Open Access

KIFC1: A Reliable Prognostic Biomarker in Rb-positive Triple-negative Breast Cancer Patients Treated With Doxorubicin in Combination With Abemaciclib

FLEISHER BRETT 1
WERKMAN CAROLIN 1
JACOBS BREHANNA 1
VARKEY JUSTIN 1
TAHA KAREEM 1
  &  
AIT-OUDHIA SIHEM 2

1Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, College of Pharmacy, Orlando, FL, U.S.A.

2Quantitative Pharmacology and Pharmacometrics (QP2), Merck & Co., Inc, Kenilworth, NJ, U.S.A.

Cancer Diagnosis & Prognosis Sep-Oct; 2(5): 525-532 DOI: 10.21873/cdp.10137
Received 17 June 2022 | Revised 29 March 2024 | Accepted 19 July 2022
Corresponding author
Sihem Ait-Oudhia, MS, PharmD, Ph.D., Quantitative Pharmacology and Pharmacometrics (QP2), Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ 07033, U.S.A. Tel: +1 7162509828 sb.manuscript.submission@gmail.com

Abstract

Background/Aim: Triple-negative breast cancer (TNBC) prevalence and risk of relapse are greatest in African American (AA) patients. Doxorubicin (DOX) and abemaciclib (ABE) synergism in Rb-positive TNBC cells (MDA-MB-231), and antagonism in Rb-negative TNBC cells (MDA-MB-468) have been previously shown. Here, we assessed Kinesin-like protein 1 (KIFC1) as an ethnic-specific prognostic biomarker of the DOX+ABE combination for the Rb-status in TNBC. Materials and Methods: Literature search for TNBC prognostic biomarkers in the AA population was conducted. MDA-MB-231 and MDA-MB-468 cells were exposed over 72 h to four treatment arms: 1) control (medium without drug), 2) DOX at 50% inhibitory concentration in MDA-MB-231 (0.565 μM) and MDA-MB-468 (0.121 μM), 3) ABE alone (2 μM), and 4) DOX+ABE combination at their corresponding concentrations in each cell-line. KIFC1 protein expression and temporal changes were quantified in MDA-MB-231 cells using western blot. Results: KIFC1, Kaiso, and Annexin A2 are literature-identified AA-specific TNBC prognostic biomarkers. KIFC1 was found to be uncorrelated to other proposed biomarkers, suggesting it may predict risk independently of other TNBC biomarkers. In both cell lines, DOX alone did not significantly change KIFC1 expression relative to control. Conversely, ABE reduced KIFC1 expression in MDA-MB-231 but not in MDA-MB-468 cells. The combination DOX+ABE resulted in a greatest reduction in KIFC1 in MDA-MB-231 cells with a more rapid time-to-full inhibition of KIFC1 compared to ABE alone. Conclusion: Change in KIFC1 expression is primarily driven by ABE in Rb-positive TNBC cells. DOX increases ABE speed to achieve a full inhibition of KIFC1 in Rb-positive, yet, without influencing its expression in Rb-negative TNBC cells.
Keywords: prognosis, combination therapy, CDK4/6 inhibitor, anthracycline

Triple-negative breast cancer (TNBC) is a breast cancer (BC) subtype characterized by the lack of the three hallmark receptors: estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). TNBC occurs in about 10-20% of diagnosed BC patient population and is more aggressive compared to other BC subtypes (1). Diagnoses of TNBC occur at a younger age in African Americans (AA) (average age <50 years) and with more aggressive phenotypes as compared to Caucasian Americans (CA) (1,2). The American Society of Clinical Oncology (ASCO) recommends TNBC patients be treated with a cytotoxic chemotherapeutic regimen based on anthracyclines and taxanes (3). Despite an initial response to chemotherapy, the risk of TNBC is high (4,5), especially in AA patients (6), prompting the necessity for enhanced therapeutic alternatives in AA patients with TNBC.

We recently reported a clinical translation of in vitro data using mathematical modeling of the interaction of an anthracycline, doxorubicin (DOX), and the CDK4/6 inhibitor, abemaciclib (ABE) (7). The mechanism of action of DOX and ABE involves separate intracellular molecular circuitries. DOX induces cytotoxicity by forming a covalent bond between topoisomerase II and DNA in addition to arresting TNBC cells in the G2/M phase of the cell cycle (8,9). ABE inhibits CDK4/6, thereby, halting the G1 to S phase transition and preventing CDK4/6 signaling-dependent cell replication (10). The nature of the drug-drug interaction (synergism, additivity, antagonism) in in vitro combinations was dependent on the Rb status of the TNBC (7). The combination produced synergism in the MDA-MB-231 cells, an Rb-expressing and highly metastatic TNBC cell line. Conversely, the combination produced antagonism in the MDA-MB-468 cells, an Rb-negative TNBC cell line (9,11-13).

