Skip to main content
  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Molecular Cancer Therapeutics
Molecular Cancer Therapeutics
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Review

Engaging Anaphase Catastrophe Mechanisms to Eradicate Aneuploid Cancers

Masanori Kawakami, Lisa Maria Mustachio, Xi Liu and Ethan Dmitrovsky
Masanori Kawakami
1Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lisa Maria Mustachio
1Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xi Liu
1Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ethan Dmitrovsky
1Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
2Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
3Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: edmitrovsky@mdanderson.org
DOI: 10.1158/1535-7163.MCT-17-1108 Published April 2018
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Cancer cells often have supernumerary centrosomes that promote genomic instability, a pathognomonic feature of cancer. During mitosis, cancer cells with supernumerary centrosomes undergo bipolar cell division by clustering centrosomes into two poles. When supernumerary centrosome clustering is antagonized, cancer cells are forced to undergo multipolar division leading to death of daughter cells. This proapoptotic pathway, called anaphase catastrophe, preferentially eliminates aneuploid cancer cells and malignant tumors in engineered mouse models. Anaphase catastrophe occurs through the loss or inhibition of the centrosomal protein CP110, a direct cyclin-dependent kinase 1 (CDK1) and CDK2 target. Intriguingly, CP110 is repressed by the KRAS oncoprotein. This sensitizes KRAS-driven lung cancers (an unmet medical need) to respond to CDK2 inhibitors. Anaphase catastrophe-inducing agents like CDK1 and CDK2 antagonists are lethal to cancer cells with supernumerary centrosomes, but can relatively spare normal cells with two centrosomes. This mechanism is proposed to provide a therapeutic window in the cancer clinic following treatment with a CDK1 or CDK2 inhibitor. Taken together, anaphase catastrophe is a clinically tractable mechanism that promotes death of neoplastic tumors with aneuploidy, a hallmark of cancer. Mol Cancer Ther; 17(4); 724–31. ©2018 AACR.

Introduction

For duplicated chromosomes to segregate faithfully into two daughter cells during cell division, proper bipolar spindle formation is required (1, 2). There must be precisely two centrosomes that serve as spindle poles during cell mitosis (1, 2). While centrosome numbers are tightly controlled in normal cells, aberrant centrosome numbers are detected as frequent features of both solid and hematological cancers (3–7). This is associated with genomic instability (8), a hallmark of cancer (9, 10). Excessive centrosomes in cancer cells lead to multipolar spindle assembly, causing asymmetric chromosome segregation and aneuploidy in daughter cells after multipolar cell division (4, 11–13). This contributes to tumor initiation or evolution (14–19) by increasing the proliferative advantage of some cellular populations through the loss of a chromosome domain that contains tumor suppressor genes or by gain of a region containing oncogenes (8, 20–23). The appearance of supernumerary centrosomes is associated with tumor progression and an unfavorable clinical outcome (7, 11). This links centrosome alterations to cancer progression (24).

There is also another important aspect of the biology of supernumerary centrosomes. Daughter cancer cells with excessive aneuploidy after multipolar cell division of parental cells with supernumerary centrosomes can compromise their survival (4, 25–27). Cancer cells circumvent these detrimental effects through several mechanisms. One involves clustering of supernumerary centrosomes into two spindle poles during mitosis so that they preserve bipolar spindle assembly and engage bipolar mitosis (25, 28–31). Several other pathways and regulators are involved in this process including motor proteins, centrosomal proteins, kinetochore proteins, spindle assembly checkpoint proteins, microtubule-associated proteins, and components of the actin cytoskeleton (25, 29, 32, 33).

Herein, we describe a distinct type of mitotic catastrophe called anaphase catastrophe. This is conferred by inhibition of centrosome clustering in cells with supernumerary centrosomes. It causes death of daughter cells after forcing them to undergo multipolar cell division (34, 35). As supernumerary centrosomes are not typically found in normal diploid cells with some exceptions like polyploid hepatocytes (36, 37), targeting centrosome clustering would theoretically affect only chromosomally unstable cancer cells while sparing normal cells from exhibiting anaphase catastrophe (25, 33–35, 38, 39). Given this, inducing anaphase catastrophe is an attractive strategy to explore for cancer therapy (35, 40–42). Interestingly, several agents that antagonize cyclin-dependent kinase 1 (CDK1) or CDK2 activities were reported to cause anaphase catastrophe in cancer cells (34, 43, 44).

In this review, the molecular mechanisms that are the basis for activating anaphase catastrophe following CDK2 or CDK1 antagonism are presented. Anaphase catastrophe confers apoptotic death of cancer cells while relatively sparing normal cells, as will be discussed. Intriguingly, this mechanism is preferentially engaged in KRAS-mutant oncoprotein-expressing lung cancer (34, 35, 44). The mechanistic basis for this association will be discussed. Furthermore, the translational research implications of this finding will be highlighted.

