Abstract
Chromosomal instability (CIN), or the dynamic change in chromosome number and composition, has been observed in cancer for decades. Recently, this phenomenon has been implicated as facilitating the acquisition of cancer hallmarks and enabling the formation of aggressive disease. Hence, CIN has the potential to serve as a therapeutic target for a wide range of cancers. CIN in cancer often occurs as a result of disrupting key regulators of mitotic fidelity and faithful chromosome segregation. As a consequence of their essential roles in mitosis, dysfunctional centrosomes can induce and maintain CIN. Centrosome defects are common in breast cancer, a heterogeneous disease characterized by high CIN. These defects include amplification, structural defects, and loss of primary cilium nucleation. Recent studies have begun to illuminate the ability of centrosome aberrations to instigate genomic flux in breast cancer cells and the tumor evolution associated with aggressive disease and poor patient outcomes. Here, we review the role of CIN in breast cancer, the processes by which centrosome defects contribute to CIN in this disease, and the emerging therapeutic approaches that are being developed to capitalize upon such aberrations.
Keywords: breast cancer, centrosome, chromosomal instability, genome instability, microtubule, primary cilium
In 2011, Hanahan and Weinberg defined genome instability and mutation as an enabling characteristic that facilitates the acquisition of cancer hallmarks (1). Several forms of genome instability can occur, one of which is chromosomal instability (CIN) (2, 3). This refers to defects in chromosome segregation resulting in gains, losses, and alterations in portions of, or entire, chromosomes (a genomic state known as aneuploidy). These errors ultimately induce gene dosage changes for oncogenes and tumor suppressors (4). Importantly, CIN contributes to the advent of distinct phenotypes by promoting tumor evolution. CIN during early cancer development is associated with poor patient prognosis (5, 6). Furthermore, continuous evolution of genomically unstable tumors leads to more aggressive cancers. Thus, a more comprehensive understanding of the mechanisms leading to CIN should provide targets that can ultimately be leveraged for developing therapies for patients with genomically unstable tumors. Due to their essential role in mitosis, centrosomes provide 1 such target. Abnormalities affecting centrosome number, structure, and nucleation of the primary cilium have been identified as drivers of CIN. This review discusses the various centrosomal defects that contribute to CIN and how they impact breast cancer evolution, patient prognosis, and treatment.
Chromosomal Instability and Cancer
For decades, a high incidence of CIN-induced aneuploidy has been observed in most types of cancer. Technological advances such as cytogenetic and chromosomal array comparative genomic hybridization (CGH) have revealed that the vast majority of tumor cells contain 1 or more chromosomal aberrations (3, 7-10). For example, in breast cancer, it is not uncommon for a tumor to have lost over half of its alleles (11). The vast collection of genetic alterations caused by CIN leads to genetically heterogeneous cell populations within a tumor that continue to diversify with subsequent cell divisions (12). The genomic complexity of these populations contributes to significant intratumoral heterogeneity, resulting in tumor cells that are more adaptive, metastatic, and resistant to treatment (13-15); in other words, CIN imparts a fitness advantage (Fig. 1). Previously, CIN was thought to be tangentially related to the hallmarks of cancer (16). However, it has become evident that the extensive genomic variability induced by CIN produces discrete populations of cells that display an enhanced ability to sustain cancer hallmarks (1, 17). Hence, it has been recently proposed that the processes that foster CIN, including DNA damage/replication stress and mitotic stress should be included in the list of cancer hallmarks (17, 18), highlighting the importance of this enabling phenotype in tumor initiation and progression.
Figure 1.
Centrosome abnormalities induce CIN in cancer cells. Acquired centrosomal abnormalities impart a cellular fitness advantage through CIN, allowing cancer evolution and the generation of tumor cells that are more adaptable, metastatic, and treatment resistant. By inhibiting centrosome function using agents such as kinase inhibitors or centrosomal declustering agents, tumors already experiencing CIN can be further pushed to develop excessive CIN. This leads to cell senescence or death and ultimately, tumor stasis or regression.
Overview of CIN
Imprecision during chromosome condensation, spindle assembly checkpoint, centrosome duplication, DNA replication, and cytokinesis often result in CIN (19, 20), ultimately leading to both large- and small-scale genomic changes. Large genomic alterations, or whole CIN (wCIN), consists of a gain or loss of entire chromosomes causing variations in gene dosage of oncogenes and tumor suppressors (4). In contrast, structural CIN (sCIN) involves translocations, deletions, inversions, insertions, and fragmentations of subchromosomal regions (19). Both wCIN and sCIN events result in unstable aneuploid cells, a dynamic genomic state associated with evolving karyotypes across the tumor cell population. It is notable that a single cell division error can lead to progressive aneuploidy (21). Moreover, focal CIN can occur that results from the shattering and reformation of a single chromosome. This is known as chromothripsis and is a rapid form of karyotypic evolution (22) that leads to dysregulated and disrupted genes within the chromothriptic region.
CIN and Cancer Cell Fitness
As indicated above, the constant flux in chromosome composition in unstable aneuploid cells, along with associated changes in gene expression, causes extensive cell-to-cell variation in tumors (23, 24). This heterogeneity can enhance tumor aggressiveness (25-27). Paradoxically, CIN can also be detrimental to cancers. This paradigm can be explained by biological optimization or the Goldilocks Principle. In the framework of this theory, when CIN levels are “just right” cancer cells have various fitness and oncogenic advantages, including disabled cell cycle checkpoints and loss of tumor suppressor expression. However, cells with CIN levels at the extreme margins (excessive or low CIN) exhibit no evolutionary advantage. Cells with low CIN are unable to adapt. In contrast, excessive CIN can act as a vulnerability where the loss of essential genes/chromosomes leads to cell senescence or death and ultimately, tumor stasis or regression (5, 28, 29) (Fig. 1). Indeed, numerous studies have demonstrated that high CIN enhances sensitivity to cytotoxic therapies and radiotherapy (30-34). Hence, drugs that induce mitotic defects can push modestly genomically unstable cells from intermediate CIN to intolerable CIN (35). To maximally utilize this approach, it is first necessary to identify which cancer types and targets can be leveraged to shift cells to an intolerable state of CIN.
Breast Cancer and CIN
Breast cancer encompasses a number of diseases that are molecularly, morphologically, genomically, and clinically distinct. Clinically, breast cancers are classified by expression of estrogen (ER) or progesterone (PR) receptors and amplification of epidermal growth factor receptor 2 (HER2). Tumors lacking these receptors are referred to as triple negative breast cancer (TNBC). Breast cancers can also be categorized molecularly using gene expression profiles. Molecular subtypes include Luminal A (typically ER/PR+, HER2–), Luminal B (typically ER/PR/HER2+), HER2 (ER/PR–, HER2+), or basal-like (ER/PR/HER2–), with basal-like being further subclassified. The majority of TNBC are the basal subtype (36-38). Despite their discrete attributes, all breast cancer subtypes exhibit some degree of CIN. Indeed, in a study of 2201 tumor samples from 24 cancer types, breast cancers have one of the highest proportions of altered genomes as measured by copy number variation and chromosome segmental alterations (3). The heterogeneity in this disease has also been defined using cytogenetics, chromosomal, and microarray-based CGH, and massively parallel sequencing (14, 39-47). Strikingly, approximately 60% to 80% of breast tumors deviate from a normal diploid karyotype, with about 50% exhibiting genome doubling (48). Breast tumors with higher rates of CIN are associated with poorer patient outcomes, therapeutic resistance, metastasis, and recurrence (33, 36, 49).