Historically, clinical monitoring of disease-associated biomarkers improves patient survival by serving as reliable targets for efficacious treatment (predictive biomarkers) or prognostic biomarkers (14). While Rb may be a predictive biomarker supporting the mechanistic rationale of utilizing CDK4/6 inhibitors in TNBC subtypes, a prognostic biomarker can help preemptively estimate TNBC progression in AA patients following treatment with novel therapeutic options.

In the present work, we first collected from literature reports an up-to-date list of candidate prognostic TNBC biomarkers that were proven specific to AA patients. Then, we analyzed the reported correlations between the biomarker of interest and AA ethnicity as well as TNBC aggressiveness. Our results show that while each biomarker of interest from the TNBC specimens in each study correlates positively with the AA ethnicity within the TNBC samples, the biomarker expression correlates negatively with the survival in donors. Hence, of all the examined biomarkers, we report the time-course change in the negative prognostic biomarker, Kinesin-like protein 1 (KIFC1).

Materials and Methods

Literature search of African American-specific prognostic biomarkers in TNBC. PubMed Central (PMC) and Google Scholar search engines were used to conduct an intense literature search to screen studies examining the role of biomarkers specific for AA TNBC. First, studies were identified using keywords such as “triple-negative breast cancer” and “biomarker” and “African American.” Second, only studies with measured biomarker expression using immunohistochemistry techniques in TNBC tissue microarrays from AA and CA donors were selected for the analysis. Lastly, a biomarker was considered as an AA-specific negative prognostic biomarker if the results in the published study (A) report a comparison of the frequency of expression of the biomarker of interest in TNBC samples collected in both AA and CA TNBC donors, (B) report that the biomarker of interest correlates positively with an AA ethnicity, and (C) report that the biomarker of interest correlates negatively with the patients’ overall survival.

Cell culture and reagents. The two TNBC cells lines MDA-MB-231 and MDA-MB-468 were obtained from American Type Culture Collection (Manassas, VA, USA). Both were cultured in Dulbecco’s Modified Eagle Medium (DMEM) with 10% sterile filtered fetal bovine serum (Sigma-Aldrich, Saint Louis, MO, USA) and 1% penicillin/streptomycin. The cells were incubated at 37˚C in an incubator supplied with 5% CO2. DOX and ABE were purchased from Selleck Chemicals (Houston, TX, USA). The stock solutions of DOX (30 mM) and ABE (10 mM) were prepared in Millipore water, aliquoted at 20 μl and then stored at −80˚C.

Sample collection. The cells were seeded in 6-well plates at density of cells 3×105 cells/well. After allowing for an overnight cell adherence, drugs or fresh media alone were added to the corresponding wells and left for incubation at varying time intervals spanning from 0 to 72 h. At the end of each incubation time, all cells (i.e., floating in medium and adherent) were collected via trypsinization, pooled, centrifuged at 180 × g for four min, and washed with PBS. Then, cells were re-suspended in 75 μl ice-cold Milliplex® MAP Lysis Buffer supplemented with protease/phosphatase inhibitor. After that the tube containing the cells was shaken for 10 min at 4˚C, incubated on ice for 10 min, and centrifuged for 1 min at 10,000 × g. The supernatant was then collected, aliquoted, and stored at −80˚C. The total cell protein was quantified using the BCA Protein Assay Kit (Thermo Fisher Scientific, Waltham, MA, USA).

Measurement of KIFC1. The western blot technique was used to quantify the change in the expression of the KIFC1 protein. In brief, rabbit primary antibodies against KIFC1 (ab72452; Abcam, Cambridge, UK) and the GAPDH housekeeping protein (2118S Cell Signaling, Danvers, MA, USA) were used. The 12% MP TGX Stain-Free Gel was loaded with 30 μg total proteins per lane, then transferred onto trans-Blot Turbo Mini PVDF (Bio-Rad, Hercules, CA, USA). A 5% non-fat dry milk in 1x phosphate-buffered saline with tween (PBST; 9809S; Cell Signaling Technologies, Danvers, MA, USA) was used for 60 min at room temperature to block (i.e., saturate) the membrane. The latter was then incubated overnight with gentle agitation at 4˚C with either rabbit KIFC1 at 1:2,000 dilution or GAPDH at 1:1,000 dilution. Following the incubation, the membrane was thoroughly washed with PBST, and then incubated for 1 h at room temperature with a secondary antibody. The bands were developed using electrochemiluminescence (ECL; Bio-Rad, Hercules, CA, USA). Using ImageLab, a densitometric analysis was carried out to measure the levels of total KIFC1, which were normalized to the total GAPDH.