Anaphase Catastrophe

CDK1 or CDK2 inhibition induces anaphase catastrophe

The association between anaphase catastrophe and CDK2 inhibition was first uncovered after treating lung cancers with seliciclib (CYC202, Cyclacel), an orally bioavailable and fully reversible inhibitor of CDK2 activity with less evident effects against CDK5, CDK7, and CDK9 activity (IC50 for CDK2: 100 nmol/L, CDK5: 160 nmol/L, CDK7: 540 nmol/L, and CDK9: 950 nmol/L; refs. 34, 35, 44–47). When aneuploid lung cancer cells were exposed to seliciclib, irreversible antiproliferative effects were unexpectedly observed (34). In the pursuit of a mechanism responsible for these surprising irreversible actions, seliciclib was found to promote multipolar spindle formation during cell mitosis (34). This process does not disrupt the temporal sequence of cell mitosis, but forces cells to undergo multipolar cell division leading to apoptosis of daughter cells (34, 35, 48), as shown in Fig. 1A. Because apoptosis was triggered by this aberrant mitosis after anaphase, this proapoptotic death program was called anaphase catastrophe (34, 35). Live-cell imaging and cytochrome C staining of the progeny of seliciclib-treated cells revealed that affected cells with multipolar anaphases underwent apoptosis (43, 48). This indicated that anaphase catastrophe caused apoptosis after engaging cell division (48).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Summary of anaphase catastrophe. A, The flow of mitosis with three spindle poles is shown to represent multipolar mitosis with normal bipolar mitosis for comparison. Although the temporal sequence of mitosis is maintained, chromosomes segregate inappropriately in multipolar cell mitosis, leading to apoptotic death of daughter cells. B, CDK1 or CDK2 inhibition each can antagonize clustering of supernumerary centrosomes into two poles. This forces cells to undergo multipolar cell division. This leads to apoptotic death of daughter cells, known as anaphase catastrophe. The arrow size displays the relative extent of the indicated engaged pathway. The red rectangle indicates CDK1 or CDK2 inhibition blocks centrosome clustering. C, There are three types of indicated cancer cells identified after treatment with CDK2 inhibitors: normal cells with two centrosomes and spindles (N cells), cells with clustered supernumerary centrosomes with bipolar spindles (C cells), and cells with supernumerary centrosomes and multipolar spindles (M cells). Left, Representative immunofluorescence images of these different cell types and the respective schematic diagrams are displayed. Staining with DAPI (blue), α-tubulin (red), and γ-tubulin (green) are each shown. Right, Pie charts indicate representative cell population changes for N cells, C cells, and M cells after treatment with a CDK2 inhibitor. Although cell populations of these cell types can differ between examined cancer cells, C cells are typically decreased and M cells are increased while the total cell population of cells with supernumerary centrosomes (C + M cells) is often not appreciably changed after treatment with a CDK2 inhibitor, as shown in these pie charts.

The major molecular pharmacologic target of seliciclib is CDK2 activity (45–47). Given this, it was hypothesized that CDK2 inhibition was responsible for conferring anaphase catastrophe. To study the direct and specific effects of CDK2 inhibition, CDK2 was genetically targeted for repression by transfection of siRNAs into lung cancer cells. Anaphase catastrophe was observed by this knockdown of CDK2 and this finding independently confirmed the pharmacologic effects of CDK2 antagonists (34, 44). In addition to seliciclib, anaphase catastrophe engagement was observed using other selective CDK2 or pan-CDK inhibitors, that include dinaciclib (SCH727965, Merck; IC50 for CDK1: 3 nmol/L, CDK2: 1 nmol/L, CDK5: 1 nmol/L, and CDK9: 4 nmol/L; refs. 43, 49), CCT68127 (Cyclacel; IC50 for CDK2: 30 nmol/L and CDK9: 110 nmol/L; ref. 44), and CYC065 (PubChem ID: 24983461, Cyclacel; IC50 for CDK2: 5 nmol/L and CDK9: 26 nmol/L; ref. 44). Notably, anaphase catastrophe induced after CDK2 inhibitor treatment was observed not only in lung cancer cells, but also in preliminary evidence from other aneuploid cancer cells (unpublished observations from Ethan Dmitrovsky Laboratory). This indicates that anaphase catastrophe is a common pathway engaged in chromosomally unstable cancer cells.

Because specific inhibitors can affect multiple CDKs in addition to CDK2, the potential contribution of other CDKs in mediating anaphase catastrophe was investigated directly by downregulating CDK1, CDK5, and CDK9 individually using independent siRNAs. Knockdown of CDK1 was shown to augment anaphase catastrophe as did CDK2 repression (43).

Anaphase catastrophe occurs by inhibiting centrosome clustering

Because multipolar spindle formation is linked to the presence of supernumerary centrosomes, spindle formation and centrosome number were each analyzed by respective α-tubulin and γ-tubulin staining of cancer cells following treatment with vehicle or with specific CDK2 inhibitors (50). Using these methods, it was found that anaphase catastrophe occurs via inhibition of supernumerary centrosomes clustering after treatment with CDK2 inhibitors, as shown in Fig. 1B.

The examined lung cancer cells after the treatments were divided into three groups based on centrosome and spindle numbers at anaphase, as depicted in Fig. 1C. The first includes bipolar normal cells with two centrosomes and spindles (N cells). The second describes cells with supernumerary centrosomes clustered into two poles with bipolar spindles formation (C cells). In this type of cell, chromosomes are segregated equally, despite an aberrant number of centrosomes. The third represents cells with supernumerary centrosomes and multipolar spindle formation without centrosome clustering (M cells). This type of cell undergoes multipolar cell division and anaphase catastrophe is subsequently conferred.

It was uncovered that the total population of cells with supernumerary centrosomes (the sum of the number of C and M cells) was unaffected by treatment with a selective CDK2 inhibitor, as indicated in pie charts in Fig. 1C. This indicated that a CDK2 inhibitor does not cause de novo supernumerary centrosomes formation. However, the population of C cells, in which supernumerary centrosomes are clustered into two poles, was reduced and that of M cells, which have multipolar spindles without centrosome clustering, was correspondingly increased after the CDK2 inhibitor treatment, as depicted in the pie charts displayed in Fig. 1C. This revealed that a CDK2 inhibitor inhibits clustering of supernumerary centrosomes. Thus, CDK2 inhibitors cause anaphase catastrophe by opposing the clustering of preexisting supernumerary centrosomes and not by causing de novo supernumerary centrosomes to form. Therefore, anaphase catastrophe following CDK2 antagonism is detected preferentially in cells having supernumerary centrosomes while relatively sparing normal cells with two centrosomes. Consistent with this view, it was experimentally shown that anaphase catastrophe was not substantially observed after treatment of immortalized pulmonary epithelial cells that are not chromosomally unstable as are lung cancer cells (34, 44). Normal cells like hematopoietic and gastric cells are affected by conventional chemotherapy because of their proliferative properties. Yet, anaphase catastrophe engagement is expected to spare even these proliferating normal cells. This is because anaphase catastrophe is conferred by the presence of aneuploidy rather than the rate of proliferation of a cancer cell (44).

Inhibition of CP110 centrosome protein mediates anaphase catastrophe

To elucidate the mechanisms underlying induced anaphase catastrophe after CDK2 antagonism, proteins reported as substrates of CDK2 phosphorylation were screened to uncover candidate mediators of anaphase catastrophe conferred by CDK2 inhibitor treatment (48). Among these proteins, the centrosomal protein CP110 was functionally highlighted (48). Indeed, CP110 knockdown can trigger multipolar anaphases in examined cancer cells (48).