CIN and TNBC Outcomes
Among breast cancer subtypes, basal (TNBC) breast cancers rank the highest in several categories of CIN (Fig. 2). This is associated with whole genome doubling or tetraploidization events (37, 38) and is prognostic of TNBC patient outcomes (50). A seminal study by Carter and colleagues developed a gene signature indicative of aneuploidy (51). By conducting a meta-analysis of 1944 tumors spanning 9 cancer types, the authors correlated the expression of individual genes with total functional aneuploidy levels (a proxy measure of aneuploidy that utilizes DNA copy number profiles and spectral karyotyping data). This analysis revealed that tumors with functional aneuploidy were enriched for the expression of 70 genes, now commonly known as the CIN70 gene signature. Birkbak et al. divided TNBC patients into quartiles based on gene expression levels of these CIN70 signature within their tumors and these groups were associated with differences in outcomes. Patients with intermediate (third quartile) CIN70 expression exhibited decreased recurrence-free survival when compared with patients with either extreme (fourth quartile) or low (first and second quartiles) CIN (50). These results exemplify the CIN and cancer fitness paradigm discussed previously. Underscoring the utility of targeting intermediate CIN to induce excessive genomic disruption, TNBC is often treated with taxanes and radiotherapy, both of which induce DNA and chromosomal damage (52-54). Radiotherapy induces excessive sCIN and causes chaotic mitoses that can lead to wCIN (30, 55), whereas taxanes stabilize microtubules and prevent proper chromosome segregation, also leading to wCIN (53, 54). By inducing extreme CIN, these therapies cause senescence, apoptosis, and, ultimately, tumor regression (28, 31, 56-59). Moreover, elevated CIN sensitizes breast cancers to taxanes. While such therapies are initially highly effective, resistance and recurrence is common, suggesting that the level of CIN is insufficient to uniformly induce cell death or irreversible senescence (60-62). Identifying druggable factors that further drive intolerable CIN in a tumor cell–selective manner should lead to highly effective therapies for breast cancer and other malignancies with intrinsic CIN.
Figure 2.
Basal breast tumors are genomically unstable. Breast cancer samples within The Cancer Genome Atlas (TCGA) classified as Luminal A, Luminal B, HER2+, or Basal were queried for (A) aneuploidy score, (B) fraction genome altered (FGA) using CBioportal. n = 945. (C) Expression level of the CIN70 gene signature in breast cancer patients within the TCGA. Note: 63/70 of the CIN70 genes were detected and reported in the dataset. *P < .05 when comparing each subtype with basal (148, 149).
The Centrosome and CIN
Centrosomes play key roles in regulating CIN and can be leveraged to treat breast cancers and other tumor types. Normally, these organelles are precisely regulated to maintain faithful chromosome segregation and cytokinesis. However, cancers often harbor centrosome aberrations that can lead to abnormal mitoses, CIN, and intratumoral heterogeneity (63) (Fig. 3). Notably, many of the CIN70 genes are regulators of centrosome function such as TPX2, NEK2, and TTK. Structural, numerical, and functional defects of centrosomes have been observed in breast cancers and are correlated with elevated CIN and poorer patient prognoses (64-67). The centrosome has also been implicated in driving the development of CIN in early breast lesions and inhibition of centrosome function can promote intolerable CIN. Importantly, the centrosome is composed of and regulated by many enzymatic components that are inherently druggable. For these reasons, the centrosome is a promising therapeutic target in cancers with high CIN.
Figure 3.
Centrosome function is necessary to maintain genome stability and tissue architecture. (A) Centrosome structure, number, and functionality are necessary for maintaining chromosome stability by nucleating mitotic spindles. The centrosome also defines cell polarity via its role in nucleating the primary cilium. (B) Increased centrosome volume and/or centrosome amplification (CA) are well documented occurrences in breast cancer that have been linked to CIN and intratumoral heterogeneity. (C) Loss of cell polarity due to primary cilium defects are less well defined, but is a common feature in cancer that can contribute to dysregulated cell signaling, chromosome missegregation, and disorganized tissue architecture, phenotypes commonly associated with aggressive cancer types.
Overview of the Centrosome
The Centrosome and Mitosis
The centrosome is an organelle that critically regulates cell polarity as well as chromosomal segregation during mitosis (68-70) (Fig. 3A). This multifunctional, microtubule-based structure comprises 2 centrioles surrounded by an electron-dense matrix known as pericentriolar material (PCM). The centrosome is the microtubule-organizing center; that is, it is the nucleating site for microtubule networks established during interphase and mitosis (65, 71, 72). The centrosome defines the number, polarity, and organization of microtubules. Microtubule nucleation is regulated by serine/threonine phosphorylation events that either promote or restrict the PCM from recruiting microtubule-organizing proteins (71, 73). Normally, centrosomes are tightly regulated by a number of proteins that control their number and spatial positioning throughout the cell cycle (74-77). During G1 of the cell cycle, normal cells have a single centrosome comprised of a pair of centrioles, a “daughter” and a “mother,” designated solely by the timing of their formation in the prior duplication cycle (76). During S phase, the mother and daughter centrioles both duplicate to generate 2 pairs of centrioles. At the onset of prophase, the centrosomes begin to separate and move to opposite poles of the dividing cell (78). At their respective poles, they nucleate bipolar mitotic spindles (79, 80) that are essential for chromosome alignment on the metaphase plate, sister chromatid separation, and cytokinesis (80). Upon forming the mitotic spindle, microtubules emanating from the centrosome elongate toward, and ultimately connect to, kinetochores within chromosomes. Once a centrosome-originating microtubule attaches to a kinetochore in the correct orientation, the microtubule is stabilized (81). Forces generated by centrosome-microtubule-kinetochore attachments pull the sister chromatids to opposite poles of the cell during anaphase. Following sister chromatid separation, the mitotic spindle will also determine the point of cell cleavage during cytokinesis, ensuring that both daughter cells inherit a complete and equal set of chromosomes within symmetrical cells. As the core of the mitotic spindle, centrosomes dictate correct chromosome segregation, cytokinesis, and cell cycle signaling, with centrosome defects being associated with prolonged mitosis, CIN, and failure to complete subsequent mitotic events (82, 83).
The Centrosome and the Primary Cilium
While the centrosome’s role in mitosis is well known, this organelle is also essential for formation of the primary cilium, a specialized, solitary, and nonmotile projection found on the surface of most animal cells that senses and transmits extracellular cues and establishes cell polarity (84, 85). Following mitosis, the mother centriole migrates toward the cell surface to form a basal body that acts as a scaffold for microtubule nucleation. In this case, the microtubules form the interior of the primary cilium (86, 87). Microtubule minus-ends anchor to the basal body near the apical membrane of the cell, adjacent to the lumen, while the plus-ends extend to the basal compartment, adjacent to the basement membrane (88). Through this directional anchoring, microtubules emanating from the centrosome dictate apical–basal polarity. Loss of cell polarity due to abnormalities in the primary cilium disrupt normal tissue architecture, a common hallmark of cancer.
Centrosome Dysfunction and CIN
Centrosome Amplification
Centrosome amplification (CA) refers to an abnormal increase in the number of centrosomes in the cell. This includes centrosomes that contain more than 4 centrioles and/or the acquisition of more than 1 pair of centrosomes when cells are not actively engaged in the cell cycle (64-66, 87). CA is highly prevalent in solid tumors and cell lines, with ≥75% of breast cancers displaying CA (90). Compared with the NCI-60 panel of cell lines originating from 9 different tissue types, breast cancer cell lines across all subtypes have significantly higher CA (90). CA arises through several processes, including hormone signaling perturbation (91, 92), excessive recruitment of PCM (93-95), abnormal kinase regulation, mutation-induced dysfunction of centrosome proteins, centrosome gene dysregulation, cytokinesis failure, or cell–cell fusion (96, 97). Downstream, CA induces CIN by initiating multipolar spindles during prophase, leading to chromosome missegregation (98) (Fig. 3B). Abnormal chromosomal attachments arising from multipolar spindles cause an array of mitotic defects including lagging or broken chromosomes that generate micronuclei, chromosome rearrangements, and chromothripsis (98, 99); all of which contribute to CIN.
Similar centrosome defects are observed in patient samples. In a cohort of 362 breast cancer patients, CA was assessed by immunohistochemistry for pericentrin, a component of PCM, and polyglutamylated tubulin (γ-tubulin), a marker of centrioles. The number of centrosomes per cell in tumor tissue was nearly double that of normal breast epithelium. The population of patients with the highest number of centrosomes were those in the HER2+ and TNBC cohorts (97). These data affirm results from 2 smaller studies, both using similar immunohistochemistry methods to quantify centrosome number (64, 66). In a separate study, basal breast cancers were also shown to have significantly elevated CA when compared with receptor-positive subtypes (Luminal A/B, HER2+) in cell lines and patient tumor samples (90, 100). Overall, patients with higher centrosome number exhibit higher stage and grade tumors (97), with the most aggressive breast cancer subtypes (TNBC, HER2+) having more extensive CA. Patients with tumors that harbor CA are also more likely to have decreased progression-free survival, or shorter time to recurrence or metastasis, compared with patients with less centrosome aberrations (100). On a molecular level, overexpression of 5 centrosome-related genes, CETN2 (centrin-2), TUBG1 (γ-tubulin), PCNT2 (pericentrin), PLK4 (polo-like kinase 4), and CCNE1 (cyclin E1), are associated with decreased overall survival regardless of breast cancer subtype (100).