The drug concentrations producing 50% of maximum inhibitory effects (IC50) for DOX in the TNBC cells lines MDA-MB-231 and MDA-MB-468 were previously reported (7). In order to reproduce the same synergistic interaction of DOX with ABE (7), MDA-MB-231 and MDA-MB-468 cells were exposed to either the medium alone (control), the IC50 concentration of DOX that is specific to each cell line, which is 0.565 μM for MDA-MB-231 and 0.121 μM for MDA-MB-468, the ABE alone at 2 μM, or the combination of DOX and ABE over different exposure times spanning the interval of 0-72 h. The time-course analysis of KIFC1 normalized to GAPDH (KIFC1/GAPDH) over 72 h was conducted in MDA-MB-231 cells only. The KIFC1/GAPDH in treated cells was further normalized to KIFC1/GAPDH of control at the respective times. The GraphPad Prism Version 5 for windows (GraphPad Software Inc., San Diego, CA, USA) was used for plotting the results.

Statistical analysis. The statistical differences between the mean values of the measurements obtained in the different treatment arms versus controls were calculated using the GraphPad Prism Version 5 software. The one-way analysis of variance test was used, followed by the Tukey test. A p-value of <0.05 was used as a threshold to consider the difference between groups as statistically significant.

Results

AA-specific biomarkers. The identified specific TNBC prognostic biomarkers specific to AA patients included KIFC1 (15) (also known as HSET), Kaiso (16), and Annexin A2 (ANXA2) (17).

ABE inhibits KIFC1 in MDA-MB-231 but not MDA-MB-468 cells. The expression of KIFC1, normalized to GAPDH, was measured in in MDA-MB-231 and MDA-MB-468 cells following treatment with single agents, DOX and ABE, as well as their combination. In the control treatment arm, the baseline level of KIFC1/GAPDH was identical in MDA-MB-231 and MDA-MB-468 (Figure 1). The levels of KIFC1 decreased by 0.6-fold relative to control in MDA-MB-231 cells treated with DOX alone, despite the non-statistical significance of this decrease. The KIFC1 decreased by 0.8-fold compared to control in MDA-MB-231 cells treated with ABE alone, and decreased by 0.9-fold compared to control when the same cell line was treated with the combination of DOX+ABE. In both groups (ABE single agent and combination DOX+ABE), the difference from control was statistically significant. In MDA-MB-468 cells exposed to either DOX alone or the combination DOX+ABE, KIFC1 increased by 0.3-fold compared to control. However, this difference was not statistically significant. Similarly, MDA-MB-468 cells exposed to ABE alone did not impact KIFC1 expression levels.

The combination of DOX with ABE inhibits KIFC1 faster than single agent ABE. The analysis of the time trajectory of KIFC1/GAPDH fold change from baseline in MDA-MB-231 showed that ABE decreased KIFC1/GAPDH to 0.5-fold that of control at 12 h after administration and resulted in about complete inhibition of KIFC1/GAPDH at 48 h post-exposure (Figure 2). The single agent DOX induced nearly 1.5-fold increase in KIFC1/GAPDH compared to control. The combination DOX+ABE exhibited the same pattern in KIFC1/GAPDH reduction as ABE single agent during the first 6 h, followed by a decay to non-quantifiable levels at 24 h post-administration.

Discussion

The aggressive progression of TNBC is reported to be more frequent in AA women than in any other ethnic subgroup, despite considerations of patients’ age, stage of the disease or even the socioeconomic status of the patients (6,18). One of the major factors involved in this inequal risk between populations is the inherent heterogeneity in tumor biology between different ethnicities, hence warranting the search for diagnostic tools that are specific to each ethnicity. Ethnic-specific prognostic biomarkers are biological entities associated with patients’ outcome such as patient-free or overall survival allowing oncologists to anticipate the therapeutic risk in cancer patients (19). Following an intense literature search, three published prognostic biomarkers met our selection criteria of negative patient’s clinical outcome in AA population Kaiso (16), ANXA2 (17), and KIFC1 (15).

Kaiso and ANXA2 appear to confer patient mortality via the Epidermal Growth Factor Receptor (EGFR) pathway. EGFR activity, a well-studied negative prognostic biomarker in TNBC patients (20-23), induces Kaiso sublocalization to the nucleus (24), where Kaiso, a bi-modal transcription factor (25), silences E-cadherin. The net result is the loss of adherent junctions between cancer cells (16,24,26,27). EGFR also mediates Src-induced phosphorylation of ANXA2, a calcium-dependent phospholipid protein, which increases cell-surface localization and activity of ANXA2 (28,29). Phosphorylated ANXA2 then mediates the plasmin-induced modification of extracellular matrix (ECM) and the release of angiogenic growth factor (30-32). All three biomarkers induce epithelial-mesenchymal transition (EMT) of the tumor, a process in which modifying cell-cell adhesion or the surrounding ECM attributes to cancer cells a highly metastatic potential (21).