CP110 is a direct phosphorylation target of cyclin E/CDK2, cyclin A/CDK2, and cyclin B/CDK1 (51), as summarized in Fig. 2A. CP110 is not known to have enzymatic activity. However, it interacts with distinct protein complexes and regulates microtubule growth as well as centriole length (52–57). It plays pivotal roles in centrosome duplication (51), maturation (54), separation (51), and cytokinesis (58, 59). CP110 levels as well as its localization to the centrosome are each strictly controlled and depend on the phase of the cell cycle (51). During the G1–S cell-cycle phase, CP110 is prominently expressed coincident with initiation of centrosome duplication, peaking during the S cell-cycle phase; it is involved in centrosome duplication and maturation (51, 54). CP110 protein expression diminishes substantially during the G2–M and G0–G1 cell-cycle phases. During M phase, CP110 regulates centrosome separation and cytokinesis (58, 59). CP110 knockdown or the mutation of its CDK2 phosphorylation sites each results in the unscheduled separation of centrosomes (51). From these findings, it is proposed that CP110 positively regulates centrosome duplication and inhibits premature centrosome separation. CP110 is also known to suppress primary cilia formation in noncycling cells and cells in the G0 cell-cycle phase (59–61).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

CP110 is a mediator of anaphase catastrophe. A, CP110 is phosphorylated by CDK1 or CDK2. B, The ten potential CDK2 phosphorylation sites within the CP110 amino acid sequence are displayed. Critical phosphorylation sites engaged in triggering anaphase catastrophe after CDK2 antagonism include residues serine 170 and threonine 194. These are highlighted with rectangles. C, CP110 protein expression is regulated through proteasomal degradation after ubiquitination via the SCFcyclin F ubiquitin ligase complex. D, In KRAS-mutant oncoprotein-expressing lung cancer cells, high levels of cyclin F protein, the F-box protein of SCFcyclin F, promote ubiquitination and thereby proteasomal degradation of CP110 protein. This leads to reduced levels of CP110 protein as compared with KRAS wild-type expressing cancer cells. The font size is meant to display the relative contribution of the highlighted species. The arrow size displays the relative extent of the indicated engaged pathways.

CP110 is involved in centrosome function, especially centrosome separation, and it is a CDK1 and CDK2 phosphorylation target (51). Given this, CP110 was hypothesized to control supernumerary centrosome clustering and to trigger anaphase catastrophe after CDK2 antagonism (48). Engineered knockdown of CP110 conferred anaphase catastrophe and augmented this response after treatment with the CDK2 inhibitor, seliciclib (48). As expected, gain of CP110 expression antagonized this inhibition of centrosome clustering as well as anaphase catastrophe induction caused by either genetic or pharmacological antagonism of CDK2 (48). These findings were confirmed by use of different individual or combined CDK1 or CDK2 pharmacologic inhibitors including seliciclib (48), dinaciclib (43), CCT68127 (44), and CYC065 (44). Also, expression of a mutant CP110 species with all potential CDK2 phosphorylation sites replaced by alanine residues did not antagonize consequences of CDK2 inhibition (48). Based on these findings, it was hypothesized (48) that CDK2 antagonism disrupts effective clustering of supernumerary centrosomes via inhibition of CP110 phosphorylation, causing anaphase catastrophe.

There are ten potential CDK2 phosphorylation sites present in CP110. The role of each of these potential phosphorylation sites in CDK2 inhibitor–mediated anaphase catastrophe was interrogated by transversing each or all of these sites with alanine residues within CP110 expression vector constructs and independently by individually restoring these mutated sites to their respective wild-type sequences containing serine or threonine residues (50). Based on these experiments, serine 170 and threonine 194 residues were found as the sites responsible for causing anaphase catastrophe after CDK2 inhibition (50), as summarized in Fig. 2B. It was also confirmed that these two sites were involved in the inhibition of centrosome clustering after CDK2 antagonism (50). CP110 serine 170 and threonine 194 residues are located between the CP110 coiled coil and destruction box (D-box) domains (51) as shown in Fig. 2B. These sites are likely important for interactions between CP110 and other centrosomal proteins that control centrosome clustering.

CP110 expression and the KRAS oncoprotein

CP110 protein levels are tightly controlled during the cell cycle to prevent errors in centrosome duplication, separation, or cytokinesis (51). CP110 expression is reduced at the early G1 cell-cycle phase and is increased during the G1–S transition (51). These levels begin to decline at the G2 cell-cycle phase and diminish after cell mitosis (51). Ubiquitination mechanisms are engaged in regulating this CP110 protein expression (62, 63). During the G2 phase of the cell cycle, CP110 associates with the F-box protein cyclin F and is ubiquitylated via the SCF (Skp1-Cul1-F-box protein)cyclin F ubiquitin ligase complex and it is then degraded (62, 63), as shown in Fig. 2C. The siRNA-mediated depletion of cyclin F in the G2 cell-cycle phase causes centrosome and mitotic abnormalities that are reversed by cosilencing CP110 (62). This indicates that SCFcyclinF-mediated degradation of CP110 is necessary for proper mitosis and genomic integrity (62). Besides cyclin F, Neuralized homolog 4 (Neurl4), a member of Neuralized family, and EDD-DYRK2-DDB1VprBP, an E3 ligase, can also regulate CP110 expression, respectively, through ubiquitination (64–66).

Deubiquitination is critical for regulating CP110 protein expression (63). The ubiquitin-specific protease 33 (USP33), a deubiquitinating enzyme, deubiquitinates CP110 in a cell-cycle–dependent manner, thereby counteracting SCFcyclinF-mediated ubiquitination (67). Ablation of USP33 destabilizes CP110 protein and by this inhibits centrosome amplification (67). The mitotic defects caused by siRNA-mediated depletion of SCFcyclinF are reduced by codepletion of USP33 (67). Pertinent to the molecular pharmacologic role of CP110, USP33 knockdown enhanced anaphase catastrophe caused by CDK2 inhibition (50).

