Loading

Review Article Open Access
Volume 2 | Issue 3 | DOI: https://doi.org/10.33696/Signaling.2.053

TTK: A Promising Target in Malignant Tumors

  • 1Graduate Department, Bengbu Medical College, Bengbu, Anhui, 23300, PR China
  • 2Oncology Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, PR China
  • #These authors contribute equally to the review
+ Affiliations - Affiliations

*Corresponding Author

Xiaodong Liang, lxdctopone@sina.com
Haibo Zhang, zhbdoctor@163.com

Received Date: July 12, 2021

Accepted Date: September 15, 2021

Abstract

TTK is a dual-specific protein kinase that phosphorylates serine and threonine. The spindle assembly checkpoint (SAC) acts as a molecular monitoring mechanism that regulates mitosis and ensures accurate separation of chromosomes. TTK is a key regulator of the SAC. Activated TTK localizes to kinetochores and activates SAC function. Simultaneously, TTK promotes error correction by phosphorylating several substrates. When all chromosomes are correctly aligned, TTK is separated from kinetochores, and SAC function is turned off, thus initiating the anaphase of mitosis. TTK is highly expressed in various malignant tumors and is negatively associated with survival. TTK inhibition inhibits malignant tumor cells growth, and the combination of TTK inhibition and chemotherapy enhances anti-tumor effects in vitro. The combination of TTK inhibitors and chemotherapy increased efficacy without increasing adverse reactions. The addition of TTK inhibition also enhanced the effect of radiotherapy both in vitro and in vivo. The combination of TTK inhibition and radiotherapy results in more DNA double-strand damage and mitotic catastrophe, with the involved mechanisms including homologous recombination pathways and non-homologous end-joining pathways. The mechanism of combination therapy is complex and requires further study. Five small-molecule TTK inhibitors have been tested in clinical trials and were used in combination with paclitaxel because the combination could enhance the killing of cancer cells and alleviate side effects by reducing the drug dosage. Because of its high expression in malignant tumors, TTK is expected to be developed as an effective anti-tumor vaccine. TTK is a promising treatment target and further investigation is warranted.

Keywords

TTK, Cancer, SAC, Inhibitor, Vaccine, Mitotic catastrophe

Introduction

TTK, also known as MPS1 (the monopolar spindle 1)/ MPS1L1, is located on chromosome 6q13-q21 and encodes a dual-specific protein kinase that phosphorylates serine and threonine [1]. The spindle assembly checkpoint (SAC) plays a key role in mitosis. The SAC acts as a molecular monitoring mechanism, which delays mitosis until all chromosomes are properly attached to the spindle microtubules. As a key regulator of the SAC, TTK plays an important role in controlling cell cycle progression and maintaining genomic integrity [2]. TTK is vital for the recruitment of kinetochore components to unattached kinetochores and is essential for correcting improperly attached chromosomes. Interestingly, TTK is highly expressed in many types of malignant tumors [3]. However, TTK expression is low in most organs, except in the testis and placenta. Once TTK is inhibited, cancer cells exit mitosis prematurely, with more chromosome segregation errors and aneuploids. After several rounds of cell division, the accumulation of chromosome segregation errors may lead to cancer cell death [4]. Therefore, TTK has gradually become a research hotspot for anticancer drugs, and TTK inhibitors are increasingly being investigated in clinical trials.

The combination of TTK inhibitors and radiation therapy can increase mitotic errors, promote aneuploidy formation, inhibit DNA damage repair, and ultimately lead to mitotic catastrophe and necrosis [5]. Studies have also shown that the combination of TTK inhibitors and radiotherapy can improve the efficacy in breast cancer, liver cancer, and brain glioma treatment [6-8]. Therefore, it is necessary to further study the molecular mechanism of combining radiation therapy and TTK inhibitors in various tumors to develop better strategies for treatment.