Unlike the other biomarkers, KIFC1 may not be correlated to other commonly examined TNBC markers, including EGFR (14,15). The KIFC1 protein acts by directing the spindle assembly of chromosomes with supernumerary centrosomes such as the ones present in aggressive TNBC cells (33-37). It cross-links the microtubule minus-end of spindles (33), which ultimately will converge at the acentrosomal microtubule-organizing centers (38). The latter mechanism allows cancer cells to continuously divide. Ogden et al. (15) demonstrated that the nuclear KIFC1 is a reliable predictive biomarker of patients’ survival in AA women with TNBC. Additional studies have associated the expression of KIFC1 with the increased risk of brain metastases in other types of neoplasms (39). This finding suggests a causal link between the KIFC1 protein and the aggressive progression of cancers. Hence, altogether, these results indicate that KIFC1 protein may be a promising prognostic biomarker candidate that may be very useful in predicting the risk of TNBC aggressiveness in AA independently of EGFR.

We previously published the effects of the combination of DOX+ABE in Rb-positive TNBC cells (MDA-MB-231) as synergistic and in Rb-negative TNBC cells (MDA-MB-468) as antagonistic (7). Here, we sought to further understand whether a change in KIFC1 levels following treatment with single agents DOX or ABE, and both agents in combination DOX+ABE reflects the nature of DOX and ABE interaction (synergism or antagonism) in both cell lines. The baseline levels of KIFC1 protein expression were similar in MDA-MB-231 and MDA-MB-468 cells (Figure 1). This result is in alignment with prior observations (40). The IC50 of DOX after 72 h exposure was 0.565 μM in MDA-MB-231 and 0.121 μM in MDA-MB-468 cells (7). These concentrations of DOX altered the expression of KIFC1 protein in both cell lines, albeit neither change was statistically significant compared to control. Next, the KFC1 protein expression was significantly reduced under ABE exposure in MDA-MB-231 cells but remained unchanged in MDA-MB-468cells. The addition of DOX to ABE resulted in a higher decrease in KIFC1 protein expression in MDA-MB-231 cells, although the latter was not statistically significant compared to the effects of ABE as a single agent. The time course profile of the KIFC1 protein expression following treatment with DOX+ABE also revealed that the time to achieve a full inhibition of KIFC1 expression is reduced compared to single agent ABE (Figure 2). Altogether, these findings suggest that the proposed combination therapy of ABE+DOX will result in a more rapid reduction of the prognostic biomarker KIFC1 in Rb-positive TNBC, but will not have an impact on Rb-negative TNBC.

TNBC cells with supernumerary centrosomes, including MDA-MB-231 and MDA-MB-468 cells (41), divide to produce DOX-resistant progeny through centrosomal clustering. Anthracycline-induced DNA damage increases centrosome clustering via up-regulation of the KIFC1 pathway (42). DOX exposure increased KIFC1 in both cell lines, whereas the addition of ABE decreased KIFC1 in MDA-MB-231 cells alone (Figure 1 and Figure 2). One hypothesis for the observed reduction in the expression of KIFC1 protein in Rb-positive TNBC is that it may result from a cooperative effect of ABE and DOX on the inhibition of the cell cycle during the replication phase (9,10). Indeed, the kinesin-14 family is primarily synthesized during the G2 phase of the cell cycle, then is rapidly degraded post-replication (43). An increase in cellular cytostasis following ABE+DOX combination treatment may inhibit the synthesis of the KIFC1 protein without affecting the basal KIFC1 degradation. This hypothesis is in support with the unanticipated increased expression of KIFC1 protein in MDA-MB-468 following exposure to ABE, since ABE exerts its activity on Rb-negative TNBC cells via mechanisms that are off-target rather than Rb-mediated cytostasis (44,45).

It is important to point out that our work presents a few limitations. First, the experiments presented here were conducted using one Rb-positive TNBC cell line, which is MDA-MB-231. In order to examine the hypothesis that the reduction of KIFC1 protein expression is specific to RB-positive TNBC, we may characterize the concentration effect of ABE on KIFC1 protein expression using a second Rb-positive mesenchymal-like TNBC cell line such as HS-578T or MDA-MB-157 (46). Second, it remains unclear whether the metastatic risk attributed to the nuclear KIFC1 activity occurs only during the mitosis phase or also occurs during the interphase (47,48). One of the suggested roles for the nuclear KIFC1 protein during interphase is keeping the integrity and organization of the Golgi apparatus (48) and aiding in vesicle transport and processing (49,50). Future examination of the KIFC1 protein degradation rates and KIFC1 interphase activity in TNBC are needed to elucidate the role of KIFC1 as a surrogate for mitosis activity or whether the KIFC1 protein expression is directly influenced by the Rb-CDK4/6 pathway.