Intriguingly, in KRAS-mutant versus wild-type expressing lung cancer cells, the basal protein expression of cyclin F was detected as markedly elevated (50). Cyclin F is a key negative regulator of CP110 protein expression (62). Gain of expression of the KRAS oncoprotein led to reduced expression of CP110 protein (50), as summarized in Fig. 2D. The direct effect of the KRAS oncoprotein on cyclin F and CP110 proteins was experimentally shown by loss of KRAS expression by use of siRNAs (50). When KRAS was repressed in lung cancer cells, cyclin F expression decreased and CP110 expression subsequently increased (50). This indicated that the KRAS oncoprotein downregulated CP110 protein by upregulating cyclin F expression (50). The precise mechanism through which the KRAS oncoprotein upregulates cyclin F expression is not yet elucidated, but cyclin F expression is affected by DNA damage and repair pathways (68, 69). This links cyclin F expression to the KRAS oncoprotein that can confer intrinsic genotoxic stress to cancer cells (70).

CP110 immunohistochemical analysis revealed that both engineered murine lung cancer models and human lung cancer cases that expressed the KRAS oncoprotein substantially lowered CP110 expression as compared with KRAS wild-type expressing lung cancers (48). KRAS mutations are linked to centrosome amplification and chromosomal instability (71–73). These associations provide a mechanistic basis for the observed link between destabilization of CP110 protein and KRAS oncoprotein expression in lung cancer and potentially other cancer cell contexts (44).

Translational perspectives

Mutant KRAS species regulate expression of CP110 protein. In turn, CP110 is a mediator of anaphase catastrophe caused by CDK2 inhibition. Given this, the effect of KRAS mutations on activities of CDK2 inhibitors in cancers was analyzed. Notably, lung cancer cells with KRAS mutations were particularly sensitive to CDK2 inhibition (34, 43, 44). High throughput pharmacogenomic analyses revealed that KRAS mutations expressed in lung cancer cells led to a statistically significantly more sensitive response to seliciclib (34) and CCT68127 (44) than KRAS wild-type expressing lung cancer cells. Because CP110 expression is reduced in KRAS-mutant lung cancer cells (48), CDK2 inhibitors are proposed to inhibit CP110 activity more readily in these cancer cells. This is proposed to confer an efficient induction of anaphase catastrophe, as shown in Fig. 3.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Anaphase catastrophe induction in KRAS oncoprotein-driven cancer cells. In KRAS-mutant oncoprotein-expressing lung cancer cells, basal expression levels of CP110 protein are reduced. As a result, anaphase catastrophe is readily conferred after inhibiting CP110 activity by use of a CDK2 inhibitor. This increases sensitivity of KRAS-mutant lung cancer cells to treatment with a CDK1 or CDK2 inhibitor. The font size is meant to convey the relative contribution of the highlighted species. The arrow size displays the relative extent of the indicated engaged pathway.

This hypothesis has translational research implications because the KRAS oncogene is often activated via mutation within diverse cancers. Likewise, the KRAS oncoprotein has been a challenging pharmacologic target; it is also associated with a poor clinical prognosis (74–77). This makes lung cancers driven by the KRAS oncoprotein an unmet medical need for innovative therapy (76, 78). KRAS mutations are present in nearly 30% of lung cancers and are especially common in smokers, who develop pulmonary adenocarcinomas (74). These alterations frequently confer resistance to chemotherapeutic agents that are otherwise effective against KRAS wild-type cancers (75, 79, 80). Despite the considerable efforts already invested in targeting KRAS-mutant cancers, this has not yet translated into clinical advances.

New insights are needed to address this refractory cancer problem (81). In this regard, the observed preclinical response of KRAS-mutant lung cancers to CDK2 inhibitors has the prospect of producing substantial clinical benefits (34, 35, 44). Thus, clinical trials using either CDK1 or CDK2 inhibitors especially for combating KRAS-mutant lung cancer cases are warranted. Because KRAS mutations are detected at a high frequency in other clinically challenging cancers like pancreatic and colon cancers (82), the pharmacologic engagement of anaphase catastrophe following CDK1 or CDK2 inhibition could provide a new way to target broadly KRAS mutant–driven cancers. Added support for this view comes from in vivo evidence that found CDK2 inhibition substantially repressed growth of syngeneic murine lung cancers whether or not these models expressed wild-type or mutant KRAS species (34, 35, 44). Normal bipolar cells are less sensitive to the consequence of anaphase catastrophe than are aneuploid cells. Thus, anaphase catastrophe induction after treatment with a CDK1 or CDK2 inhibitor has a clinically relevant therapeutic window.

Conclusions

This review summarized molecular insights and clinical implications of a previously unrecognized proapoptotic death program known as anaphase catastrophe. It preferentially eradicates aneuploid cancer cells with supernumerary centrosomes while relatively sparing bipolar cells. Specific CDK antagonists inhibit supernumerary centrosome clustering and trigger anaphase catastrophe via reduced phosphorylation of the centrosomal protein CP110, a direct CDK1 and CDK2 target (48). Notably, CP110 protein is regulated by the KRAS oncoprotein (50). Given this, induced anaphase catastrophe following treatment with CDK2 (or CDK1) inhibitors has therapeutic implications for otherwise therapeutically refractory cancers that harbor KRAS mutations, such as lung cancers. These antineoplastic agents engage a previously unrecognized mechanism that is worthy of study in the laboratory and in cancer patients.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

We thank all members of the Dmitrovsky Laboratory for their helpful consultation. This work was supported by National Institutes of Health (NIH) and National Cancer Institute (NCI) grants R01-CA087546 (E. Dmitrovsky) and R01-CA190722 (E. Dmitrovsky), a Samuel Waxman Cancer Research Foundation Award (E. Dmitrovsky), a UT-STARs award (E. Dmitrovsky), and an American Cancer Society Clinical Research Professorship (E. Dmitrovsky).

  • Received November 13, 2017.
  • Revision received January 16, 2018.
  • Accepted February 16, 2018.
  • Published first March 20, 2018.
  • ©2018 American Association for Cancer Research.