The Role of TTK in Mitosis

Cell mitosis is a basic activity that maintains the normal function of organs and tissues. The SAC is a key surveillance and correction system for mitosis. The SAC is activated when chromosomes are not properly captured by the spindle [9]. TTK is the key regulator of SAC function and plays an important role in the mitotic process. Once activated in early mitosis, TTK localizes to kinetochores and phosphorylates multiple residues on SAC proteins, thus activating SAC function. Simultaneously, TTK promotes error correction by phosphorylating several substrates [10,11]. When the SAC signal is activated, a mitotic checkpoint complex (MCC) is formed [12]. TTK also plays a key role in the assembly of the interphase MCC [13]. Subsequently, the anaphase-promoting complex (APC/C) function is inhibited by the MCC, and the initiation of anaphase mitosis is delayed [12,14-19]. When all chromosomes are correctly aligned, TTK is separated from the kinetochores, and the SAC function is turned off. Then, the MCC is dissociated, and the APC/C is activated, which initiates anaphase of mitosis [20-28].

TTK is a Promising Target in Malignant Tumors

TTK is highly expressed in many types of malignant tumors. However, TTK expression is low in most organs, except in the testis and placenta [3]. In addition, high expression of TTK has been found to be associated with poor prognosis in various malignant tumors. Therefore, TTK is potentially specific antitumor target. Preclinical studies have shown that TTK inhibitors or TTK knockdown-induced mitotic aberrancies inhibit the growth of malignant tumor cells [29-31]. Moreover, TTK inhibition combined with chemotherapy has a synergistic effect in some malignant tumors [32-34]. Because of the difference in expression between malignant tumors and normal tissues, the combination of TTK inhibitors and paclitaxel increased the efficacy without increasing the adverse reactions in animal models [35].

Glioma

TTK is significantly elevated in gliomas. Overexpression of TTK was positively associated with tumor grade and negatively associated with survival in patients with glioma [32,36]. TTK knockdown resulted in mitotic aberrancies and inhibited the proliferation of glioblastoma cells. MPS1-IN-3, a selective inhibitor of TTK, caused gross chromosome segregation defects in glioblastoma cells. In addition, inhibition of TTK with MPS1-IN-3 increased the sensitivity of glioblastoma cells to vincristine both in vitro and in vivo. Moreover, the combination of MPS1-IN-3 with vincristine did not increase toxicity in animal models.

Breast cancer

Daniel et al. found that the expression levels of TTK mRNA and protein were elevated and correlated positively with aneuploidy and basal-like phenotype in breast cancer. Expression levels of TTK were positively correlated with tumor grade, p53 mutation, and poor survival. TTK knockdown caused aberrant mitosis and induced apoptosis both in vitro and in vivo. TTK inhibition tends to selectively kill cancer cells with high aneuploidy. Maire et al. reported that TTK was overexpressed in breast cancer patients, regardless of the histological type. However, the expression level of TTK was higher in triple-negative breast cancer, whereas TTK was not detected in healthy breast tissues. TTK knockdown induced mitotic catastrophe and apoptosis in triple-negative breast cancer cells. NTRC 0066-0, a selective TTK inhibitor, was developed by Maia et al. NTRC 0066-0 was found to inhibit tumor growth both in vitro and in vivo. Furthermore, simultaneous administration of NTRC 0066-0 with docetaxel extended survival and tumor remission without toxicity in a triplenegative breast cancer mouse model [33,37-39].

Hepatocellular carcinoma

Chemotherapy is usually ineffective in patients with advanced liver cancer with limited life expectancy. Sorafenib is often prescribed, and drug resistance soon emerges. Our previous study showed that TTK is significantly overexpressed in hepatocellular carcinoma tissues. TTK overexpression promotes hepatocellular carcinoma cell proliferation and resistance to sorafenib in vitro and in vivo. In contrast, TTK knockdown inhibits cell growth and reduces resistance to sorafenib in hepatocellular carcinoma (HCC) cells [40]. Multi-omics analysis showed that TTK mRNA levels were negatively correlated with relapse-free survival (RFS) and overall survival (OS) in patients with HCC after surgery. This indicates its potential as a prognostic biomarker [41]. It has been reported that TTK is overexpressed in 77.63% of HCC specimens, and the increased TTK expression is closely associated with tumor size and the presence of portal vein tumor thrombus. Demethylation of the TTK promoter can increase its expression in HCC. In vitro studies have shown that TTK can induce cell proliferation, colony formation and migration of HCC cells, thereby increasing the degree of malignancy of the tumor. Further studies have shown that TTK activates the Akt/mTOR pathway through p53. TTK inhibitors can inhibit the growth of HCC cells [42]. Therefore, TTK has potential therapeutic value for HCC, which encouraged us to conduct further clinical studies on TTK inhibitors in the treatment of HCC.