Conclusion

In this work, we examined published negative prognostic biomarkers specific to the AA TNBC population. Biomarkers that met our criteria included Kaiso (16), ANXA2 (17), and KIFC1 (15). The combination of ABE+DOX produced the greatest mean reduction in KIFC1. Further, this combination resulted in a more rapid decay in the expression of KIFC1 protein than the corresponding single agents in Rb-positive TNBC, without affecting KIFC1 expression in Rb-negative TNBC. In conclusion, KIFC1 appears to have potential as a robust prognostic biomarker for TNBC in AA patients considered for treatment with the combination therapy of ABE+DOX.

Conflicts of Interest

Sihem Ait-Oudhia is currently employed by Merck & Co Inc, Kenilworth, New Jersey, USA. Brett Fleisher is currently employed by Genentech Inc, South San Francisco, CA, USA. The Authors declare that there are no conflicts of interest.

Authors’ Contributions

BF designed the study, conducted the experiments, and wrote the manuscript. CW, BJ, JV, KT helped conducting the experiments, SAO designed the study, provided scientific mentoring to all co-authors, and edited the manuscript.

Acknowledgements

The Authors acknowledge Dr. Folakemi Odedina for supporting the Comprehensive Cancer Research Training Opportunities for Outstanding Leaders (C-ReTOOL) Program and Maria Liebhart for technical assistance. This work was partly funded by the American Foundation for Pharmaceutical Education Pre-doctoral Award to Brett Fleisher (AFPE, VA, USA).