Several features of aggressive breast cancers are associated with CA, including loss of estrogen-dependence and p53 function, elevated epidermal growth factor receptor expression, tumor dedifferentiation, and increased metastatic potential (64, 97, 101). In an independent study using METABRIC and TCGA datasets, a signature of 20 centrosome structural genes, named the CA20, was developed to assess the association of CA with clinical outcome in breast cancer patients. This study found that tumors with a high CA20 signature were associated with decreased overall patient survival compared with tumors that had a lower CA20 score (102). The CA20 score was also highly correlated with a smaller subset of the CIN70 gene signature, known as CIN25 (90). Together, these data indicate that CA may drive malignant phenotypes of breast cancer and affirm the relationship between CA and CIN.
The aggressive nature of tumors harboring CA may be due to its ability to drive aneuploidy, CIN, and intratumoral heterogeneity. In the previously mentioned cohort of 362 patients, fluorescence in situ hybridization analyses revealed that CA positively correlates with tumor ploidy and an increase in nonmodal number of chromosomes, both indicators of CIN (97). Unsurprisingly, patients with both high CA and high CIN had worse overall survival (97). In cell line models, higher rates of CA are also associated with CIN, including abnormal mitotic events characterized by multipolar spindles, lagging chromosomes, and cytokinetic defects (66, 97). Breast tumors comprising cells with severe CA are also more likely to have aberrant spindle morphology, a common precursor of CIN, and higher mitotic indices, which correlate with worse prognosis (66).
Direct evidence that CA impacts various stages of tumor development, including tumor initiation and progression, has been obtained using rat and mouse models of hormonally controlled mammary tumorigenesis (91, 92, 103-107). Matthews and colleagues found that glucocorticoid receptor (GR) localization at the mitotic spindle, especially in metaphase and anaphase cells, is necessary for mitotic progression. Specifically, loss of GR at the mitotic spindle led to CA, chromosome missegregation, aneuploidy, and increased incidence of tumor formation (91). Importantly, the disruption of the DNA-binding domain of GR alone was insufficient to induce CA and mitotic defects. Hence, these data suggest that GR may have a nontranscriptional role in regulating mitotic progression. In a second model of rat tumorigenesis, long-term estrogen stimulation elevated the expression of the centrosome protein Aurora Kinase A (AURKA), CA, CIN, and tumorigenesis (92). These results contrast with another study utilizing a rat model of pregnancy-induced protection from breast cancer. In this case, short-term estrogen treatment coupled with progesterone, which mimics pregnancy, prevented AURKA overexpression, CA, and CIN. This approach revealed that parity, or hormone signaling resembling parity, caused resistance to carcinogen-induced tumorigenesis in rats (104). These studies indicate the important role of hormones and their timing in the regulation of genomic stability and how modulation of these control mechanisms can impact breast tumorigenesis. Further supporting the tumorigenic role of CA, a study involving overexpression of AURKA in the murine mammary gland demonstrated that CA led to premature sister chromatid segregation, chromosome tetraploidization, CIN, and the formation of mammary tumors (103). A second centrosome associated protein, ninein-like protein (Nlp) has been found to be overexpressed in 80% of breast cancer patients (107). Transgenic mice that globally overexpress Nlp also form spontaneous mammary tumors, with CA occurring in tumors as well as mouse embryonic fibroblasts (89, 107). These findings revealed that dysregulated centrosome-associated proteins lead to CA and that this may induce breast tumorigenesis. These in vivo models highlight the impact that CA has on breast cancer initiation and demonstrate that CA can occur prior to detectable lesions.
This same ability of CA to induce CIN also drives tumor progression through the acquisition of intratumoral heterogeneity. As a genomically unstable tumor develops, clonal outgrowth contributes to branched evolution. In the MDA-MB-231 model of claudin-low TNBC, cells with CA demonstrated increased migratory capacity compared with cells devoid of CA (100), a phenotype associated with invasive, metastatic disease. To assess whether CA drives aggressive phenotypes, this same study overexpressed another centrosomal kinase, PLK4, in the nontransformed mammary epithelial cell line MCF10A. PLK4 also induced CA with subsequently elevated invasion and migration (100). These findings indicate that CA is an important driver of aggressive phenotypes in cancer. This could be due to elevated CIN and subsequent evolution and intratumoral heterogeneity. Alternatively, the centrosome also controls cell polarization, intracellular positioning of the small GTPase, Rac1, and cell migration (108-110). Thus, CA may also directly stimulate migration and invasion of cancer cells. This possibility is supported by the ability of PLK4 and NEK2, another centrosome kinase, to induce CA and migration when overexpressed. This occurs in a short time frame before selection of promigratory genomic changes (111, 112), thus elevated migration could be a direct effect of altering centrosome function rather than its indirect impact on CIN. The presence of CA is associated with breast tumor initiation and tumor progression, thus CA has the potential to be both a diagnostic and prognostic feature of breast cancer.
Centrosome Structural Abnormalities
Maintaining the structure of the centrosome and its centrioles is as vital as controlling centrosome number. Structural abnormalities can be classified as defects in centriole shape or in the amount of surrounding PCM. Mechanisms underlying such abnormalities are less well studied than CA; however, altered expression of genes that control centrosome structure may be a key factor (113). These abnormalities can present in a few ways, but the most common phenomenon in breast cancer is overelongated centrioles (90, 100). Centrioles are the microtubule-based cylinders that constitute the centrosome and are typically ≈450 nm in length. In the NCI-60 cell line panel, breast cancer cell centrioles have a median length of ≈546 nm, making them the cancer type with the longest centrioles across the panel (90). Overly elongated centrioles are prone to fragmentation, resulting in the formation of ectopic procentrioles. When centrosome elongation occurs asymmetrically, 1 centriole nucleates more microtubules and becomes unstable. Subsequently, centriole fragmentation causes CA and increases the number of sites for microtubule nucleation (90). This ultimately leads to CIN (Fig. 3B). The second mechanism through which CA can occur is as a result of de novo ectopic procentriole formation (90). Rather than fragmenting, overly long centrioles can generate more than 1 daughter centriole along its length, compared with normal cells where each centriole typically only nucleates the formation of a single daughter (90). As a result, ectopic procentriole formation increases the number of centrioles (>4) and centrosomes per cell. Lastly, overly long centrioles can cause the centrosome to be overactive, leading to excessive microtubule-nucleation in the absence of overt fragmentation. This is not considered CA because a normal centriole number is observed, but it does cause the same defect in microtubule organization. The clinical relevance of increased microtubule-nucleating capacity observed in cell lines was affirmed using breast adenocarcinoma tissue samples (114), where microtubules were found to originate from multiple regions within tumor cells (114). Centrosomes with increased microtubules can generate unbalanced forces on chromosomes, multipolar spindles, and chromosome missegregation, all of which can induce CIN and intratumoral heterogeneity. Confirming a direct link between centriole overelongation and CIN, overexpression of CPAP, a promoter of centriole elongation, was found to induce centriole overelongation in a luminal A breast cancer cell line (MDA-MB-435) and this resulted in chromosome missegregation and CIN (90). These results indicate that centriole overelongation contributes to intratumoral heterogeneity in breast cancer either due to secondary CA or as a result of increased microtubule-nucleating capacity.
A second form of centrosome structural abnormality involves elevated centrosome volume, or centrosome hypertrophy. Comparison of centrosome volume using γ-tubulin immunostaining in breast cancer samples revealed a significant increase in centrosome size compared with normal adjacent breast epithelium. Strikingly, centrosome size was also found to be subtype specific, with TNBC samples having larger centrosomes by volume than non-TNBC samples (100). Similar results were obtained using centrin immunostaining. In this case, centrosomes in breast tumor cells were approximately 7 to 10 times larger than those in normal breast (65). Centrosome hypertrophy may contribute to CIN and intratumoral heterogeneity by disrupting normal mitoses. In all reported cases of structural abnormalities, features of CIN were present, indicating varied irregular centrosome pathologies can independently contribute to mitotic dysfunction and chromosome segregation errors.