Pancreatic cancer

TTK is associated with poor survival of pancreatic ductal adenocarcinoma (PDAC) cells. Compared with normal pancreatic cells, PDAC is more sensitive to TTK inhibition [30]. Kaistha et al. found that TTK expression is significantly elevated in PDAC. TTK plays an important role in the growth and proliferation of PDAC cells. Loss of TTK activity can induce cell death through chromosome segregation errors. In contrast, immortalized normal pancreatic hTERT-HPNE cell lines were unaffected by TTK activity [40].

Prostate cancer

TTK inhibition has been reported to be significantly associated with the recurrence of prostate cancer, and in androgen receptor-positive prostate cancer cells, TTK inhibition enhances the antiproliferative effects of antiandrogens [43,44]. Therefore, TTK has potential clinical value in the treatment of prostate cancer.

Melanoma

Liu et al. found that in melanoma, activated B-Raf (V600E) prevented TTK degradation and increased the amount of TTK protein, which led to centrosome amplification and incorrect chromosomal segregation [45]. B-Raf/MEK/ERK signaling and Mps1/Akt constitute an automatically regulated negative feedback loop in melanoma cells. Oncogenic B-RAF (V600E) can eliminate the negative feedback loop and cause TTK dysfunction to induce chromosome instability and tumorigenesis [46]. Therefore, it has been suggested that combined targeted therapy of B-RAF and TTK should be applied in clinical practice.

Lung cancer

Suda et al. established a strong cytotoxic T lymphocytes (CTL) clone stimulated by TTK-567 with specific killing activity against HLA-A24-positive lung cancer and esophageal cancer cells [47]. This feature illustrates the potential of TTK as a cancer vaccine. TTK peptide vaccine has also been shown to be safe and well-tolerated in latestage clinical trials in patients with advanced or recurrent non-small cell lung cancer (NSCLC) [48].

Esophageal cancer

Mizukami et al. found that in patients with esophageal squamous cell carcinoma (ESCC), TTK antigen can induce a specific T cell response [49]. In clinical trials, the efficacy and safety of the TTK vaccine for advanced esophageal cancer have been confirmed [50]. This will encourage us to further study the use of a TTK tumor vaccine in advanced ESCC.

Biliary tract cancer

In clinical trials, the TTK peptide vaccine induced specific T cell immune responses in patients with advanced biliary tract cancer (BTC) and achieved good efficacy. This will lead us to regard TTK vaccination therapy as one of the few treatment options for advanced BTC [51].

TTK Inhibitors

Researchers have used small molecule compounds to block the function of the SAC by inhibiting TTK activity, resulting in the disruption of mitotic stability. This feature can be exploited to develop cancer treatment strategies. To date, several types of small-molecule compounds that inhibit TTK activity have been identified or developed. These small molecule compounds can be divided into four broad groups, including N-phenylpyrimidin-2-amine, N-phenylpyridine, heterocyclic small compounds in 3-phenylindazole, and 5-membered bridged six-membered bridged [52] (Table 1). Based on the mechanism of action of TTK inhibitors, researchers used TTK inhibitors in combination with microtubule-targeting agents (MTAs) to increase chromosomal separation errors and kill cancer cells more efficiently. Some studies have shown that TTK inhibitors combined with chemotherapeutic agents can effectively increase the killing effect of tumor cells. Wu et al. found that CC-671 (a highly selective inhibitor of TTK and CLK2) could inhibit the drug efflux activity of ABCG2 in lung cancer cells, thus increasing the level of intracellular chemotherapy drugs. This will help improve the efficacy of chemotherapy in lung cancer [34]. Tannous et al. developed a selective small molecule inhibitor of TTK, Mps1-IN-3, which caused mitotic abnormalities in glioblastoma cells, and the combination of Mps1-IN-3 and vincristine increased aneuploidy and cell death. In addition, Mps1- IN-3 increases the susceptibility of glioblastoma cells to mitotic drugs [32]. Similarly, inhibition of TTK enhanced the efficacy of docetaxel in a triple-negative breast cancer model [33].