References

1 Kumar P & Aggarwal R An overview of triple-negative breast cancer. Arch Gynecol Obstet. 293(2) 247 - 269 2016. PMID: 26341644. DOI: 10.1007/s00404-015-3859-y
2 Sturtz LA Melley J Mamula K Shriver CD & Ellsworth RE Outcome disparities in African American women with triple negative breast cancer: a comparison of epidemiological and molecular factors between African American and Caucasian women with triple negative breast cancer. BMC Cancer. 14 62 2014. PMID: 24495414. DOI: 10.1186/1471-2407-14-62
3 Van Poznak C Harris LN & Somerfield MR Use of biomarkers to guide decisions on systemic therapy for women with metastatic breast cancer: American Society of Clinical Oncology clinical practice guideline. J Oncol Pract. 11(6) 514 - 516 2015. PMID: 29424583. DOI: 10.1200/JOP.2015.005215
4 Liedtke C Mazouni C Hess KR André F Tordai A Mejia JA Symmans WF Gonzalez-Angulo AM Hennessy B Green M Cristofanilli M Hortobagyi GN & Pusztai L Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol. 26(8) 1275 - 1281 2008. PMID: 18250347. DOI: 10.1200/JCO.2007.14.4147
5 Yu KD Zhu R Zhan M Rodriguez AA Yang W Wong S Makris A Lehmann BD Chen X Mayer I Pietenpol JA Shao ZM Symmans WF & Chang JC Identification of prognosis-relevant subgroups in patients with chemoresistant triple-negative breast cancer. Clin Cancer Res. 19(10) 2723 - 2733 2013. PMID: 23549873. DOI: 10.1158/1078-0432.CCR-12-2986
6 Pastoriza JM Karagiannis GS Lin J Lanjewar S Entenberg D Condeelis JS Sparano JA Xue X Rohan TE & Oktay MH Black race and distant recurrence after neoadjuvant or adjuvant chemotherapy in breast cancer. Clin Exp Metastasis. 35(7) 613 - 623 2018. PMID: 30136072. DOI: 10.1007/s10585-018-9932-8
7 Fleisher B Lezeau J Werkman C Jacobs B & Ait-Oudhia S In vitro to clinical translation of combinatorial effects of doxorubicin and abemaciclib in Rb-positive triple negative breast cancer: a systems-based pharmacokinetic/pharmacodynamic modeling approach. Breast Cancer (Dove Med Press). 13 87 - 105 2021. PMID: 33628047. DOI: 10.2147/BCTT.S292161
8 Tacar O Sriamornsak P & Dass CR Doxorubicin: an update on anticancer molecular action, toxicity and novel drug delivery systems. J Pharm Pharmacol. 65(2) 157 - 170 2013. PMID: 23278683. DOI: 10.1111/j.2042-7158.2012.01567.x
9 McClendon AK Dean JL Rivadeneira DB Yu JE Reed CA Gao E Farber JL Force T Koch WJ & Knudsen ES CDK4/6 inhibition antagonizes the cytotoxic response to anthracycline therapy. Cell Cycle. 11(14) 2747 - 2755 2012. PMID: 22751436. DOI: 10.4161/cc.21127
10 Knudsen ES Hutcheson J Vail P & Witkiewicz AK Biological specificity of CDK4/6 inhibitors: dose response relationship, in vivo signaling, and composite response signature. Oncotarget. 8(27) 43678 - 43691 2017. PMID: 28620137. DOI: 10.18632/oncotarget.18435
11 Camirand A Fadhil I Luco AL Ochietti B & Kremer RB Enhancement of taxol, doxorubicin and zoledronate anti-proliferation action on triple-negative breast cancer cells by a PTHrP blocking monoclonal antibody. Am J Cancer Res. 3(5) 500 - 508 2013. PMID: 24224127.
Pubmed |
12 Tapia MA González-Navarrete I Dalmases A Bosch M Rodriguez-Fanjul V Rolfe M Ross JS Mezquita J Mezquita C Bachs O Gascón P Rojo F Perona R Rovira A & Albanell J Inhibition of the canonical IKK/NF kappa B pathway sensitizes human cancer cells to doxorubicin. Cell Cycle. 6(18) 2284 - 2292 2007. PMID: 17890907. DOI: 10.4161/cc.6.18.4721
13 Echavarria I López-Tarruella S Picornell A García-Saenz JÁ Jerez Y Hoadley K Gómez HL Moreno F Monte-Millan MD Márquez-Rodas I Alvarez E Ramos-Medina R Gayarre J Massarrah T Ocaña I Cebollero M Fuentes H Barnadas A Ballesteros AI Bohn U Perou CM & Martin M Pathological response in a triple-negative breast cancer cohort treated with neoadjuvant carboplatin and docetaxel according to Lehmann’s refined classification. Clin Cancer Res. 24(8) 1845 - 1852 2018. PMID: 29378733. DOI: 10.1158/1078-0432.CCR-17-1912
14 Fleisher B Clarke C & Ait-Oudhia S Current advances in biomarkers for targeted therapy in triple-negative breast cancer. Breast Cancer (Dove Med Press). 8 183 - 197 2016. PMID: 27785100. DOI: 10.2147/BCTT.S114659
15 Ogden A Garlapati C Li XB Turaga RC Oprea-Ilies G Wright N Bhattarai S Mittal K Wetherilt CS Krishnamurti U Reid MD Jones M Gupta M Osan R Pattni S Riaz A Klimov S Rao A Cantuaria G Rida PC & Aneja R Multi-institutional study of nuclear KIFC1 as a biomarker of poor prognosis in African American women with triple-negative breast cancer. Sci Rep. 7 42289 2017. PMID: 28218233. DOI: 10.1038/srep42289