Primary Cilium Aberrations
While the centrosome’s role in mitosis has been extensively studied, this organelle also acts as the basal body, which anchors microtubules to form the primary cilium and establishes apical–basal cell polarity. The directionality of the microtubules emanating from the basal body establishes the polarity of the cell (Fig. 3A) and loss of cellular polarity due to centrosome aberrations can disrupt normal tissue architecture (Fig. 3C). This is particularly important in secretory tissues such as the mammary gland that comprises alveoli that produce milk and ducts that transport it during lactation. The ducts and alveoli are lined by 2 layers of cells: the luminal epithelial layer which is closest to the lumen of the duct and the myoepithelial layer attached to the basement membrane (115). Proper cellular polarity is necessary for the formation of complex branching units within the mammary gland (115). The cells themselves are also highly organized and oriented such that the nucleus resides at the basal portion of the cell while the centrosome is positioned at the luminal portion of the mammary duct, providing directionality for milk secretion. Loss of this organizational structure, including apical–basal polarity, is often observed during breast cancer progression (115). Because the centrosome dictates this polarity, it is not surprising that centrosome aberrations can induce irregular breast tissue architecture in the setting of breast cancer.
In normal breast tissue and tissue adjacent to breast tumors, epithelial cells form highly organized ring-like structures with centrin/centrosomes being localized near the luminal membrane (65). In contrast, breast cancer cells containing centrosome abnormalities display an architecture that is chaotic and disarrayed, losing the luminal structure that is characteristic of the terminal ductal lobular units (97). Disrupted tissue organization is often associated with a relocalization of centrosomes to regions near the nucleus rather than at the apical pole of the cell (116). This abnormal positioning prevents proper apical localization of the primary cilium and impedes normal polarization. In other cases, centrin can be diffusely localized throughout the cytoplasm of cancer cells, resulting in the inability to coalesce a functional centrosome. This also results in the absence of a primary cilium, disruption of polarity, and architectural disarray (65). Notably, loss of the primary cilium occurs in most breast cancers (117). In cell lines, the loss of the primary cilium was more prominent in transformed and aggressive derivatives than in parental nontransformed cells (117). Moreover, in vitro studies of mammary epithelial cells grown in 3D cultures demonstrated that loss of tissue architecture was associated with increased chromosome missegregation, perhaps due to disrupted cell polarity. Mature mammary spheroids that had an organized tissue architecture similar to normal breast tissue lacked CIN phenotypes. However, immature mammary spheroids that lacked tissue architecture had elevated rates of chromosomal missegregation (118). While incompletely understood at a mechanistic level, these data indicate that the loss of cellular polarity cues imparted by the primary cilium is associated with inaccurate chromosome segregation. Further studies will be necessary to elucidate how primary cilium defects result in disorganized tissue architecture and how this contributes to CIN. This is especially important because the morphological complexity in invasive ductal breast carcinoma is linked to both disease-specific and overall survival and is more strongly associated with outcomes than other standard prognosticators such as nodal status or grade (119).
Therapeutic Potential of Targeting Centrosomes
As indicated above, CIN and its associated intratumoral heterogeneity contribute to significant disease burden, cancer cell fitness, and therapeutic recurrence in breast cancer (120, 121). However, CIN can also be a therapeutic vulnerability that can be leveraged to induce cell death and tumor regression or suppression (5, 28, 29, 59) (Fig. 1). CIN-elevating strategies typically exacerbate existing instability by inducing extreme levels of chromosome missegregation or DNA damage and loss of vital genomic information (35). CIN-targeting therapies typically disrupt mitosis and chromosome dynamics. For example, taxanes, including paclitaxel and docetaxel, are one of the most established classes of chemotherapy. These drugs stabilize microtubules, resulting in mitotic arrest due to activation of the spindle assembly checkpoint (54, 122). A number of other drugs function in a similar capacity to inhibit microtubules and induce chromosome missegregation. Furthermore, drugs targeting mitotic kinases, chromatin modifications, and microtubule-associated motor proteins are also inducers of excessive CIN (35). An additional druggable target related to CIN in breast cancer is the centrosome itself. Given the high incidence of centrosomal abnormalities in this disease and their relationship to chromosome segregation, targeting centrosomes is a rational avenue for cancer therapy. This can involve several different approaches including therapeutics that are directed at CA, primary cilium defects, phosphorylation dynamics, upstream activating kinases, nucleating capacity of the centrosome, or overexpression of proteins involved in centrosome structure or function (123). Several therapies that directly or indirectly impact these mechanisms have been developed and ongoing efforts focus on assessing their efficacy as single agents and in combination with other therapies.
Centrosomal Kinase Inhibitors
Given the diverse roles of various kinases in cancer initiation and progression, suppressing kinase activity has been a favorable approach for cancer drug development (124). Several centrosome kinase inhibitors are already in clinical trials, including those that target polo-like kinases (PLK), cell cycle related kinases, and Aurora kinases (125-127). Given the frequency of centrosome aberrations in breast cancers, centrosome kinase inhibitors should be highly effective in this disease. As a recent example, inhibitors of PLK4 have been assessed in breast cancers with TRIM37 amplification (128). TRIM37 limits the accumulation of PCM proteins, thus preventing the formation of noncentrosomal PCM foci. These foci are essential for chromosome segregation and maintenance of euploidy in cells that are viable but lack centrosomes. Thus, the overexpression of TRIM37 provides a vulnerability of tumor cells to agents that impact centrosomes. Notably, the inhibition of PLK4 causes centrosome depletion and is synthetically lethal with TRIM37 overexpression in breast cancer cells (128). PLK4 inhibition using CFI-400945 CFI-400945 is also effective in breast cancer cells with centrosome abnormalities due to its ability to induce catastrophic defects in centrosome function (129).
Aurora kinases are also important targets in breast cancer that control centrosomes or microtubules. AURKA acts at centrosomes to permit spindle bipolarity by regulating microtubule assembly and centrosome organization. In contrast, Aurora kinase B (AURKB) is a member of the chromosome passenger complex that localizes to chromosomes and assists in kinetochore–microtubule attachment during metaphase (130). Both AURKA and AURKB are overexpressed or amplified in breast cancer and are associated with poor outcomes, making them promising therapeutic targets (131). As such, a number of Aurora kinase inhibitors are currently being evaluated in breast cancer clinical trials (126, 132-136). In addition to the goal of treating primary disease, centrosome kinases may also be leveraged to combat therapeutic resistance. Taxane resistance is common in breast cancer patients and remains a significant therapeutic obstacle. We recently reported that paclitaxel-resistant cells have CA and express elevated levels of NIMA-related kinase 2 (NEK2), a kinase that controls centrosome separation and bipolar spindle formation in mitotic cells (137). These findings indicated that NEK2 may be a potential target to combat paclitaxel resistance. Indeed, inhibiting NEK2 with small molecule inhibitors or siRNA resensitized breast cancer cell lines to taxanes. Moreover, the combination of paclitaxel with NEK2 inhibitors suppressed tumor growth more so than either drug alone in multiple in vivo models of breast cancer (138). These data support the use of centrosome kinase inhibitors as either first line, targeted, or combination therapy in breast cancer.
Centrosome Declustering Agents
Tool compounds have also been developed that target compensatory mechanisms utilized by cells to sustain viability within the context of CA. As stated above, CA is a common feature of breast tumors that can lead to multipolar spindles and CIN. To overcome CA and prevent chaotic and fatal mitoses, these cells undergo an adaptive process called centrosome clustering that involves the coalescence of multiple centrosomes into 2 functional spindle poles comprised of megacentrosomes (139). This process is beneficial in tumor cells for 2 reasons: (1) it prevents excessive CIN and (2) it still enables low-grade chromosome missegregation and sustains tumor evolution (139, 140). Centrosome clustering is common in centrosome-amplified breast cancer tumors, with more than 75% of cells having this observable phenotype in tumors and cell lines (141). Multiple drugs and targets have been identified to disrupt centrosome clustering. Kinesin Family Member C1 (KIFC1, also known as HSET) is a nonessential kinesin motor protein that is overexpressed in breast cancer cells. Along with its binding partner CEP215, KIFC1 is essential for centrosome clustering and survival in breast cancer cell lines (142). Genetic depletion of either KIFC1 or CEP215 significantly increases multipolar spindles, centrosome scattering, and death in cell lines with CA (143, 144). Multiple small molecule inhibitors of KIFC1 have been developed, including CW069 and AZ282, and both phenocopy the effects of KIFC1 genetic depletion. Indirect inhibition of KIFC1 using a small molecule inhibitor of poly(ADP-ribose) polymerase (PARP) (PJ34) also blocks breast cancer cell growth due to a loss of centrosome clustering (142, 145). Lastly, KIFC1 depletion sensitizes breast cancer cells to ionizing radiation (IR) (146). IR induces CA and protective centrosome clustering, therefore genomic depletion of KIFC1 provides a selective vulnerability, making these cells more sensitive to IR (146). Griseofulvin (139) and noscapinoids (147) are additional examples of microtubule-inhibiting drugs that also prevent centrosome clustering. Discerning whether drugs targeting centrosome clustering are effective in patients awaits the development of clinically relevant molecules.