References

  1. 1.↵
    1. Bornens M
    . Centrosome composition and microtubule anchoring mechanisms. Curr Opin Cell Biol 2002;14:25–34.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Bettencourt-Dias M,
    2. Glover DM
    . Centrosome biogenesis and function: centrosomics brings new understanding. Nat Rev Mol Cell Biol 2007;8:451–63.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Lingle WL,
    2. Lutz WH,
    3. Ingle JN,
    4. Maihle NJ,
    5. Salisbury JL
    . Centrosome hypertrophy in human breast tumors: implications for genomic stability and cell polarity. Proc Natl Acad Sci U S A 1998;95:2950–5.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Nigg EA
    . Centrosome aberrations: cause or consequence of cancer progression? Nat Rev Cancer 2002;2:815–25.
    OpenUrlCrossRefPubMed
  5. 5.↵
    1. Kramer A,
    2. Neben K,
    3. Ho AD
    . Centrosome replication, genomic instability and cancer. Leukemia 2002;16:767–75.
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. Zyss D,
    2. Gergely F
    . Centrosome function in cancer: guilty or innocent? Trends Cell Biol 2009;19:334–46.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Chan JY
    . A clinical overview of centrosome amplification in human cancers. Int J Biol Sci 2011;7:1122–44.
    OpenUrlCrossRefPubMed
  8. 8.↵
    1. Ganem NJ,
    2. Godinho SA,
    3. Pellman D
    . A mechanism linking extra centrosomes to chromosomal instability. Nature 2009;460:278–82.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Hanahan D,
    2. Weinberg RA
    . The hallmarks of cancer. Cell 2000;100:57–70.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Hanahan D,
    2. Weinberg RA
    . Hallmarks of cancer: the next generation. Cell 2011;144:646–74.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Pihan GA,
    2. Purohit A,
    3. Wallace J,
    4. Knecht H,
    5. Woda B,
    6. Quesenberry P,
    7. et al.
    Centrosome defects and genetic instability in malignant tumors. Cancer Res 1998;58:3974–85.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    1. Giehl M,
    2. Fabarius A,
    3. Frank O,
    4. Hochhaus A,
    5. Hafner M,
    6. Hehlmann R,
    7. et al.
    Centrosome aberrations in chronic myeloid leukemia correlate with stage of disease and chromosomal instability. Leukemia 2005;19:1192–7.
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Chng WJ,
    2. Ahmann GJ,
    3. Henderson K,
    4. Santana-Davila R,
    5. Greipp PR,
    6. Gertz MA,
    7. et al.
    Clinical implication of centrosome amplification in plasma cell neoplasm. Blood 2006;107:3669–75.
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    1. Godinho SA,
    2. Pellman D
    . Causes and consequences of centrosome abnormalities in cancer. Philos Trans R Soc Lond B Biol Sci 2014;369.
  15. 15.↵
    1. Nigg EA,
    2. Cajanek L,
    3. Arquint C
    . The centrosome duplication cycle in health and disease. FEBS Lett 2014;588:2366–72.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Basto R,
    2. Brunk K,
    3. Vinadogrova T,
    4. Peel N,
    5. Franz A,
    6. Khodjakov A,
    7. et al.
    Centrosome amplification can initiate tumorigenesis in flies. Cell 2008;133:1032–42.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Raff JW,
    2. Basto R
    . Centrosome amplification and cancer: a question of sufficiency. Dev Cell 2017;40:217–8.
    OpenUrl
  18. 18.↵
    1. Nigg EA
    . Origins and consequences of centrosome aberrations in human cancers. Int J Cancer 2006;119:2717–23.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Lingle WL,
    2. Barrett SL,
    3. Negron VC,
    4. D'Assoro AB,
    5. Boeneman K,
    6. Liu W,
    7. et al.
    Centrosome amplification drives chromosomal instability in breast tumor development. Proc Natl Acad Sci U S A 2002;99:1978–83.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. Rajagopalan H,
    2. Lengauer C
    . Aneuploidy and cancer. Nature 2004;432:338–41.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Gao C,
    2. Furge K,
    3. Koeman J,
    4. Dykema K,
    5. Su Y,
    6. Cutler ML,
    7. et al.
    Chromosome instability, chromosome transcriptome, and clonal evolution of tumor cell populations. Proc Natl Acad Sci U S A 2007;104:8995–9000.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    1. Silkworth WT,
    2. Nardi IK,
    3. Scholl LM,
    4. Cimini D
    . Multipolar spindle pole coalescence is a major source of kinetochore mis-attachment and chromosome mis-segregation in cancer cells. PLoS ONE 2009;4:e6564.
    OpenUrlCrossRefPubMed
  23. 23.↵
    1. Beach RR,
    2. Ricci-Tam C,
    3. Brennan CM,
    4. Moomau CA,
    5. Hsu PH,
    6. Hua B,
    7. et al.
    Aneuploidy causes non-genetic individuality. Cell 2017;169:229–42 e21.
    OpenUrlCrossRef
  24. 24.↵
    1. Godinho SA,
    2. Picone R,
    3. Burute M,
    4. Dagher R,
    5. Su Y,
    6. Leung CT,
    7. et al.
    Oncogene-like induction of cellular invasion from centrosome amplification. Nature 2014;510:167–71.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Kwon M,
    2. Godinho SA,
    3. Chandhok NS,
    4. Ganem NJ,
    5. Azioune A,
    6. Thery M,
    7. et al.
    Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev 2008;22:2189–203.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    1. Colombo R,
    2. Caldarelli M,
    3. Mennecozzi M,
    4. Giorgini ML,
    5. Sola F,
    6. Cappella P,
    7. et al.
    Targeting the mitotic checkpoint for cancer therapy with NMS-P715, an inhibitor of MPS1 kinase. Cancer Res 2010;70:10255–64.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    1. Janssen A,
    2. Kops GJ,
    3. Medema RH
    . Elevating the frequency of chromosome mis-segregation as a strategy to kill tumor cells. Proc Natl Acad Sci U S A 2009;106:19108–13.
    OpenUrlAbstract/FREE Full Text
  28. 28.↵
    1. Ring D,
    2. Hubble R,
    3. Kirschner M
    . Mitosis in a cell with multiple centrioles. J Cell Biol 1982;94:549–56.
    OpenUrlAbstract/FREE Full Text
  29. 29.↵
    1. Quintyne NJ,
    2. Reing JE,
    3. Hoffelder DR,
    4. Gollin SM,
    5. Saunders WS
    . Spindle multipolarity is prevented by centrosomal clustering. Science 2005;307:127–9.
    OpenUrlAbstract/FREE Full Text
  30. 30.↵
    1. Brinkley BR
    . Managing the centrosome numbers game: from chaos to stability in cancer cell division. Trends Cell Biol 2001;11:18–21.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Godinho SA,
    2. Kwon M,
    3. Pellman D