16 Bassey-Archibong BI Hercules SM Rayner LGA Skeete DHA Smith Connell SP Brain I Daramola A Banjo AAF Byun JS Gardner K Dushoff J & Daniel JM Kaiso is highly expressed in TNBC tissues of women of African ancestry compared to Caucasian women. Cancer Causes Control. 28(11) 1295 - 1304 2017. PMID: 28887687. DOI: 10.1007/s10552-017-0955-2
17 Gibbs LD Chaudhary P Mansheim K Hare RJ Mantsch RA & Vishwanatha JK ANXA2 expression in African American triple-negative breast cancer patients. Breast Cancer Res Treat. 174(1) 113 - 120 2019. PMID: 30478786. DOI: 10.1007/s10549-018-5030-5
18 Newman LA Griffith KA Jatoi I Simon MS Crowe JP & Colditz GA Meta-analysis of survival in African American and white American patients with breast cancer: ethnicity compared with socioeconomic status. J Clin Oncol. 24(9) 1342 - 1349 2006. PMID: 16549828. DOI: 10.1200/JCO.2005.03.3472
19 McShane LM Altman DG Sauerbrei W Taube SE Gion M Clark GM & Statistics Subcommittee of the NCI-EORTC Working Group on Cancer Diagnostics Reporting recommendations for tumour MARKer prognostic studies (REMARK). Br J Cancer. 93(4) 387 - 391 2005. PMID: 16106245. DOI: 10.1038/sj.bjc.6602678
20 Wang T Yuan J Zhang J Tian R Ji W Zhou Y Yang Y Song W Zhang F & Niu R Anxa2 binds to STAT3 and promotes epithelial to mesenchymal transition in breast cancer cells. Oncotarget. 6(31) 30975 - 30992 2015. PMID: 26307676. DOI: 10.18632/oncotarget.5199
21 Madame Curie Bioscience Database [Internet]. Austin (TX): Landes Bioscience; 2000-2013. Available at: https://www.ncbi.nlm.nih.gov/books/NBK5974.
22 Bassey-Archibong BI Rayner LG Hercules SM Aarts CW Dvorkin-Gheva A Bramson JL Hassell JA & Daniel JM Kaiso depletion attenuates the growth and survival of triple negative breast cancer cells. Cell Death Dis. 8(3) e2689 2017. PMID: 28333150. DOI: 10.1038/cddis.2017.92
23 Park HS Jang MH Kim EJ Kim HJ Lee HJ Kim YJ Kim JH Kang E Kim SW Kim IA & Park SY High EGFR gene copy number predicts poor outcome in triple-negative breast cancer. Mod Pathol. 27(9) 1212 - 1222 2014. PMID: 24406864. DOI: 10.1038/modpathol.2013.251
24 Jones J Wang H Karanam B Theodore S Dean-Colomb W Welch DR Grizzle W & Yates C Nuclear localization of Kaiso promotes the poorly differentiated phenotype and EMT in infiltrating ductal carcinomas. Clin Exp Metastasis. 31(5) 497 - 510 2014. PMID: 24570268. DOI: 10.1007/s10585-014-9644-7
25 Daniel JM & Reynolds AB The catenin p120(ctn) interacts with Kaiso, a novel BTB/POZ domain zinc finger transcription factor. Mol Cell Biol. 19(5) 3614 - 3623 1999. PMID: 10207085. DOI: 10.1128/MCB.19.5.3614
26 Kwiecien JM Bassey-Archibong BI Dabrowski W Rayner LG Lucas AR & Daniel JM Loss of Kaiso expression in breast cancer cells prevents intra-vascular invasion in the lung and secondary metastasis. PLoS One. 12(9) e0183883 2017. PMID: 28880889. DOI: 10.1371/journal.pone.0183883
27 Bassey-Archibong BI Kwiecien JM Milosavljevic SB Hallett RM Rayner LG Erb MJ Crawford-Brown CJ Stephenson KB Bédard PA Hassell JA & Daniel JM Kaiso depletion attenuates transforming growth factor-β signaling and metastatic activity of triple-negative breast cancer cells. Oncogenesis. 5 e208 2016. PMID: 26999717. DOI: 10.1038/oncsis.2016.17
28 Chaudhary P Thamake SI Shetty P & Vishwanatha JK Inhibition of triple-negative and Herceptin-resistant breast cancer cell proliferation and migration by Annexin A2 antibodies. Br J Cancer. 111(12) 2328 - 2341 2014. PMID: 25321192. DOI: 10.1038/bjc.2014.542
29 Zhang Y Bi J Zhu H Shi M & Zeng X ANXA2 could act as a moderator of EGFR-directed therapy resistance in triple negative breast cancer. Biosci Biotechnol Biochem. 82(10) 1733 - 1741 2018. PMID: 29912633. DOI: 10.1080/09168451.2018.1484275
30 Sharma M Ownbey RT & Sharma MC Breast cancer cell surface annexin II induces cell migration and neoangiogenesis via tPA dependent plasmin generation. Exp Mol Pathol. 88(2) 278 - 286 2010. PMID: 20079732. DOI: 10.1016/j.yexmp.2010.01.001
31 Shetty PK Thamake SI Biswas S Johansson SL & Vishwanatha JK Reciprocal regulation of annexin A2 and EGFR with Her-2 in Her-2 negative and herceptin-resistant breast cancer. PLoS One. 7(9) e44299 2012. PMID: 22957061. DOI: 10.1371/journal.pone.0044299
32 Gibbs LD & Vishwanatha JK Prognostic impact of AnxA1 and AnxA2 gene expression in triple-negative breast cancer. Oncotarget. 9(2) 2697 - 2704 2017. PMID: 29416802. DOI: 10.18632/oncotarget.23627
33 Wu J Mikule K Wang W Su N Petteruti P Gharahdaghi F Code E Zhu X Jacques K Lai Z Yang B Lamb ML Chuaqui C Keen N & Chen H Discovery and mechanistic study of a small molecule inhibitor for motor protein KIFC1. ACS Chem Biol. 8(10) 2201 - 2208 2013. PMID: 23895133. DOI: 10.1021/cb400186w