Conclusions and Future Directions
Intratumoral heterogeneity and CIN contribute to evolving genomic landscapes that result in aggressive breast cancers. A limited understanding of the many mechanisms that lead to CIN have precluded efforts to develop strategies that combat tumor evolution. In this regard, the centrosome offers a promising prognostic tool and therapeutic target in breast cancer. Centrosome aberrations are a driving force in breast cancer that cause chromosome missegregation, CIN, and tumorigenesis. While abnormal centrosomes participate in tumor evolution and are associated with worse outcomes in breast cancer, they also represent an attractive vulnerability. Many therapies are at different stages of development for targeting aberrant centrosomes in breast cancer. However, a major looming limitation of anticentrosome therapies is the advent of drug resistance. Generating additional CIN could expedite stochastic evolution as a result of chromosome missegregation. Cells able to evade death from centrosome targeting will likely have elevated CIN compared with the remaining tumor bulk and have the ability to resist further treatment. Thus, it will be essential for centrosome-targeting drugs to induce intolerable levels of CIN that ensure cell death. Elucidating mechanisms of resistance that could develop in response to these therapies is necessary before wide-scale implementation in breast cancer patients. It is likely that evoking durable responses to centrosome-targeting drugs will require their combination with additional CIN-inducing therapies to prevent adaptive and resistant disease. Discovering effective biomarkers predictive of susceptibility to centrosome-targeted therapies would also facilitate their clinical development and deployment. These may include direct assessments of centrosome number and size as well as using molecular profiling to assess the CA20 score (102). Overall, it remains imperative to understand how to therapeutically induce excessive CIN while simultaneously preventing further evolution. Dysfunctional centrosomes are a partial and promising answer to this challenging issue in breast cancer.
Acknowledgments
Funding: This work was supported by National Institute of Health grants R01CA206505 (RAK), R01CA257502 (RAK), T32GM007250 (KMP), and a Department of Defense Breast Cancer Research Program grant W81XWH-18–1–0455 (LJA).
Glossary
Abbreviations
- CA
centrosome amplification
- CIN
chromosomal instability
- ER
estrogen receptor
- GR
glucocorticoid receptor
- HER2
epidermal growth factor receptor 2
- PCM
pericentriolar material
- PR
progesterone receptor
- TNBC
triple negative breast cancer
Additional Information
Disclosure Statement: The authors have nothing to disclose.
Data Availability
Some or all data generated or analyzed during this study are included in this published article or in the data repositories listed in References 148 and 149.
References
- 1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-674. [DOI] [PubMed] [Google Scholar]
- 2. Vargas-Rondon N, Villegas VE, Rondon-Lagos M. The role of chromosomal instability in cancer and therapeutic responses. Cancers. 2017;10(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Pikor L, Thu K, Vucic E, Lam W. The detection and implication of genome instability in cancer. Cancer Metastasis Rev. 2013;32(3-4):341-352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Roschke AV, Rozenblum E. Multi-layered cancer chromosomal instability phenotype. Front Oncol. 2013;3:302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Giam M, Rancati G. Aneuploidy and chromosomal instability in cancer: a jackpot to chaos. Cell Div. 2015;10:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Burkard ME, Weaver BA. Tuning chromosomal instability to optimize tumor fitness. Cancer Discov. 2017;7(2):134-136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Mertens F, Johansson B, Höglund M, Mitelman F. Chromosomal imbalance maps of malignant solid tumors: a cytogenetic survey of 3185 neoplasms. Cancer Res. 1997;57(13):2765-2780. [PubMed] [Google Scholar]
- 8. Hyman E, Kauraniemi P, Hautaniemi S, et al. Impact of DNA amplification on gene expression patterns in breast cancer. Cancer Res. 2002;62(21):6240-6245. [PubMed] [Google Scholar]
- 9. Wolf M, Mousses S, Hautaniemi S, et al. High-resolution analysis of gene copy number alterations in human prostate cancer using CGH on cDNA microarrays: impact of copy number on gene expression. Neoplasia. 2004;6(3):240-247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Beroukhim R, Mermel CH, Porter D, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463(7283):899-905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in human cancers. Nature. 1998;396(6712):643-649. [DOI] [PubMed] [Google Scholar]
- 12. Bakhoum SF, Landau DA. Chromosomal instability as a driver of tumor heterogeneity and evolution. Cold Spring Harb Perspect Med. 2017;7(6):a029611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Chen G, Mulla WA, Kucharavy A, et al. Targeting the adaptability of heterogeneous aneuploids. Cell. 2015;160(4):771-784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Navin N, Kendall J, Troge J, et al. Tumour evolution inferred by single-cell sequencing. Nature. 2011;472(7341):90-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Yates LR, Gerstung M, Knappskog S, et al. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat Med. 2015;21(7):751-759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57-70. [DOI] [PubMed] [Google Scholar]
- 17. Negrini S, Gorgoulis VG, Halazonetis TD. Genomic instability–an evolving hallmark of cancer. Nat Rev Mol Cell Biol. 2010;11(3):220-228. [DOI] [PubMed] [Google Scholar]
- 18. Luo J, Solimini NL, Elledge SJ. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell. 2009;136(5):823-837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Jelluma N, Kops GJ. Collateral genome instability by DNA damage in mitosis. Cancer Discov. 2014;4(11):1256-1258. [DOI] [PubMed] [Google Scholar]
- 20. Heng HH, Bremer SW, Stevens JB, et al. Chromosomal instability (CIN): what it is and why it is crucial to cancer evolution. Cancer Metastasis Rev. 2013;32(3-4):325-340. [DOI] [PubMed] [Google Scholar]
- 21. Umbreit NT, Zhang CZ, Lynch LD, et al. Mechanisms generating cancer genome complexity from a single cell division error. Science. 2020;368(6488):eaba0712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Leibowitz ML, Zhang CZ, Pellman D. Chromothripsis: a new mechanism for rapid karyotype evolution. Annu Rev Genet. 2015;49(1):183-211. [DOI] [PubMed] [Google Scholar]
- 23. Heng HH, Bremer SW, Stevens JB, et al. Chromosomal instability (CIN): what it is and why it is crucial to cancer evolution. Cancer Metastasis Rev. 2013;32(3-4):325-340. [DOI] [PubMed] [Google Scholar]
- 24. McGranahan N, Burrell RA, Endesfelder D, Novelli MR, Swanton C. Cancer chromosomal instability: therapeutic and diagnostic challenges. EMBO Rep. 2012;13(6):528-538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Ramon YCS, Sese M, Capdevila C, et al. Clinical implications of intratumor heterogeneity: challenges and opportunities. J Mol Med. 2020;98(2):161-177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Dexter DL, Leith JT. Tumor heterogeneity and drug resistance. J Clin Oncol. 1986;4(2):244-257. [DOI] [PubMed] [Google Scholar]
- 27. Durrett R, Foo J, Leder K, Mayberry J, Michor F. Intratumor heterogeneity in evolutionary models of tumor progression. Genetics. 2011;188(2):461-477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Funk LC, Zasadil LM, Weaver BA. Living in CIN: mitotic infidelity and its consequences for tumor promotion and suppression. Dev Cell. 2016;39(6):638-652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Silk AD, Zasadil LM, Holland AJ, Vitre B, Cleveland DW, Weaver BA. Chromosome missegregation rate predicts whether aneuploidy will promote or suppress tumors. Proc Natl Acad Sci U S A. 2013;110(44):E4134-E4141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Bakhoum SF, Kabeche L, Wood MD, et al. Numerical chromosomal instability mediates susceptibility to radiation treatment. Nat Commun. 2015;6:5990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Swanton C, Nicke B, Schuett M, et al. Chromosomal instability determines taxane response. Proc Natl Acad Sci U S A. 2009;106(21):8671-8676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Jamal-Hanjani M, A’Hern R, Birkbak NJ, et al. Extreme chromosomal instability forecasts improved outcome in ER-negative breast cancer: a prospective validation cohort study from the TACT trial. Ann Oncol. 2015;26(7):1340-1346. [DOI] [PubMed] [Google Scholar]