Chemical structure Drug name
1. Compounds with N-phenylpyrimidin-2-amine scaffolds Reversine, MPI-0479605, Mps1-IN-3, AZ 3146, CC-671, BOS- 172722, Mps1-IN-2, NTRC 0066-0, NMS-P715 et al.
2. Compounds with N-phenylpyridine scaffolds TC Mps112, Mps1-IN-1, CCT251455 et al.
3. Compounds with 3-phenylindazole scaffold SP600125, CFI-400936, CFI-401870 et al.
4. Compounds with five-membered bridged six-membered heterocyclic scaffolds Mps-BAY1, Mps-BAY2a, Mps-BAY2b, BAY 1161909, BAY 1217389, CFI-402257, PF-7006, PF-3837 et al.

Table 1: TTK Inhibitors Classified by Chemical Structure.

TTK Inhibitors in Clinical Trials

There are currently five small-molecule TTK inhibitors in clinical trials, namely BAY-1217389 [35,53,54], BAY- 1161909 [35,53,54], BOS-172722 [55,56], CFI-402257 [2,57-60], and S-81694 (Table 2). Among them, BAY- 1217389, BOS-172722, and S-81694 completed phase I or II trials. The CFI-402257-related clinical trial is still in its recruitment phase. However, clinical trials on BAY- 1161909 have been terminated. All five inhibitors were used in combination with paclitaxel, because the combination not only increased the sensitivity of cancer cells, but also reduced side effects by reducing the dose of the drug. It is worth noting that determining the tolerance and toxicity due to long-term use of TTK inhibitors requires further study [61,62]. When cancer cells are resistant to one TTK inhibitor, they can be combined with several other sensitive TTK inhibitors to solve the problem of resistance. TTK inhibitors have unique advantages for tumor therapy. TTK inhibitors can target excessive TTK in tumors with little effect on normal cells, and can improve the sensitivity of cancer cells to paclitaxel and other chemotherapy drugs. Therefore, TTK inhibitors can be used in combination with existing anti-tumor drugs or treatment regimens to enhance tumor killing and improve survival in cancer patients.

Drug name Study Title Disease Intervention Phase NCT Identifier
BAY-1217389
(Completed)
Phase I Study of Oral BAY- 1217389 in Combination With Intravenous Paclitaxel Advanced malignancies (solid tumors) BAY-1217389 +
paclitaxel
I NCT02366949
BAY-1161909
(Terminated)
Phase I Dose Escalation of Oral BAY-1161909 in Combination With Intravenous Paclitaxel Advanced malignancies (solid tumors) BAY-1161909 +
paclitaxel
I NCT02138812
BOS-172722
(Completed)
Study of Paclitaxel in Combination With BOS-172722 in Patients With Advanced Nonhaematologic Malignancies Advanced Nonhaematologic Malignancies (ANM) BOS-172722 +
paclitaxel
I NCT03328494
CFI-402257
(Recruiting)
A Study of Investigational Drug CFI-402257 in Patients With Advanced Solid Tumors Advanced Solid Cancers (ASC), Breast Cancer CFI-402257 +
Fulvestrant
I NCT02792465
CFI-402257 in Combination With Paclitaxel in Patients With Advanced/Metastatic HER2- Negative Breast Cancer Breast Cancer CFI-402257 +
paclitaxel
II NCT03568422
S-81694
(Completed)
S-81694 Plus Paclitaxel in Metastatic Breast Cancer Metastatic Breast Cancer (mBC) S-81694 +
paclitaxel
I NCT03411161
Metastatic Triple Negative Breast Cancer (mTNBC) S-81694 +
paclitaxel
II

Table 2: TTK Inhibitors in Clinical Trials.

Inhibition of TTK Improve Radiosensitivity in Malignant Tumors

Radiotherapy is an important treatment method for malignant tumors. Both TTK and radiotherapy affect mitosis and DNA damage repair, and the combination of TTK inhibition and radiotherapy may have a synergistic effect. Some preclinical studies have investigated such effects and their mechanisms in malignant tumor models (Table 3).

Breast Cancer [6,65] Impair DNA damage repair Enhance mitotic catastrophe Impair homologous recombination
Glioblastoma [7,63,64] Impair DNA damage repair Enhance mitotic catastrophe
Impair homologous recombination and non-homologous end joining
Induces tumor suppressor PDCD4 and MSH2 through miR-21
HLF/miR-132/TTK axis regulates radiosensitivity of glioma cells
Liver Cancer [8] Impair DNA damage repair Enhance mitotic catastrophe
Up-regulating p21 and increasing G2/M blocking

Table 3: The mechanism of combination of TTK inhibition and radiotherapy.

