidy (65, 66). Although evidence of proteome of CIN as treatment of tumors with low CIN might induce homeostasis as an aneuploidy maintenance mechanism has intermediate levels of CIN and a negative impact on patient been gained from in vitro systems, studies in breast cancer survival. Similarly, treatment of tumors bearing mutations in supports the notion that attenuation of the effect of chromo- CIN-tolerance genes such as TP53 and BCL9L should be carefully some imbalances on protein levels also occurs in human evaluated as they might reduce the therapeutic benefit. Thus, the tumors (67). success of Mps1 inhibitors will greatly depend on the develop- ment of methods to classify patients according to levels of CIN and existence of CIN-tolerance mutations in order to avoid Exploring Advantageous Karyotypes treatment of patients that will not benefit from treatment with Studies of the intrinsic tolerance of specifickaryotypesin these compounds. yeast and mammalian cells have also been performed; Pavelka Combination of Mps1 inhibitors with chemotherapy agents and colleagues (18) derived a wide range of aneuploid yeast must also be carefully evaluated. Conceivably, increasing strains from euploid laboratory strains. Most aneuploid strains genetic heterogeneity in tumors using Mps1 inhibitors might showed impaired fitness, but some aneuploid strains did not enhance drug resistance by selection of advantageous karyo- show any proliferation defects when compared with their types. However, combination of Mps1 inhibitors with low- euploid parental strains, supporting that certain karyotypes do dose taxanes resulted in a synergistic effect due to an increase not compromise cell fitness and can be tolerated. In contrast, in aberrant mitoses (57, 70). This synergy is currently being aneuploid strains showed a general growth advantage under tested in clinical trials and could lead to a reduction in taxane restrictive growth conditions or during treatment with drugs. doses, thereby preventing toxicity and drug-resistance. This report highlighted that the effect of aneuploidy on cell Although a good overall tolerability has already been con- fitness is not general, but context specific, and depends on firmed in vivo, with gastrointestinal and hematologic toxicity defined aneuploidies and selective pressures. as the most important toxicity effects (56, 57), the safety Similar approaches have been carried out in mammalian cells. profile of Mps1 inhibitors will be confirmed when the current Sheltzer and colleagues (68) generated a number of aneuploid trials are finally completed. Specifically, the long-term expan- mouse embryonic fibroblasts and aneuploid lines from diploid sion of de novo aneuploid cells in healthy tissues should be parental cell lines and showed that single chromosome trisomies carefully assessed. limit proliferation and tumorigenic capacity in the short term. The mechanisms whereby cells sense missegragation remain However, prolonged growth induced accumulation of additional an open question in the field of CIN-tolerance. A clear example chromosomal alterations and increased fitness and tumorigene- of this controversy is p53 accumulation. So far, the pathways sis. Regardless of the technical limitations of this elegant study, leading to p53 stabilization depend on the experimental model this report clearly shows the prominent short-term tumor sup- used to induce segregation errors. Perhaps, additional efforts to pressor role of aneuploidy in mammalian cells. Only when classify the types of missegregation that occur in tumors, additional karyotypes arise are cells able to explore optimal instead of cell lines, could clarify the elusive processes ulti- karyotypes, resulting in adaptation and tumorigenesis. mately driving p53 accumulation. A similar problem applies to The selection of karyotypes that are nondetrimental for caspase-2, where the mechanism of segregation error detection cell fitness has also been studied using a computational remains unclear. Recent evidence reporting that cytosolic approach that integrates the distribution of tumor suppressor dsDNA leaked from micronuclei is sensed by the NF-kBpath- genes and on human chromosomes (69). This way (71, 72), suggest that this system should be further approach showed that a near-triploid karyotype maximizes explored as a potential mechanism to detect missegregation cell fitness, which is supported by the high frequency of this and limit the propagation of CIN. An additional aspect of karyotypes in many cancer types (Fig. 2). The same study caspase-2 in the context of CIN tolerance, is its regulation by also predicted that this near-triploid karyotype is more easily Wnt-signaling. Therapies targeting this oncogenic pathway achieved from an initial tetraploid karyotype (Fig. 2), an ob- might lead to caspase-2 depletion rendering cells unprotected servation that is compatible with the frequency of genome- against CIN. A better understanding of the link between Wnt- doubling in cancer. signaling and caspase-2 activity will shed light on the potential shortcomings of targeting the Wnt-signaling pathway to treat cancer. Future Directions Understanding the levels of CIN tolerated by cancer cells, along with the mechanisms allowing cells to survive missegregation Disclosure of Potential Conflicts of Interest has an obvious interest in cancer therapeutics. Treatment with No potential conflicts of interest were disclosed. Mps1 inhibitors to induce toxic levels of CIN is a promising therapeutic approach currently being evaluated in four phase I Received June 25, 2018; revised August 24, 2018; accepted September 12, clinical trials (NCT03328494, NCT02366949, NCT02138812, 2018; published first November 12, 2018.