- 33. Roylance R, Endesfelder D, Gorman P, et al. Relationship of extreme chromosomal instability with long-term survival in a retrospective analysis of primary breast cancer. Cancer Epidemiol Biomarkers Prev. 2011;20(10):2183-2194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Zaki BI, Suriawinata AA, Eastman AR, Garner KM, Bakhoum SF. Chromosomal instability portends superior response of rectal adenocarcinoma to chemoradiation therapy. Cancer. 2014;120(11):1733-1742. [DOI] [PubMed] [Google Scholar]
- 35. Thompson LL, Jeusset LMP, Lepage CC, et al. Evolving therapeutic strategies to exploit chromosome instability in cancer. Cancers. 2017;9(11):151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Donepudi MS, Kondapalli K, Amos SJ, Venkanteshan P. Breast cancer statistics and markers. J Cancer Res Ther. 2014;10(3):506-511. [DOI] [PubMed] [Google Scholar]
- 37. Smid M, Hoes M, Sieuwerts AM, et al. Patterns and incidence of chromosomal instability and their prognostic relevance in breast cancer subtypes. Breast Cancer Res Treat. 2011;128(1):23-30. [DOI] [PubMed] [Google Scholar]
- 38. López S, Lim EL, Horswell S, et al. ; TRACERx Consortium . Interplay between whole-genome doubling and the accumulation of deleterious alterations in cancer evolution. Nat Genet. 2020;52(3):283-293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Cottu PH, Asselah J, Lae M, et al. Intratumoral heterogeneity of HER2/neu expression and its consequences for the management of advanced breast cancer. Ann Oncol. 2008;19(3):596-597. [DOI] [PubMed] [Google Scholar]
- 40. Navin N, Krasnitz A, Rodgers L, et al. Inferring tumor progression from genomic heterogeneity. Genome Res. 2010;20(1):68-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Shah SP, Morin RD, Khattra J, et al. Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature. 2009;461(7265):809-813. [DOI] [PubMed] [Google Scholar]
- 42. Shah SP, Roth A, Goya R, et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature. 2012;486(7403):395-399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Torres L, Ribeiro FR, Pandis N, Andersen JA, Heim S, Teixeira MR. Intratumor genomic heterogeneity in breast cancer with clonal divergence between primary carcinomas and lymph node metastases. Breast Cancer Res Treat. 2007;102(2):143-155. [DOI] [PubMed] [Google Scholar]
- 44. Park SY, Gönen M, Kim HJ, Michor F, Polyak K. Cellular and genetic diversity in the progression of in situ human breast carcinomas to an invasive phenotype. J Clin Invest. 2010;120(2):636-644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Pandis N, Teixeira MR, Adeyinka A, et al. Cytogenetic comparison of primary tumors and lymph node metastases in breast cancer patients. Genes Chromosomes Cancer. 1998;22(2):122-129. [PubMed] [Google Scholar]
- 46. Geyer FC, Weigelt B, Natrajan R, et al. Molecular analysis reveals a genetic basis for the phenotypic diversity of metaplastic breast carcinomas. J Pathol. 2010;220(5):562-573. [DOI] [PubMed] [Google Scholar]
- 47. Nik-Zainal S, Alexandrov LB, Wedge DC, et al. Mutational processes molding the genomes of 21 breast cancers. Cell. 2012;149(5):979-993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Carter SL, Cibulskis K, Helman E, et al. Absolute quantification of somatic DNA alterations in human cancer. Nat Biotechnol. 2012;30(5):413-421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Kronenwett U, Huwendiek S, Ostring C, et al. Improved grading of breast adenocarcinomas based on genomic instability. Cancer Res. 2004;64(3):904-909. [DOI] [PubMed] [Google Scholar]
- 50. Birkbak NJ, Eklund AC, Li Q, et al. Paradoxical relationship between chromosomal instability and survival outcome in cancer. Cancer Res. 2011;71(10):3447-3452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Carter SL, Eklund AC, Kohane IS, Harris LN, Szallasi Z. A signature of chromosomal instability inferred from gene expression profiles predicts clinical outcome in multiple human cancers. Nat Genet. 2006;38(9):1043-1048. [DOI] [PubMed] [Google Scholar]
- 52. de Ruijter TC, Veeck J, de Hoon JP, van Engeland M, Tjan-Heijnen VC. Characteristics of triple-negative breast cancer. J Cancer Res Clin Oncol. 2011;137(2):183-192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Mustacchi G, De Laurentiis M. The role of taxanes in triple-negative breast cancer: literature review. Drug Des Devel Ther. 2015;9:4303-4318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Weaver BA. How Taxol/paclitaxel kills cancer cells. Mol Biol Cell. 2014;25(18):2677-2681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Huang RX, Zhou PK. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther. 2020;5(1):60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Lee HS, Lee NC, Kouprina N, et al. Effects of anticancer drugs on chromosome instability and new clinical implications for tumor-suppressing therapies. Cancer Res. 2016;76(4):902-911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Torres K, Horwitz SB. Mechanisms of Taxol-induced cell death are concentration dependent. Cancer Res. 1998;58(16):3620-3626. [PubMed] [Google Scholar]
- 58. McClelland SE, Burrell RA, Swanton C. Chromosomal instability: a composite phenotype that influences sensitivity to chemotherapy. Cell Cycle. 2009;8(20):3262-3266. [DOI] [PubMed] [Google Scholar]
- 59. Scribano CM, Wan J, Esbona K, et al. Chromosomal instability sensitizes patient breast tumors to multipolar divisions induced by paclitaxel. Sci Transl Med. 2021;13(610):eabd4811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Jiang T, Shi W, Wali VB, et al. Predictors of chemosensitivity in triple negative breast cancer: an integrated genomic analysis. Plos Med. 2016;13(12):e1002193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Jiang L, Siu MK, Wong OG, et al. iASPP and chemoresistance in ovarian cancers: effects on paclitaxel-mediated mitotic catastrophe. Clin Cancer Res. 2011;17(21):6924-6933. [DOI] [PubMed] [Google Scholar]
- 62. Nitta M, Kobayashi O, Honda S, et al. Spindle checkpoint function is required for mitotic catastrophe induced by DNA-damaging agents. Oncogene. 2004;23(39):6548-6558. [DOI] [PubMed] [Google Scholar]
- 63. Jusino S, Fernandez-Padin FM, Saavedra HI. Centrosome aberrations and chromosome instability contribute to tumorigenesis and intra-tumor heterogeneity. J Cancer Metastasis Treat. 2018;4:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Lingle WL, Barrett SL, Negron VC, et al. Centrosome amplification drives chromosomal instability in breast tumor development. Proc Natl Acad Sci U S A. 2002;99(4):1978-1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Lingle WL, Lutz WH, Ingle JN, Maihle NJ, Salisbury JL. Centrosome hypertrophy in human breast tumors: implications for genomic stability and cell polarity. Proc Natl Acad Sci U S A. 1998;95(6):2950-2955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. D’Assoro AB, Barrett SL, Folk C, et al. Amplified centrosomes in breast cancer: a potential indicator of tumor aggressiveness. Breast Cancer Res Treat. 2002;75(1):25-34. [DOI] [PubMed] [Google Scholar]
- 67. Godinho SA, Pellman D. Causes and consequences of centrosome abnormalities in cancer. Philos Trans R Soc Lond B Biol Sci. 2014;369(1650):20130467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Guo HQ, Gao M, Ma J, et al. Analysis of the cellular centrosome in fine-needle aspirations of the breast. Breast Cancer Res. 2007;9(4):R48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Doxsey S. Duplicating dangerously: linking centrosome duplication and aneuploidy. Mol Cell. 2002;10(3):439-440. [DOI] [PubMed] [Google Scholar]