Gliomas are most widely reported in this field. Maachani et al. found that a TTK inhibitor, NMS-P715, increased the radiosensitivity of glioblastoma (GBM) cells by reducing DNA double-strand break repair and inducing mitotic catastrophe after radiation. The involved mechanisms included both homologous recombination and nonhomologous end-joining pathways in that study [7]. Further studies showed that TTK inhibition regulates the tumor suppressors PDCD4 and MSH2 through miR- 21. MiR-21 has been shown to be elevated after radiation and mediates the radiation resistance of glioblastoma cells by regulating PDCD4 and MSH2 [63]. Another study found that hepatic leukemic factor (HLF) can inhibit the expression of TTK through miR-132, thereby inhibiting the proliferation, metastasis, and radiation resistance in GBM cells [64].

The combination of TTK and radiotherapy has also attracted attention in other cancers. Our recent study showed that the inhibition of TTK blocked G2/M transition in the cell cycle by upregulating p21 and enhancing radiosensitivity in liver cancer cells [8]. We also found that the combination of TTK inhibition and radiotherapy enhanced mitotic catastrophe and DNA damage. This opens up a new field for the treatment of liver cancer. Interestingly, low-dose (non-cytotoxic) TTK inhibitors showed radiosensitization in this study. The combination of low-dose TTK inhibitors and radiotherapy is expected to improve tumor control without increasing adverse reactions, which warrants further study. In addition, TTK may be responsible for radiation resistance in patients with basal breast cancer. Chandler et al. found that radiosensitivity was enhanced by the inhibition of TTK both in vitro and in vivo in basal-like breast cancer cells. TTK inhibition leads to unrepaired double-stranded DNA damage by reducing the repair efficiency of the homologous recombination system. However, nonhomologous endjoining was not considered in this study [6,65].

Conclusions

TTK is the key regulator of the SAC, which regulates mitosis and ensures the accurate separation of chromosomes. TTK is highly expressed in various malignant tumors, while its expression is low in most normal organs. TTK is positively associated with grade and associated negatively with survival in various malignant tumors. Preclinical studies show that TTK inhibition induces mitotic aberrancies and high aneuploidy and inhibits the growth of malignant tumor cells. The combination of TTK inhibitors and chemotherapy increases efficacy without increasing adverse reactions in animal models. The addition of TTK inhibition also enhanced the effects of radiotherapy. The combination of TTK inhibition and radiotherapy results in more DNA double-strand damage and mitotic catastrophe. Involved mechanisms may include homologous recombination, non-homologous end joining pathways, or both. The mechanism of combination therapy has not yet been clarified. Several types of smallmolecule TTK inhibitors have been developed. Five smallmolecule TTK inhibitors have been tested in clinical trials. All five inhibitors were used in combination with paclitaxel because the combination may not only enhance the killing of cancer cells, but also alleviate side effects by reducing the dose of paclitaxel (Figure 1). Because of its high specific expression in malignant tumors, TTK is expected to be developed as an effective anti-tumor vaccine. The efficacy and safety of the TTK vaccine have been tested in advanced esophageal, biliary tract, and lung cancer. In conclusion, targeting TTK has broad prospects, not only is the TTK inhibitor expected to be combined with radiotherapy and chemotherapy safely to further improve the curative effect, but the TTK vaccine is expected to be effective in the treatment of malignant tumors. However, the biomarkers for predicting the efficacy of TTK inhibitors are still lacking, the specific molecular mechanism of TTK inhibitors needs to be further studied, and the drug resistance mechanism of TTK inhibitors is not well understood. The sequence of radiotherapy and chemotherapy combined with TTK inhibitors and the adjustment of dose also need to be further studied.

Declaration of Competing Interest

The authors have no interest to declare.

Acknowledgments

This study was supported in part by grants from the Zhejiang Province Medical and Health Care Project (2019KY279 to Xiaodong Liang) and the National Natural Science Foundation of China (Grant number:82003236 to Haibo Zhang).

References

1. Hiruma Y, Koch A, Hazraty N, Tsakou F, Medema RH, Joosten RP, et al. Understanding inhibitor resistance in Mps1 kinase through novel biophysical assays and structures. Journal of Biological Chemistry. 2017 Sep 1;292(35):14496-504.