References 1. Bakhoum SF, Compton DA. Cancer: CINful centrosomes. Curr Biol 2009; 2. Bakhoum SF, Genovese G, Compton DA. Deviant kinetochore microtubule 19:R642–5. dynamics underlie chromosomal instability. Curr Biol 2009;19:1937–42.

www.aacrjournals.org Cancer Res; 78(23) December 1, 2018 OF5

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 12, 2018; DOI: 10.1158/0008-5472.CAN-18-1958

Gronroos and Lopez-García

3. Bakhoum SF, Thompson SL, Manning AL, Compton DA. Genome stability 26. Cancer Genome Atlas Research N, Kandoth C, Schultz N, Cherniack AD, is ensured by temporal control of kinetochore-microtubule dynamics. Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial Nat Cell Biol 2009;11:27–35. carcinoma. Nature 2013;497:67–73. 4. Cahill DP, Lengauer C, Yu J, Riggins GJ, Willson JK, Markowitz SD, et al. 27. Cianchi F, Balzi M, Becciolini A, Giache V, Messerini L, Palomba A, et al. Mutations of mitotic checkpoint genes in human . Nature 1998; Correlation between DNA content and p53 deletion in colorectal cancer. 392:300–3. Eur J Surg 1999;165:363–8. 5. Ertych N, Stolz A, Stenzinger A, Weichert W, Kaulfuss S, Burfeind P, et al. 28. Rowan A, Halford S, Gaasenbeek M, Kemp Z, Sieber O, Volikos E, et al. Increased microtubule assembly rates influence chromosomal instability Refining molecular analysis in the pathways of colorectal carcinogenesis. in colorectal cancer cells. Nat Cell Biol 2014;16:779–91. Clin Gastroenterol Hepatol 2005;3:1115–23. 6. Sotillo R, Hernando E, Diaz-Rodriguez E, Teruya-Feldstein J, Cordon- 29. Tang R, Changchien CR, Wu MC, Fan CW, Liu KW, Chen JS, et al. Colorectal Cardo C, Lowe SW, et al. Mad2 overexpression promotes aneuploidy and cancer without high microsatellite instability and chromosomal tumorigenesis in mice. Cancer Cell 2007;11:9–23. instability—an alternative genetic pathway to human colorectal 7. Vitre BD, Cleveland DW. Centrosomes, chromosome instability (CIN) and cancer. Carcinogenesis 2004;25:841–6. aneuploidy. Curr Opin Cell Biol 2012;24:809–15. 30. Li M, Fang X, Baker DJ, Guo L, Gao X, Wei Z, et al. The ATM-p53 pathway 8. Burrell RA, McClelland SE, Endesfelder D, Groth P, Weller MC, Shaikh N, suppresses aneuploidy-induced tumorigenesis. Proc Natl Acad Sci U S A et al. Replication stress links structural and numerical cancer chromosomal 2010;107:14188–93. instability. Nature 2013;494:492–6. 31. Soto M, Raaijmakers JA, Bakker B, Spierings DCJ, Lansdorp PM, 9. Chan KL, North PS, Hickson ID. BLM is required for faithful chromosome Foijer F, et al. p53 prohibits propagation of chromosome segregation segregation and its localization defines a class of ultrafine anaphase errors that produce structural aneuploidies. Cell Rep 2017;19: bridges. EMBO J 2007;26:3397–409. 2423–31. 10. Crasta K, Ganem NJ, Dagher R, Lantermann AB, Ivanova EV, Pan Y, et al. 32. Foijer F, Xie SZ, Simon JE, Bakker PL, Conte N, Davis SH, et al. Chromo- DNA breaks and chromosome pulverization from errors in mitosis. Nature some instability induced by Mps1 and p53 mutation generates aggressive 2012;482:53–8. lymphomas exhibiting aneuploidy-induced stress. Proc Natl Acad Sci U S A 11. Janssen A, van der Burg M, Szuhai K, Kops GJ, Medema RH. Chromosome 2014;111:13427–32. segregation errors as a cause of DNA damage and structural chromosome 33. Grim JE, Knoblaugh SE, Guthrie KA, Hagar A, Swanger J, Hespelt J, et al. aberrations. Science 2011;333:1895–8. Fbw7 and p53 cooperatively suppress advanced and chromosomally 12. Kawabata T, Luebben SW, Yamaguchi S, Ilves I, Matise I, Buske T, et al. unstable intestinal cancer. Mol Cell Biol 2012;32:2160–7. Stalled fork rescue via dormant replication origins in unchallenged S phase 34. Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, et al. promotes proper chromosome segregation and tumor suppression. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of Mol Cell 2011;41:543–53. human intestinal organoids. Nat Med 2015;21:256–62. 13. Pampalona J, Soler D, Genesca A, Tusell L. Whole chromosome loss is 35. Wang P, Lushnikova T, Odvody J, Greiner TC, Jones SN, Eischen CM. promoted by telomere dysfunction in primary cells. Genes Chromosomes Elevated Mdm2 expression induces chromosomal instability and confers a Cancer 2010;49:368–78. survival and growth advantage to B cells. 2008;27:1590–8. 