- 70. Sen S. Aneuploidy and cancer. Curr Opin Oncol. 2000;12(1):82-88. [DOI] [PubMed] [Google Scholar]
- 71. Bettencourt-Dias M, Glover DM. Centrosome biogenesis and function: centrosomics brings new understanding. Nat Rev Mol Cell Biol. 2007;8(6):451-463. [DOI] [PubMed] [Google Scholar]
- 72. Wu J, Akhmanova A. Microtubule-organizing centers. Annu Rev Cell Dev Biol. 2017;33:51-75. [DOI] [PubMed] [Google Scholar]
- 73. Wang G, Jiang Q, Zhang C. The role of mitotic kinases in coupling the centrosome cycle with the assembly of the mitotic spindle. J Cell Sci. 2014;127(Pt 19):4111-4122. [DOI] [PubMed] [Google Scholar]
- 74. Nigg EA, Holland AJ. Once and only once: mechanisms of centriole duplication and their deregulation in disease. Nat Rev Mol Cell Biol. 2018;19(5):297-312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Nigg EA, Stearns T. The centrosome cycle: centriole biogenesis, duplication and inherent asymmetries. Nat Cell Biol. 2011;13(10):1154-1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Hatch E, Stearns T. The life cycle of centrioles. Cold Spring Harb Symp Quant Biol. 2010;75:425-431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Holland AJ, Lan W, Cleveland DW. Centriole duplication: a lesson in self-control. Cell Cycle. 2010;9(14):2731-2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Barr AR, Gergely F. Aurora-A: the maker and breaker of spindle poles. J Cell Sci. 2007;120(Pt 17):2987-2996. [DOI] [PubMed] [Google Scholar]
- 79. Meraldi P. Centrosomes in spindle organization and chromosome segregation: a mechanistic view. Chromosome Res. 2016;24(1):19-34. [DOI] [PubMed] [Google Scholar]
- 80. Petry S. Mechanisms of mitotic spindle assembly. Annu Rev Biochem. 2016;85(1):659-683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Kirschner M, Mitchison T. Beyond self-assembly: from microtubules to morphogenesis. Cell. 1986;45(3):329-342. [DOI] [PubMed] [Google Scholar]
- 82. Wong YL, Anzola JV, Davis RL, et al. Cell biology. Reversible centriole depletion with an inhibitor of Polo-like kinase 4. Science. 2015;348(6239):1155-1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Sir JH, Pütz M, Daly O, et al. Loss of centrioles causes chromosomal instability in vertebrate somatic cells. J Cell Biol. 2013;203(5):747-756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Joukov V, De Nicolo A. The centrosome and the primary cilium: the yin and yang of a hybrid organelle. Cells. 2019;8(7):701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Badano JL, Teslovich TM, Katsanis N. The centrosome in human genetic disease. Nat Rev Genet. 2005;6(3):194-205. [DOI] [PubMed] [Google Scholar]
- 86. Sorokin S. Centrioles and the formation of rudimentary cilia by fibroblasts and smooth muscle cells. J Cell Biol. 1962;15(2):363-377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Pan J, Snell W. The primary cilium: keeper of the key to cell division. Cell. 2007;129(7):1255-1257. [DOI] [PubMed] [Google Scholar]
- 88. Müsch A. Microtubule organization and function in epithelial cells. Traffic. 2004;5(1):1-9. [DOI] [PubMed] [Google Scholar]
- 89. D’Assoro AB, Lingle WL, Salisbury JL. Centrosome amplification and the development of cancer. Oncogene. 2002;21(40):6146-6153. [DOI] [PubMed] [Google Scholar]
- 90. Marteil G, Guerrero A, Vieira AF, et al. Over-elongation of centrioles in cancer promotes centriole amplification and chromosome missegregation. Nat Commun. 2018;9(1):1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Matthews LC, Berry AA, Morgan DJ, et al. Glucocorticoid receptor regulates accurate chromosome segregation and is associated with malignancy. Proc Natl Acad Sci U S A. 2015;112(17):5479-5484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Li JJ, Weroha SJ, Lingle WL, Papa D, Salisbury JL, Li SA. Estrogen mediates Aurora-A overexpression, centrosome amplification, chromosomal instability, and breast cancer in female ACI rats. Proc Natl Acad Sci U S A. 2004;101(52):18123-18128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Antonczak AK, Mullee LI, Wang Y, et al. Opposing effects of pericentrin and microcephalin on the pericentriolar material regulate CHK1 activation in the DNA damage response. Oncogene. 2016;35(15):2003-2010. [DOI] [PubMed] [Google Scholar]
- 94. Woodruff JB, Wueseke O, Hyman AA. Pericentriolar material structure and dynamics. Philos Trans R Soc Lond B Biol Sci. 2014;369(1650). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Conduit PT, Brunk K, Dobbelaere J, Dix CI, Lucas EP, Raff JW. Centrioles regulate centrosome size by controlling the rate of Cnn incorporation into the PCM. Curr Biol. 2010;20(24):2178-2186. [DOI] [PubMed] [Google Scholar]
- 96. Zhang Y, Tian J, Qu C, et al. A look into the link between centrosome amplification and breast cancer. Biomed Pharmacother. 2020;132:110924. [DOI] [PubMed] [Google Scholar]
- 97. Denu RA, Zasadil LM, Kanugh C, Laffin J, Weaver BA, Burkard ME. Centrosome amplification induces high grade features and is prognostic of worse outcomes in breast cancer. BMC Cancer. 2016;16:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Ganem NJ, Godinho SA, Pellman D. A mechanism linking extra centrosomes to chromosomal instability. Nature. 2009;460(7252):278-282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Ogden A, Rida PCG, Aneja R. Centrosome amplification: a suspect in breast cancer and racial disparities. Endocr Relat Cancer. 2017;24(9):T47-T64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Pannu V, Mittal K, Cantuaria G, et al. Rampant centrosome amplification underlies more aggressive disease course of triple negative breast cancers. Oncotarget. 2015;6(12):10487-10497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Schneeweiss A, Sinn HP, Ehemann V, et al. Centrosomal aberrations in primary invasive breast cancer are associated with nodal status and hormone receptor expression. Int J Cancer. 2003;107(3):346-352. [DOI] [PubMed] [Google Scholar]
- 102. Ogden A, Rida PC, Aneja R. Prognostic value of CA20, a score based on centrosome amplification-associated genes, in breast tumors. Sci Rep. 2017;7(1):262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Wang X, Zhou YX, Qiao W, et al. Overexpression of aurora kinase A in mouse mammary epithelium induces genetic instability preceding mammary tumor formation. Oncogene. 2006;25(54):7148-7158. [DOI] [PubMed] [Google Scholar]
- 104. Goepfert TM, Adigun YE, Zhong L, Gay J, Medina D, Brinkley WR. Centrosome amplification and overexpression of aurora A are early events in rat mammary carcinogenesis. Cancer Res. 2002;62(14):4115-4122. [PubMed] [Google Scholar]
- 105. Goepfert TM, Moreno-Smith M, Edwards DG, Pathak S, Medina D, Brinkley WR. Loss of chromosomal integrity drives rat mammary tumorigenesis. Int J Cancer. 2007;120(5):985-994. [DOI] [PubMed] [Google Scholar]
- 106. Bergmann S, Royer-Pokora B, Fietze E, et al. YB-1 provokes breast cancer through the induction of chromosomal instability that emerges from mitotic failure and centrosome amplification. Cancer Res. 2005;65(10):4078-4087. [DOI] [PubMed] [Google Scholar]
- 107. Shao S, Liu R, Wang Y, et al. Centrosomal Nlp is an oncogenic protein that is gene-amplified in human tumors and causes spontaneous tumorigenesis in transgenic mice. J Clin Invest. 2010;120(2):498-507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Ueda M, Gräf R, MacWilliams HK, Schliwa M, Euteneuer U. Centrosome positioning and directionality of cell movements. Proc Natl Acad Sci U S A. 1997;94(18):9674-9678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Cheng HW, Hsiao CT, Chen YQ, et al. Centrosome guides spatial activation of Rac to control cell polarization and directed cell migration. Life Sci Alliance. 2019;2(1):e201800135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Zhang J, Wang YL. Centrosome defines the rear of cells during mesenchymal migration. Mol Biol Cell. 2017;28(23):3240-3251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Kazazian K, Go C, Wu H, et al. Plk4 Promotes cancer invasion and metastasis through Arp2/3 complex regulation of the actin cytoskeleton. Cancer Res. 2017;77(2):434-447. [DOI] [PubMed] [Google Scholar]