2. Thu KL, Silvester J, Elliott MJ, Ba-Alawi W, Duncan MH, Elia AC, et al. Disruption of the anaphase-promoting complex confers resistance to TTK inhibitors in triplenegative breast cancer. Proceedings of the National Academy of Sciences. 2018 Feb 13;115(7):E1570-1577.

3. Xie Y, Wang A, Lin J, Wu L, Zhang H, Yang X, et al. Mps1/TTK: a novel target and biomarker for cancer. Journal of Drug Targeting. 2017 Feb 7;25(2):112-118.

4. Foley EA, Kapoor TM. Microtubule attachment and spindle assembly checkpoint signalling at the kinetochore. Nature Reviews Molecular Cell Biology. 2013 Jan;14(1):25-37.

5. Lara-Gonzalez P, Westhorpe FG, Taylor SS. The spindle assembly checkpoint. Current Biology. 2012 Nov 20;22(22):R966-80.

6. Chandler BC, Moubadder L, Ritter CL, Liu M, Cameron M, Wilder-Romans K, et al. TTK inhibition radiosensitizes basal-like breast cancer through impaired homologous recombination. The Journal of Clinical Investigation. 2020 Feb 3;130(2):958-73.

7. Maachani UB, Kramp T, Hanson R, Zhao S, Celiku O, Shankavaram U, et al. Targeting MPS1 enhances radiosensitization of human glioblastoma by modulating DNA repair proteins. Molecular Cancer Research. 2015 May 1;13(5):852-62.

8. Zhang H, Yao W, Zhang M, Lu Y, Tang J, Jiang M, et al. TTK inhibitor promotes radiosensitivity of liver cancer cells through p21. Biochemical and Biophysical Research Communications. 2021 Apr 23;550:84-91.

9. Rieder CL, Cole RW, Khodjakov A, Sluder G. The checkpoint delaying anaphase in response to chromosome monoorientation is mediated by an inhibitory signal produced by unattached kinetochores. The Journal of Cell Biology. 1995 Aug 15;130(4):941-8.

10. Maure JF, Kitamura E, Tanaka TU. Mps1 kinase promotes sister-kinetochore bi-orientation by a tensiondependent mechanism. Current Biology. 2007 Dec 18;17(24):2175-82.

11. Dou Z, Liu X, Wang W, Zhu T, Wang X, Xu L, et al. Dynamic localization of Mps1 kinase to kinetochores is essential for accurate spindle microtubule attachment. Proceedings of the National Academy of Sciences. 2015 Aug 18;112(33):E4546-55.

12. Sudakin V, Chan GK, Yen TJ. Checkpoint inhibition of the APC/C in HeLa cells is mediated by a complex of BUBR1, BUB3, CDC20, and MAD2. The Journal of Cell Biology. 2001 Sep 3;154(5):925-36.

13. Maciejowski J, George KA, Terret ME, Zhang C, Shokat KM, Jallepalli PV. Mps1 directs the assembly of Cdc20 inhibitory complexes during interphase and mitosis to control M phase timing and spindle checkpoint signaling. Journal of Cell Biology. 2010 Jul 12;190(1):89-100.

14. Braunstein I, Miniowitz S, Moshe Y, Hershko A. Inhibitory factors associated with anaphase-promoting complex/cylosome in mitotic checkpoint. Proceedings of the National Academy of Sciences. 2007 Mar 20;104(12):4870-4875.

15. Chan GK, Jablonski SA, Sudakin V, Hittle JC, Yen TJ. Human BUBR1 is a mitotic checkpoint kinase that monitors CENP-E functions at kinetochores and binds the cyclosome/APC. The Journal of Cell Biology. 1999 Sep 6;146(5):941-54.

16. Herzog F, Primorac I, Dube P, Lenart P, Sander B, Mechtler K, et al. Structure of the anaphase-promoting complex/cyclosome interacting with a mitotic checkpoint complex. Science. 2009 Mar 13;323(5920):1477-81.

17. Musacchio A, Hardwick KG. The spindle checkpoint: structural insights into dynamic signalling. Nature reviews Molecular Cell Biology. 2002 Oct;3(10):731-41.