14. Dewhurst SM, McGranahan N, Burrell RA, Rowan AJ, Gronroos E, 36. Onel K, Cordon-Cardo C. MDM2 and prognosis. Mol Cancer Res 2004; Endesfelder D, et al. Tolerance of whole-genome doubling propagates 2:1–8. chromosomal instability and accelerates cancer genome evolution. 37. Ganguli G, Wasylyk B. p53-independent functions of MDM2. Mol Cancer Cancer Discov 2014;4:175–85. Res 2003;1:1027–35. 15. Duncan AW, Hanlon Newell AE, Smith L, Wilson EM, Olson SB, Thayer MJ, 38. Bouska A, Lushnikova T, Plaza S, Eischen CM. Mdm2 promotes genetic et al. Frequent aneuploidy among normal human hepatocytes. Gastroen- instability and transformation independent of p53. Mol Cell Biol terology 2012;142:25–8. 2008;28:4862–74. 16. Faggioli F, Wang T, Vijg J, Montagna C. Chromosome-specific accumula- 39. Sdek P, Ying H, Chang DL, Qiu W, Zheng H, Touitou R, et al. MDM2 tion of aneuploidy in the aging mouse brain. Hum Mol Genet 2012;21: promotes proteasome-dependent ubiquitin-independent degradation of 5246–53. retinoblastoma protein. Mol Cell 2005;20:699–708. 17. Iwanaga Y, Chi YH, Miyazato A, Sheleg S, Haller K, Peloponese JM Jr., et al. 40. Sansregret L, Patterson JO, Dewhurst S,Lopez-Garcia C, Koch A,McGranahan Heterozygous deletion of mitotic arrest-deficient protein 1 (MAD1) N, et al. APC/C dysfunction limits excessive cancer chromosomal instability. increases the incidence of tumors in mice. Cancer Res 2007;67:160–6. Cancer Discov 2017;7:218–33. 18. Pavelka N, Rancati G, Zhu J, Bradford WD, Saraf A, Florens L, et al. 41. Hinchcliffe EH, Day CA, Karanjeet KB, Fadness S, Langfald A, Vaughan KT, Aneuploidy confers quantitative proteome changes and phenotypic et al. Chromosome missegregation during anaphase triggers p53 variation in budding yeast. Nature 2010;468:321–5. arrest through histone H3.3 Ser31 phosphorylation. Nat Cell Biol 2016; 19. Thompson SL, Compton DA. Examining the link between chromosom- 18:668–75. al instability and aneuploidy in human cells. J Cell Biol 2008;180: 42. Lopez-Garcia C, Sansregret L, Domingo E, McGranahan N, Hobor S, 665–72. Birkbak NJ, et al. BCL9L dysfunction impairs caspase-2 expression 20. Thompson SL, Compton DA. Proliferation of aneuploid human permitting aneuploidy tolerance in colorectal cancer. Cancer Cell 2017; cells is limited by a p53-dependent mechanism. J Cell Biol 2010; 31:79–93. 188:369–81. 43. Jamal-Hanjani M, Hackshaw A, Ngai Y, Shaw J, Dive C, Quezada S, et al. 21. Schvartzman JM, Sotillo R, Benezra R. Mitotic chromosomal instability and Tracking genomic cancer evolution for precision medicine: the lung TRA- cancer: mouse modelling of the human disease. Nat Rev Cancer 2010;10: CERx study. PLoS Biol 2014;12:e1001906. 102–15. 44. Ganem NJ, Cornils H, Chiu SY, O'Rourke KP, Arnaud J, Yimlamai D, et al. 22. Birkbak NJ, Eklund AC, Li Q, McClelland SE, Endesfelder D, Tan P, et al. Cytokinesis failure triggers hippo tumor suppressor pathway activation. Paradoxical relationship between chromosomal instability and survival Cell 2014;158:833–48. outcome in cancer. Cancer Res 2011;71:3447–52. 45. Albuquerque C, Breukel C, van der Luijt R, Fidalgo P, Lage P, Slors FJ, et al. 23. Roylance R, Endesfelder D, Gorman P, Burrell RA, Sander J, Tomlinson I, The 'just-right' signaling model: APC somatic mutations are selected based et al. Relationship of extreme chromosomal instability with long-term on a specific level of activation of the beta-catenin signaling cascade. survival in a retrospective analysis of primary breast cancer. Cancer Hum Mol Genet 2002;11:1549–60. Epidemiol Biomarkers Prev 2011;20:2183–94. 46. Dorstyn L, Puccini J, Wilson CH, Shalini S, Nicola M, Moore S, et al. 24. Andor N, Graham TA, Jansen M, Xia LC, Aktipis CA, Petritsch C, et al. Pan- Caspase-2 deficiency promotes aberrant DNA-damage response and genet- cancer analysis of the extent and consequences of intratumor heterogene- ic instability. Cell Death Differ 2012;19:1288–98. ity. Nat Med 2016;22:105–13. 47. Puccini J, Shalini S, Voss AK, Gatei M, Wilson CH, Hiwase DK, et al. Loss of 25. Cancer Genome Atlas Research N. Comprehensive molecular characteri- caspase-2 augments lymphomagenesis and enhances genomic instability zation of gastric adenocarcinoma. Nature 2014;513:202–9. in Atm-deficient mice. Proc Natl Acad Sci U S A 2013;110:19920–5.