- 112. Rivera-Rivera Y, Marina M, Jusino S, et al. The Nek2 centrosome-mitotic kinase contributes to the mesenchymal state, cell invasion, and migration of triple-negative breast cancer cells. Sci Rep. 2021;11(1):9016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Wu Q, Li B, Liu L, Sun S, Sun S. Centrosome dysfunction: a link between senescence and tumor immunity. Signal Transduct Target Ther. 2020;5(1):107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Salisbury JL, Lingle WL, White RA, Cordes LE, Barrett S. Microtubule nucleating capacity of centrosomes in tissue sections. J Histochem Cytochem. 1999;47(10):1265-1274. [DOI] [PubMed] [Google Scholar]
- 115. Rejon C, Al-Masri M, McCaffrey L. Cell polarity proteins in breast cancer progression. J Cell Biochem. 2016;117(10):2215-2223. [DOI] [PubMed] [Google Scholar]
- 116. Lingle WL, Salisbury JL. Altered centrosome structure is associated with abnormal mitoses in human breast tumors. Am J Pathol. 1999;155(6):1941-1951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Yuan K, Frolova N, Xie Y, et al. Primary cilia are decreased in breast cancer: analysis of a collection of human breast cancer cell lines and tissues. J Histochem Cytochem. 2010;58(10):857-870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Knouse KA, Lopez KE, Bachofner M, Amon A. Chromosome segregation fidelity in epithelia requires tissue architecture. Cell. 2018;175(1):200-211.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Tambasco M, Eliasziw M, Magliocco AM. Morphologic complexity of epithelial architecture for predicting invasive breast cancer survival. J Transl Med. 2010;8:140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Kwei KA, Kung Y, Salari K, Holcomb IN, Pollack JR. Genomic instability in breast cancer: pathogenesis and clinical implications. Mol Oncol. 2010;4(3):255-266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Bakhoum SF, Compton DA. Chromosomal instability and cancer: a complex relationship with therapeutic potential. J Clin Invest. 2012;122(4):1138-1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Abu Samaan TM, Samec M, Liskova A, et al. Paclitaxel’s mechanistic and clinical effects on breast cancer. Biomolecules. 2019;9(12):789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Schatten H (ed.). The Impact of Centrosome Abnormalities on Breast Cancer Development and Progression with a Focus on Targeting Centrosomes for Breast Cancer Therapy, in Cell and Molecular Biology of Breast Cancer. Humana Press; 2013:261-287. [Google Scholar]
- 124. Bhullar KS, Lagarón NO, McGowan EM, et al. Kinase-targeted cancer therapies: progress, challenges and future directions. Mol Cancer. 2018;17(1):48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Schatten H, Sun QY. Functions and dysfunctions of the mammalian centrosome in health, disorders, disease, and aging. Histochem Cell Biol. 2018;150(4):303-325. [DOI] [PubMed] [Google Scholar]
- 126. Cheung CH, Coumar MS, Chang JY, Hsieh HP. Aurora kinase inhibitor patents and agents in clinical testing: an update (2009-10). Expert Opin Ther Pat. 2011;21(6):857-884. [DOI] [PubMed] [Google Scholar]
- 127. Schöffski P. Polo-like kinase (PLK) inhibitors in preclinical and early clinical development in oncology. Oncologist. 2009;14(6):559-570. [DOI] [PubMed] [Google Scholar]
- 128. Yeow ZY, Lambrus BG, Marlow R, et al. Targeting TRIM37-driven centrosome dysfunction in 17q23-amplified breast cancer. Nature. 2020;585(7825):447-452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Mason JM, Lin DC, Wei X, et al. Functional characterization of CFI-400945, a Polo-like kinase 4 inhibitor, as a potential anticancer agent. Cancer Cell. 2014;26(2):163-176. [DOI] [PubMed] [Google Scholar]
- 130. Gully CP, Zhang F, Chen J, et al. Antineoplastic effects of an Aurora B kinase inhibitor in breast cancer. Mol Cancer. 2010;9:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Tang A, Gao K, Chu L, Zhang R, Yang J, Zheng J. Aurora kinases: novel therapy targets in cancers. Oncotarget. 2017;8(14):23937-23954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. McLaughlin J, Markovtsov V, Li H, et al. Preclinical characterization of Aurora kinase inhibitor R763/AS703569 identified through an image-based phenotypic screen. J Cancer Res Clin Oncol. 2010;136(1):99-113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Carpinelli P, Ceruti R, Giorgini ML, et al. PHA-739358, a potent inhibitor of Aurora kinases with a selective target inhibition profile relevant to cancer. Mol Cancer Ther. 2007;6(12 Pt 1):3158-3168. [DOI] [PubMed] [Google Scholar]
- 134. Oslob JD, Romanowski MJ, Allen DA, et al. Discovery of a potent and selective aurora kinase inhibitor. Bioorg Med Chem Lett. 2008;18(17):4880-4884. [DOI] [PubMed] [Google Scholar]
- 135. Shang X, Burlingame SM, Okcu MF, et al. Aurora A is a negative prognostic factor and a new therapeutic target in human neuroblastoma. Mol Cancer Ther. 2009;8(8):2461-2469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Yang J, Ikezoe T, Nishioka C, et al. AZD1152, a novel and selective aurora B kinase inhibitor, induces growth arrest, apoptosis, and sensitization for tubulin depolymerizing agent or topoisomerase II inhibitor in human acute leukemia cells in vitro and in vivo. Blood. 2007;110(6):2034-2040. [DOI] [PubMed] [Google Scholar]
- 137. Fry AM. The Nek2 protein kinase: a novel regulator of centrosome structure. Oncogene. 2002;21(40):6184-6194. [DOI] [PubMed] [Google Scholar]
- 138. Roberts MS, Sahni JMB, Schrock M, et al. LIN9 and NEK2 are core regulators of mitotic fidelity that can be therapeutically targeted to overcome taxane resistance. Cancer Res. 2020;80(8):1693-1706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Rebacz B, Larsen TO, Clausen MH, et al. Identification of griseofulvin as an inhibitor of centrosomal clustering in a phenotype-based screen. Cancer Res. 2007;67(13):6342-6350. [DOI] [PubMed] [Google Scholar]
- 140. Ogden A, Cheng A, Rida PC, et al. Quantitative multi-parametric evaluation of centrosome declustering drugs: centrosome amplification, mitotic phenotype, cell cycle and death. Cell Death Dis. 2014;5:e1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Pannu V, Rida PC, Celik B, et al. Centrosome-declustering drugs mediate a two-pronged attack on interphase and mitosis in supercentrosomal cancer cells. Cell Death Dis. 2014;5:e1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Li Y, Lu W, Chen D, et al. KIFC1 is a novel potential therapeutic target for breast cancer. Cancer Biol Ther. 2015;16(9):1316-1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Kwon M, Godinho SA, Chandhok NS, et al. Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev. 2008;22(16):2189-2203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Chavali PL, Chandrasekaran G, Barr AR, et al. A CEP215-HSET complex links centrosomes with spindle poles and drives centrosome clustering in cancer. Nat Commun. 2016;7:11005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Castiel A, Visochek L, Mittelman L, Dantzer F, Izraeli S, Cohen-Armon M. A phenanthrene derived PARP inhibitor is an extra-centrosomes de-clustering agent exclusively eradicating human cancer cells. BMC Cancer. 2011;11:412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Choe MH, Kim J, Ahn J, Hwang SG, Oh JS, Kim JS. Centrosome Clustering is a tumor-selective target for the improvement of radiotherapy in breast cancer cells. Anticancer Res. 2018;38(6):3393-3400. [DOI] [PubMed] [Google Scholar]
- 147. Karna P, Rida PC, Pannu V, et al. A novel microtubule-modulating noscapinoid triggers apoptosis by inducing spindle multipolarity via centrosome amplification and declustering. Cell Death Differ. 2011;18(4):632-644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401-404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Some or all data generated or analyzed during this study are included in this published article or in the data repositories listed in References 148 and 149.