18. Peters JM. The anaphase promoting complex/ cyclosome: a machine designed to destroy. Nature reviews Molecular cell biology. 2006 Sep;7(9):644-56.

19. D’Angiolella V, Mari C, Nocera D, Rametti L, Grieco D. The spindle checkpoint requires cyclin-dependent kinase activity. Genes & Development. 2003 Oct 15;17(20):2520-5.

20. Howell BJ, McEwen BF, Canman JC, Hoffman DB, Farrar EM, Rieder CL, et al. Cytoplasmic dynein/dynactin drives kinetochore protein transport to the spindle poles and has a role in mitotic spindle checkpoint inactivation. The Journal of Cell Biology. 2001 Dec 24;155(7):1159-72.

21. Morrow CJ, Tighe A, Johnson VL, Scott MI, Ditchfield C, Taylor SS. Bub1 and aurora B cooperate to maintain BubR1-mediated inhibition of APC/CCdc20. Journal of Cell Science. 2005 Aug 15;118(16):3639-52.

22. Hardwick KG, Johnston RC, Smith DL, Murray AW. MAD3 encodes a novel component of the spindle checkpoint which interacts with Bub3p, Cdc20p, and Mad2p. The Journal of Cell Biology. 2000 Mar 6;148(5):871-82.

23. Tang Z, Bharadwaj R, Li B, Yu H. Mad2-Independent inhibition of APCCdc20 by the mitotic checkpoint protein BubR1. Developmental Cell. 2001 Aug 1;1(2):227-37.

24. Poddar A, Stukenberg PT, Burke DJ. Two complexes of spindle checkpoint proteins containing Cdc20 and Mad2 assemble during mitosis independently of the kinetochore in Saccharomyces cerevisiae. Eukaryotic Cell. 2005 May;4(5):867-78.

25. Fang G, Yu H, Kirschner MW. The checkpoint protein MAD2 and the mitotic regulator CDC20 form a ternary complex with the anaphase-promoting complex to control anaphase initiation. Genes & Development. 1998 Jun 15;12(12):1871-83.

26. Fraschini R, Beretta A, Sironi L, Musacchio A, Lucchini G, Piatti S. Bub3 interaction with Mad2, Mad3 and Cdc20 is mediated by WD40 repeats and does not require intact kinetochores. The EMBO Journal. 2001 Dec 3;20(23):6648-59.

27. Wassmann K, Benezra R. Mad2 transiently associates with an APC/p55Cdc complex during mitosis. Proceedings of the National Academy of Sciences. 1998 Sep 15;95(19):11193-8.

28. Millband DN, Hardwick KG. Fission yeast Mad3p is required for Mad2p to inhibit the anaphase-promoting complex and localizes to kinetochores in a Bub1p-, Bub3p-, and Mph1p-dependent manner. Molecular and Cellular Biology. 2002 Apr 15;22(8):2728-42.

29. Choi M, Min YH, Pyo J, Lee CW, Jang CY, Kim JE. TC Mps1 12, a novel Mps1 inhibitor, suppresses the growth of hepatocellular carcinoma cells via the accumulation of chromosomal instability. British Journal of Pharmacology. 2017 Jun;174(12):1810-25.

30. Slee RB, Grimes BR, Bansal R, Gore J, Blackburn C, Brown L, et al. Selective inhibition of pancreatic ductal adenocarcinoma cell growth by the mitotic MPS1 kinase inhibitor NMS-P715. Molecular Cancer Therapeutics. 2014 Feb 1;13(2):307-15.

31. Faisal A, Mak GW, Gurden MD, Xavier CP, Anderhub SJ, Innocenti P, et al. Characterisation of CCT271850, a selective, oral and potent MPS1 inhibitor, used to directly measure in vivo MPS1 inhibition vs therapeutic efficacy. British Journal of Cancer. 2017 Apr;116(9):1166-76.

32. Tannous BA, Kerami M, Van der Stoop PM, Kwiatkowski N, Wang J, Zhou W, et al. Effects of the selective MPS1 inhibitor MPS1-IN-3 on glioblastoma sensitivity to antimitotic drugs. Journal of the National Cancer Institute. 2013 Sep 4;105(17):1322-31.

33. Maia AR, de Man J, Boon U, Janssen A, Song JY, Omerzu M, et al. Inhibition of the spindle assembly checkpoint kinase TTK enhances the efficacy of docetaxel in a triple-negative breast cancer model. Annals of Oncology. 2015 Oct 1;26(10):2180-92.