OF6 Cancer Res; 78(23) December 1, 2018 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 12, 2018; DOI: 10.1158/0008-5472.CAN-18-1958

CIN Tolerance in Cancer

48. Oliver TG, Meylan E, Chang GP, Xue W, Burke JR, Humpton TJ, et al. 61. Rudolph KL, Millard M, Bosenberg MW, DePinho RA. Telomere dysfunc- Caspase-2-mediated cleavage of Mdm2 creates a p53-induced positive tion and evolution of intestinal carcinoma in mice and humans. Nat Genet feedback loop. Mol Cell 2011;43:57–71. 2001;28:155–9. 49. Terry MR, Arya R, Mukhopadhyay A, Berrett KC, Clair PM, Witt B, et al. 62. Hose J, Yong CM, Sardi M, Wang Z, Newton MA, Gasch AP. Dosage Caspase-2 impacts lung tumorigenesis and chemotherapy response in vivo. compensation can buffer copy-number variation in wild yeast. Elife Cell Death Differ 2015;22:719–30. 2015;4. doi: 10.7554/eLife.05462. 50. Thompson R, Shah RB, Liu PH, Gupta YK, Ando K, Aggarwal AK, et al. An 63. Torres EM, Dephoure N, Panneerselvam A, Tucker CM, Whittaker CA, Gygi inhibitor of PIDDosome formation. Mol Cell 2015;58:767–79. SP, et al. Identification of aneuploidy-tolerating mutations. Cell 2010;143: 51. Andersen JL, Johnson CE, Freel CD, Parrish AB, Day JL, Buchakjian MR, 71–83. et al. Restraint of apoptosis during mitosis through interdomain phos- 64. Stingele S, Stoehr G, Peplowska K, Cox J, Mann M, Storchova Z. Global phorylation of caspase-2. EMBO J 2009;28:3216–27. analysis of genome, transcriptome and proteome reveals the response to 52. Crockford A, Zalmas LP, Gronroos E, Dewhurst SM, McGranahan N, aneuploidy in human cells. Mol Syst Biol 2012;8:608. Cuomo ME, et al. Cyclin D mediates tolerance of genome-doubling in 65. Durrbaum M, Kruse C, Nieken KJ, Habermann B, Storchova Z. The cancers with functional p53. Ann Oncol 2016;28:149–56. deregulated microRNAome contributes to the cellular response to aneu- 53. Potapova TA, Seidel CW, Box AC, Rancati G, Li R. Transcriptome analysis of . BMC Genomics 2018;19:197. tetraploid cells identifies cyclin D2 as a facilitator of adaptation to genome 66. Vigano C, von Schubert C, Ahrne E, Schmidt A, Lorber T, Bubendorf L, et al. doubling in the presence of p53. Mol Biol Cell 2016;27:3065–84. Quantitative proteomic and phosphoproteomic comparison of human 54. Berenjeno IM, Pineiro R, Castillo SD, Pearce W, McGranahan N, Dewhurst colon cancer DLD-1 cells differing in ploidy and chromosome stability. SM, et al. Oncogenic PIK3CA induces centrosome amplification and Mol Biol Cell 2018;29:1031–47. tolerance to genome doubling. Nat Commun 2017;8:1773. 