    . Centrosomes and cancer: how cancer cells divide with too many centrosomes. Cancer Metastasis Rev 2009;28:85–98.
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. Leber B,
    2. Maier B,
    3. Fuchs F,
    4. Chi J,
    5. Riffel P,
    6. Anderhub S,
    7. et al.
    Proteins required for centrosome clustering in cancer cells. Sci Transl Med 2010;2:33ra8.
    OpenUrl
  33. 33.↵
    1. Fielding AB,
    2. Lim S,
    3. Montgomery K,
    4. Dobreva I,
    5. Dedhar S
    . A critical role of integrin-linked kinase, ch-TOG and TACC3 in centrosome clustering in cancer cells. Oncogene 2011;30:521–34.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Galimberti F,
    2. Thompson SL,
    3. Liu X,
    4. Li H,
    5. Memoli V,
    6. Green SR,
    7. et al.
    Targeting the cyclin E-cdk-2 complex represses lung cancer growth by triggering anaphase catastrophe. Clin Cancer Res 2010;16:109–20.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    1. Galimberti F,
    2. Thompson SL,
    3. Ravi S,
    4. Compton DA,
    5. Dmitrovsky E
    . Anaphase catastrophe is a target for cancer therapy. Clin Cancer Res 2011;17:1218–22.
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    1. Gentric G,
    2. Celton-Morizur S,
    3. Desdouets C
    . Polyploidy and liver proliferation. Clin Res Hepatol Gastroenterol 2012;36:29–34.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Gentric G,
    2. Desdouets C
    . Liver polyploidy: Dr Jekyll or Mr Hide? Oncotarget 2015;6:8430–1.
    OpenUrl
  38. 38.↵
    1. Rebacz B,
    2. Larsen TO,
    3. Clausen MH,
    4. Ronnest MH,
    5. Loffler H,
    6. Ho AD,
    7. et al.
    Identification of griseofulvin as an inhibitor of centrosomal clustering in a phenotype-based screen. Cancer Res 2007;67:6342–50.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. Raab MS,
    2. Breitkreutz I,
    3. Anderhub S,
    4. Ronnest MH,
    5. Leber B,
    6. Larsen TO,
    7. et al.
    GF-15, a novel inhibitor of centrosomal clustering, suppresses tumor cell growth in vitro and in vivo. Cancer Res 2012;72:5374–85.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Gergely F,
    2. Basto R
    . Multiple centrosomes: together they stand, divided they fall. Genes Dev 2008;22:2291–6.
    OpenUrlAbstract/FREE Full Text
  41. 41.↵
    1. Gascoigne KE,
    2. Taylor SS
    . How do anti-mitotic drugs kill cancer cells? J Cell Sci 2009;122(Pt 15):2579–85.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    1. Ogden A,
    2. Rida PC,
    3. Aneja R
    . Let's huddle to prevent a muddle: centrosome declustering as an attractive anticancer strategy. Cell Death Differ 2012;19:1255–67.
    OpenUrlCrossRefPubMed
  43. 43.↵
    1. Danilov AV,
    2. Hu S,
    3. Orr B,
    4. Godek K,
    5. Mustachio LM,
    6. Sekula D,
    7. et al.
    Dinaciclib induces anaphase catastrophe in lung cancer cells via inhibition of cyclin-dependent kinases 1 and 2. Mol Cancer Ther 2016;15:2758–66.
    OpenUrlAbstract/FREE Full Text
  44. 44.↵
    1. Kawakami M,
    2. Mustachio LM,
    3. Rodriguez-Canales J,
    4. Mino B,
    5. Roszik J,
    6. Tong P,
    7. et al.
    Next-generation CDK2/9 Inhibitors and anaphase catastrophe in lung cancer. J Natl Cancer Inst 2017;109:djw297.
  45. 45.↵
    1. McClue SJ,
    2. Blake D,
    3. Clarke R,
    4. Cowan A,
    5. Cummings L,
    6. Fischer PM,
    7. et al.
    In vitro and in vivo antitumor properties of the cyclin dependent kinase inhibitor CYC202 (R-roscovitine). Int J Cancer 2002;102:463–8.
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. Whittaker SR,
    2. Walton MI,
    3. Garrett MD,
    4. Workman P
    . The cyclin-dependent kinase inhibitor CYC202 (R-roscovitine) inhibits retinoblastoma protein phosphorylation, causes loss of cyclin D1, and activates the mitogen-activated protein kinase pathway. Cancer Res 2004;64:262–72.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    1. Fleming IN,
    2. Hogben M,
    3. Frame S,
    4. McClue SJ,
    5. Green SR
    . Synergistic inhibition of ErbB signaling by combined treatment with seliciclib and ErbB-targeting agents. Clin Cancer Res 2008;14:4326–35.
    OpenUrlAbstract/FREE Full Text
  48. 48.↵
    1. Hu S,
    2. Danilov AV,
    3. Godek K,
    4. Orr B,
    5. Tafe LJ,
    6. Rodriguez-Canales J,
    7. et al.
    CDK2 inhibition causes anaphase catastrophe in lung cancer through the centrosomal protein CP110. Cancer Res 2015;75:2029–38.
    OpenUrlAbstract/FREE Full Text
  49. 49.↵
    1. Parry D,
    2. Guzi T,
    3. Shanahan F,
    4. Davis N,
    5. Prabhavalkar D,
    6. Wiswell D,
    7. et al.
    Dinaciclib (SCH 727965), a novel and potent cyclin-dependent kinase inhibitor. Mol Cancer Ther 2010;9:2344–53.
    OpenUrlAbstract/FREE Full Text
  50. 50.↵
    1. Hu S,
    2. Lu Y,
    3. Orr B,
    4. Godek K,
    5. Mustachio LM,
    6. Kawakami M,
    7. et al.
    Specific CP110 phosphorylation sites mediate anaphase catastrophe after cdk2 inhibition: evidence for cooperation with USP33 knockdown. Mol Cancer Ther 2015;14:2576–85.
    OpenUrlAbstract/FREE Full Text
  51. 51.↵
    1. Chen Z,
    2. Indjeian VB,
    3. McManus M,
    4. Wang L,
    5. Dynlacht BD
    . CP110, a cell cycle-dependent cdk substrate, regulates centrosome duplication in human cells. Dev Cell 2002;3:339–50.
    OpenUrlCrossRefPubMed
  52. 52.↵
    1. Kobayashi T,
    2. Tsang WY,
    3. Li J,
    4. Lane W,
    5. Dynlacht BD
    . Centriolar kinesin Kif24 interacts with CP110 to remodel microtubules and regulate ciliogenesis. Cell 2011;145:914–25.
    OpenUrlCrossRefPubMed
  53. 53.↵
    1. Kleylein-Sohn J,
    2. Westendorf J,
    3. Le Clech M,
    4. Habedanck R,
    5. Stierhof YD,
    6. Nigg EA
    . Plk4-induced centriole biogenesis in human cells. Dev Cell 2007;13:190–202.
    OpenUrlCrossRefPubMed
  54. 54.↵
    1. Schmidt TI,
    2. Kleylein-Sohn J,
    3. Westendorf J,
    4. Le Clech M,
    5. Lavoie SB,
    6. Stierhof YD,
    7. et al.
    Control of centriole length by CPAP and CP110. Curr Biol 2009;19:1005–11.
    OpenUrlCrossRefPubMed
  55. 55.↵
    1. Kohlmaier G,
    2. Loncarek J,
    3. Meng X,
    4. McEwen BF,
    5. Mogensen MM,
    6. Spektor A,
    7. et al.
    Overly long centrioles and defective cell division upon excess of the SAS-4-related protein CPAP. Curr Biol 2009;19:1012–8.
    OpenUrlCrossRefPubMed
  56. 56.↵
    1. Tang CJ,
    2. Fu RH,
    3. Wu KS,
    4. Hsu WB,
    5. Tang TK
    . CPAP is a cell-cycle regulated protein that controls centriole length. Nat Cell Biol 2009;11:825–31.
    OpenUrlCrossRefPubMed