34 Pannu V Mittal K Cantuaria G Reid MD Li X Donthamsetty S McBride M Klimov S Osan R Gupta MV Rida PC & Aneja R Rampant centrosome amplification underlies more aggressive disease course of triple negative breast cancers. Oncotarget. 6(12) 10487 - 10497 2015. PMID: 25868856. DOI: 10.18632/oncotarget.3402
35 Kwon M Godinho SA Chandhok NS Ganem NJ Azioune A Thery M & Pellman D Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev. 22(16) 2189 - 2203 2008. PMID: 18662975. DOI: 10.1101/gad.1700908
36 Zhang W Zhai L Wang Y Boohaker RJ Lu W Gupta VV Padmalayam I Bostwick RJ White EL Ross LJ Maddry J Ananthan S Augelli-Szafran CE Suto MJ Xu B Li R & Li Y Discovery of a novel inhibitor of kinesin-like protein KIFC1. Biochem J. 473(8) 1027 - 1035 2016. PMID: 26846349. DOI: 10.1042/BJ20150992
37 Li Y Lu W Chen D Boohaker RJ Zhai L Padmalayam I Wennerberg K Xu B & Zhang W KIFC1 is a novel potential therapeutic target for breast cancer. Cancer Biol Ther. 16(9) 1316 - 1322 2015. PMID: 26177331. DOI: 10.1080/15384047.2015.1070980
38 Kleylein-Sohn J Pöllinger B Ohmer M Hofmann F Nigg EA Hemmings BA & Wartmann M Acentrosomal spindle organization renders cancer cells dependent on the kinesin HSET. J Cell Sci. 125(Pt 22) 5391 - 5402 2012. PMID: 22946058. DOI: 10.1242/jcs.107474
39 Grinberg-Rashi H Ofek E Perelman M Skarda J Yaron P Hajdúch M Jacob-Hirsch J Amariglio N Krupsky M Simansky DA Ram Z Pfeffer R Galernter I Steinberg DM Ben-Dov I Rechavi G & Izraeli S The expression of three genes in primary non-small cell lung cancer is associated with metastatic spread to the brain. Clin Cancer Res. 15(5) 1755 - 1761 2009. PMID: 19190132. DOI: 10.1158/1078-0432.CCR-08-2124
40 De S Cipriano R Jackson MW & Stark GR Overexpression of kinesins mediates docetaxel resistance in breast cancer cells. Cancer Res. 69(20) 8035 - 8042 2009. PMID: 19789344. DOI: 10.1158/0008-5472.CAN-09-1224
41 Mittal K Choi DH Ogden A Donthamsetty S Melton BD Gupta MV Pannu V Cantuaria G Varambally S Reid MD Jonsdottir K Janssen EA Aleskandarany MA Ellis IO Rakha EA Rida PC & Aneja R Amplified centrosomes and mitotic index display poor concordance between patient tumors and cultured cancer cells. Sci Rep. 7 43984 2017. PMID: 28272508. DOI: 10.1038/srep43984
42 Fan G Sun L Meng L Hu C Wang X Shi Z Hu C Han Y Yang Q Cao L Zhang X Zhang Y Song X Xia S He B Zhang S & Wang C The ATM and ATR kinases regulate centrosome clustering and tumor recurrence by targeting KIFC1 phosphorylation. Nat Commun. 12(1) 20 2021. PMID: 33397932. DOI: 10.1038/s41467-020-20208-x
43 Yount AL Zong H & Walczak CE Regulatory mechanisms that control mitotic kinesins. Exp Cell Res. 334(1) 70 - 77 2015. PMID: 25576382. DOI: 10.1016/j.yexcr.2014.12.015
44 Asghar US Barr AR Cutts R Beaney M Babina I Sampath D Giltnane J Lacap JA Crocker L Young A Pearson A Herrera-Abreu MT Bakal C & Turner NC Single-cell dynamics determines response to CDK4/6 inhibition in triple-negative breast cancer. Clin Cancer Res. 23(18) 5561 - 5572 2017. PMID: 28606920. DOI: 10.1158/1078-0432.CCR-17-0369
45 O’Brien N Conklin D Beckmann R Luo T Chau K Thomas J Mc Nulty A Marchal C Kalous O von Euw E Hurvitz S Mockbee C & Slamon DJ Preclinical activity of abemaciclib alone or in combination with antimitotic and targeted therapies in breast cancer. Mol Cancer Ther. 17(5) 897 - 907 2018. PMID: 29483214. DOI: 10.1158/1535-7163.MCT-17-0290
46 Robinson TJ Liu JC Vizeacoumar F Sun T Maclean N Egan SE Schimmer AD Datti A & Zacksenhaus E RB1 status in triple negative breast cancer cells dictates response to radiation treatment and selective therapeutic drugs. PLoS One. 8(11) e78641 2013. PMID: 24265703. DOI: 10.1371/journal.pone.0078641
47 Cai S Weaver LN Ems-McClung SC & Walczak CE Kinesin-14 family proteins HSET/XCTK2 control spindle length by cross-linking and sliding microtubules. Mol Biol Cell. 20(5) 1348 - 1359 2009. PMID: 19116309. DOI: 10.1091/mbc.e08-09-0971
48 She ZY Pan MY Tan FQ & Yang WX Minus end-directed kinesin-14 KIFC1 regulates the positioning and architecture of the Golgi apparatus. Oncotarget. 8(22) 36469 - 36483 2017. PMID: 28430595. DOI: 10.18632/oncotarget.16863
49 Dorner C Ciossek T Müller S Møller PH Ullrich A & Lammers R Characterization of KIF1C, a new kinesin-like protein involved in vesicle transport from the Golgi apparatus to the endoplasmic reticulum. J Biol Chem. 273(32) 20267 - 20275 1998. PMID: 9685376. DOI: 10.1074/jbc.273.32.20267
50 Nath S Bananis E Sarkar S Stockert RJ Sperry AO Murray JW & Wolkoff AW Kif5B and Kifc1 interact and are required for motility and fission of early endocytic vesicles in mouse liver. Mol Biol Cell. 18(5) 1839 - 1849 2007. PMID: 17360972. DOI: 10.1091/mbc.e06-06-0524