67. Myhre S, Lingjaerde OC, Hennessy BT, Aure MR, Carey MS, Alsner J, 55. Thu KL, Silvester J, Elliott MJ, Ba-Alawi W, Duncan MH, Elia AC, et al. et al. Influence of DNA copy number and mRNA levels on the Disruption of the anaphase-promoting complex confers resistance to TTK expression of breast cancer related proteins. Mol Oncol 2013;7: inhibitors in triple-negative breast cancer. Proc Natl Acad Sci U S A 1704–18. 2018;115:E1570–E7. 68. Sheltzer JM, Ko JH, Replogle JM, Habibe Burgos NC, Chung ES, Meehl CM, 56. Mason JM, Wei X, Fletcher GC, Kiarash R, Brokx R, Hodgson R, et al. et al. Single-chromosome gains commonly function as tumor suppressors. Functional characterization of CFI-402257, a potent and selective Mps1/ Cancer Cell 2016;31:240–1. TTK kinase inhibitor, for the treatment of cancer. Proc Natl Acad Sci U S A 69. Laughney AM, Elizalde S, Genovese G, Bakhoum SF. Dynamics of tumor 2017;114:3127–32. heterogeneity derived from clonal karyotypic evolution. Cell Rep 2015;12: 57. Wengner AM, Siemeister G, Koppitz M, Schulze V, Kosemund D, Klar U, 809–20. et al. Novel Mps1 kinase inhibitors with potent antitumor activity. Mol 70. Maia ARR, Linder S, Song JY, Vaarting C, Boon U, Pritchard CEJ, et al. Mps1 Cancer Ther 2016;15:583–92. inhibitors synergise with low doses of taxanes in promoting tumour cell 58. Chin K, de Solorzano CO, Knowles D, Jones A, Chou W, Rodriguez EG, death by enhancement of errors in cell division. Br J Cancer et al. In situ analyses of in breast cancer. Nat Genet 2018;118:1586–95. 2004;36:984–8. 71. Bakhoum SF, Ngo B, Laughney AM, Cavallo JA, Murphy CJ, Ly P, et al. 59. Feldmann G, Beaty R, Hruban RH, Maitra A. Molecular genetics of pancreatic Chromosomal instability drives through a cytosolic DNA intraepithelial neoplasia. J Hepatobiliary Pancreat Surg 2007;14:224–32. response. Nature 2018;553:467–72. 60. Meeker AK, Hicks JL, Platz EA, March GE, Bennett CJ, Delannoy MJ, et al. 72. Mackenzie KJ, Carroll P, Martin CA, Murina O, Fluteau A, Simpson DJ, et al. Telomere shortening is an early somatic DNA alteration in human prostate cGAS surveillance of micronuclei links genome instability to innate immu- tumorigenesis. Cancer Res 2002;62:6405–9. nity. Nature 2017;548:461–5.

www.aacrjournals.org Cancer Res; 78(23) December 1, 2018 OF7

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 12, 2018; DOI: 10.1158/0008-5472.CAN-18-1958

Tolerance of Chromosomal Instability in Cancer: Mechanisms and Therapeutic Opportunities

Eva Gronroos and Carlos López-García

Cancer Res Published OnlineFirst November 12, 2018.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-18-1958

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2018/11/09/0008-5472.CAN-18-1958. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2018 American Association for Cancer Research.