  57. 57.↵
    1. Kobayashi T,
    2. Dynlacht BD
    . Regulating the transition from centriole to basal body. J Cell Biol 2011;193:435–44.
    OpenUrlAbstract/FREE Full Text
  58. 58.↵
    1. Tsang WY,
    2. Spektor A,
    3. Luciano DJ,
    4. Indjeian VB,
    5. Chen Z,
    6. Salisbury JL,
    7. et al.
    CP110 cooperates with two calcium-binding proteins to regulate cytokinesis and genome stability. Mol Biol Cell 2006;17:3423–34.
    OpenUrlAbstract/FREE Full Text
  59. 59.↵
    1. Tsang WY,
    2. Dynlacht BD
    . CP110 and its network of partners coordinately regulate cilia assembly. Cilia 2013;2:9.
    OpenUrlCrossRefPubMed
  60. 60.↵
    1. Spektor A,
    2. Tsang WY,
    3. Khoo D,
    4. Dynlacht BD
    . Cep97 and CP110 suppress a cilia assembly program. Cell 2007;130:678–90.
    OpenUrlCrossRefPubMed
  61. 61.↵
    1. Tsang WY,
    2. Bossard C,
    3. Khanna H,
    4. Peranen J,
    5. Swaroop A,
    6. Malhotra V,
    7. et al.
    CP110 suppresses primary cilia formation through its interaction with CEP290, a protein deficient in human ciliary disease. Dev Cell 2008;15:187–97.
    OpenUrlCrossRefPubMed
  62. 62.↵
    1. D'Angiolella V,
    2. Donato V,
    3. Vijayakumar S,
    4. Saraf A,
    5. Florens L,
    6. Washburn MP,
    7. et al.
    SCF (Cyclin F) controls centrosome homeostasis and mitotic fidelity through CP110 degradation. Nature 2010;466:138–42.
    OpenUrlCrossRefPubMed
  63. 63.↵
    1. Barenz F,
    2. Hoffmann I
    . Cell biology: DUBing CP110 controls centrosome numbers. Curr Biol 2013;23:R459–60.
    OpenUrlCrossRefPubMed
  64. 64.↵
    1. Li J,
    2. Kim S,
    3. Kobayashi T,
    4. Liang FX,
    5. Korzeniewski N,
    6. Duensing S,
    7. et al.
    Neurl4, a novel daughter centriole protein, prevents formation of ectopic microtubule organizing centres. EMBO Rep 2012;13:547–53.
    OpenUrlAbstract/FREE Full Text
  65. 65.↵
    1. Loukil A,
    2. Tormanen K,
    3. Sutterlin C
    . The daughter centriole controls ciliogenesis by regulating Neurl-4 localization at the centrosome. J Cell Biol 2017;216:1287–300.
    OpenUrlAbstract/FREE Full Text
  66. 66.↵
    1. Hossain D,
    2. Javadi Esfehani Y,
    3. Das A,
    4. Tsang WY
    . Cep78 controls centrosome homeostasis by inhibiting EDD-DYRK2-DDB1(Vpr)(BP). EMBO Rep 2017;18:632–44.
    OpenUrlAbstract/FREE Full Text
  67. 67.↵
    1. Li J,
    2. D'Angiolella V,
    3. Seeley ES,
    4. Kim S,
    5. Kobayashi T,
    6. Fu W,
    7. et al.
    USP33 regulates centrosome biogenesis via deubiquitination of the centriolar protein CP110. Nature 2013;495:255–9.
    OpenUrlCrossRefPubMed
  68. 68.↵
    1. Fung TK,
    2. Siu WY,
    3. Yam CH,
    4. Lau A,
    5. Poon RY
    . Cyclin F is degraded during G2 –M by mechanisms fundamentally different from other cyclins. J Biol Chem 2002;277:35140–9.
    OpenUrlAbstract/FREE Full Text
  69. 69.↵
    1. D'Angiolella V,
    2. Donato V,
    3. Forrester FM,
    4. Jeong YT,
    5. Pellacani C,
    6. Kudo Y,
    7. et al.
    Cyclin F-mediated degradation of ribonucleotide reductase M2 controls genome integrity and DNA repair. Cell 2012;149:1023–34.
    OpenUrlCrossRefPubMed
  70. 70.↵
    1. Dietlein F,
    2. Kalb B,
    3. Jokic M,
    4. Noll EM,
    5. Strong A,
    6. Tharun L,
    7. et al.
    A synergistic interaction between Chk1- and MK2 inhibitors in KRAS-mutant cancer. Cell 2015;162:146–59.
    OpenUrlCrossRefPubMed
  71. 71.↵
    1. Harrison MK,
    2. Adon AM,
    3. Saavedra HI
    . The G1 phase cdks regulate the centrosome cycle and mediate oncogene-dependent centrosome amplification. Cell Div 2011;6:2.
    OpenUrlCrossRefPubMed
  72. 72.↵
    1. Zeng X,
    2. Shaikh FY,
    3. Harrison MK,
    4. Adon AM,
    5. Trimboli AJ,
    6. Carroll KA,
    7. et al.
    The ras oncogene signals centrosome amplification in mammary epithelial cells through cyclin D1/Cdk4 and Nek2. Oncogene 2010;29:5103–12.
    OpenUrlCrossRefPubMed
  73. 73.↵
    1. Castagnola P,
    2. Giaretti W
    . Mutant KRAS, chromosomal instability and prognosis in colorectal cancer. Biochim Biophys Acta 2005;1756:115–25.
    OpenUrlPubMed
  74. 74.↵
    1. Huncharek M,
    2. Muscat J,
    3. Geschwind JF
    . K-ras oncogene mutation as a prognostic marker in non-small cell lung cancer: a combined analysis of 881 cases. Carcinogenesis 1999;20:1507–10.
    OpenUrlCrossRefPubMed
  75. 75.↵
    1. Marchetti A,
    2. Milella M,
    3. Felicioni L,
    4. Cappuzzo F,
    5. Irtelli L,
    6. Del Grammastro M,
    7. et al.
    Clinical implications of KRAS mutations in lung cancer patients treated with tyrosine kinase inhibitors: an important role for mutations in minor clones. Neoplasia 2009;11:1084–92.
    OpenUrlCrossRefPubMed
  76. 76.↵
    1. Roberts PJ,
    2. Stinchcombe TE
    . KRAS mutation: should we test for it, and does it matter? J Clin Oncol 2013;31:1112–21.
    OpenUrlAbstract/FREE Full Text
  77. 77.↵
    1. Bournet B,
    2. Buscail C,
    3. Muscari F,
    4. Cordelier P,
    5. Buscail L
    . Targeting KRAS for diagnosis, prognosis, and treatment of pancreatic cancer: hopes and realities. Eur J Cancer 2016;54:75–83.
    OpenUrlCrossRefPubMed
  78. 78.↵
    1. Aviel-Ronen S,
    2. Blackhall FH,
    3. Shepherd FA,
    4. Tsao MS
    . K-ras mutations in non-small-cell lung carcinoma: a review. Clin Lung Cancer 2006;8:30–8.
    OpenUrlCrossRefPubMed
  79. 79.↵
    1. Massarelli E,
    2. Varella-Garcia M,
    3. Tang X,
    4. Xavier AC,
    5. Ozburn NC,
    6. Liu DD,
    7. et al.
    KRAS mutation is an important predictor of resistance to therapy with epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. Clin Cancer Res 2007;13:2890–6.
    OpenUrlAbstract/FREE Full Text
  80. 80.↵
    1. Eberhard DA,
    2. Johnson BE,
    3. Amler LC,
    4. Goddard AD,
    5. Heldens SL,
    6. Herbst RS,
    7. et al.
    Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol 2005;23:5900–9.
    OpenUrlAbstract/FREE Full Text
  81. 81.↵
    1. Stephen AG,
    2. Esposito D,
    3. Bagni RK,
    4. McCormick F
    . Dragging ras back in the ring. Cancer Cell 2014;25:272–81.
    OpenUrlCrossRefPubMed
  82. 82.↵
    1. Quinlan MP,
    2. Settleman J
    . Explaining the preponderance of Kras mutations in human cancer: an isoform-specific function in stem cell expansion. Cell Cycle 2008;7:1332–5.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top
Molecular Cancer Therapeutics: 17 (4)
April 2018
Volume 17, Issue 4
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Editorial Board (PDF)

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Molecular Cancer Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Engaging Anaphase Catastrophe Mechanisms to Eradicate Aneuploid Cancers
(Your Name) has forwarded a page to you from Molecular Cancer Therapeutics
(Your Name) thought you would be interested in this article in Molecular Cancer Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Engaging Anaphase Catastrophe Mechanisms to Eradicate Aneuploid Cancers
Masanori Kawakami, Lisa Maria Mustachio, Xi Liu and Ethan Dmitrovsky
Mol Cancer Ther April 1 2018 (17) (4) 724-731; DOI: 10.1158/1535-7163.MCT-17-1108

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Engaging Anaphase Catastrophe Mechanisms to Eradicate Aneuploid Cancers
Masanori Kawakami, Lisa Maria Mustachio, Xi Liu and Ethan Dmitrovsky
Mol Cancer Ther April 1 2018 (17) (4) 724-731; DOI: 10.1158/1535-7163.MCT-17-1108
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Anaphase Catastrophe
    • Conclusions
    • Disclosure of Potential Conflicts of Interest
    • Acknowledgments
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • PIM Kinases Promote Resistance to Therapy
  • CAR-T Cell Therapy to Target TNBC
  • Targeting Mitochondrial One-Carbon Metabolism in Cancer
Show more Review
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About MCT

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Molecular Cancer Therapeutics
eISSN: 1538-8514
ISSN: 1535-7163

Advertisement