34. Wu ZX, Yang Y, Wang G, Wang JQ, Teng QX, Sun L, et al. Dual TTK/CLK2 inhibitor, CC-671, selectively antagonizes ABCG2-mediated multidrug resistance in lung cancer cells. Cancer Science. 2020 Aug;111(8):2872.

35. Wengner AM, Siemeister G, Koppitz M, Schulze V, Kosemund D, Klar U, Stoeckigt D, Neuhaus R, Lienau P, Bader B, Prechtl S. Novel Mps1 kinase inhibitors with potent antitumor activity. Molecular Cancer Therapeutics. 2016 Apr 1;15(4):583-92.

36. Bie L, Zhao G, Cheng P, Rondeau G, Porwollik S, Ju Y, et al. The accuracy of survival time prediction for patients with glioma is improved by measuring mitotic spindle checkpoint gene expression. PloS One. 2011 Oct 12;6(10):e25631.

37. Daniel J, Coulter J, Woo JH, Wilsbach K, Gabrielson E. High levels of the Mps1 checkpoint protein are protective of aneuploidy in breast cancer cells. Proceedings of the National Academy of Sciences. 2011 Mar 29;108(13):5384- 9.

38. Maire V, Baldeyron C, Richardson M, Tesson B, Vincent-Salomon A, Gravier E, Marty-Prouvost B, De Koning L, Rigaill G, Dumont A, Gentien D. TTK/hMPS1 is an attractive therapeutic target for triple-negative breast cancer. PloS One. 2013 May 20;8(5):e63712.

39. Gyorffy B, Bottai G, Lehmann-Che J, Kéri G, Orfi L, Iwamoto T, et al. TP53 mutation-correlated genes predict the risk of tumor relapse and identify MPS1 as a potential therapeutic kinase in TP53-mutated breast cancers. Molecular oncology. 2014 May 1;8(3):508-19.

40. Liang XD, Dai YC, Li ZY, Gan MF, Zhang SR, Lu HS, Cao XQ, Zheng BJ, Bao LF, Wang DD, Zhang LM. Expression and function analysis of mitotic checkpoint genes identifies TTK as a potential therapeutic target for human hepatocellular carcinoma. PloS One. 2014 Jun 6;9(6):e97739.

41. Miao R, Luo H, Zhou H, Li G, Bu D, Yang X, et al. Identification of prognostic biomarkers in hepatitis B virus-related hepatocellular carcinoma and stratification by integrative multi-omics analysis. Journal of Hepatology. 2014 Oct 1;61(4):840-9.

42. Liu X, Liao W, Yuan Q, Ou Y, Huang J. TTK activates Akt and promotes proliferation and migration of hepatocellular carcinoma cells. Oncotarget. 2015 Oct 27;6(33):34309.

43. Shiraishi T, Terada N, Zeng Y, Suyama T, Luo J, Trock B, et al. Cancer/testis antigens as potential predictors of biochemical recurrence of prostate cancer following radical prostatectomy. Journal of Translational Medicine. 2011 Dec;9(1):1-9.

44. Dahlman KB, Parker JS, Shamu T, Hieronymus H, Chapinski C, Carver B, et al. Modulators of prostate cancer cell proliferation and viability identified by short-hairpin RNA library screening. PLoS One. 2012 Apr 11;7(4):e34414.

45. Liu J, Cheng X, Zhang Y, Li S, Cui H, Zhang L, et al. Phosphorylation of Mps1 by BRAF V600E prevents Mps1 degradation and contributes to chromosome instability in melanoma. Oncogene. 2013 Feb;32(6):713-23.

46. Zhang L, Shi R, He C, Cheng C, Song B, Cui H, et al. Oncogenic B-RafV600E abrogates the AKT/B-Raf/Mps1 interaction in melanoma cells. Cancer Letters. 2013 Aug 28;337(1):125-32.

47. Suda T, Tsunoda T, Daigo Y, Nakamura Y, Tahara H. Identification of human leukocyte antigen-A24-restricted epitope peptides derived from gene products upregulated in lung and esophageal cancers as novel targets for immunotherapy. Cancer Science. 2007 Nov;98(11):1803- 8.

48. Suzuki H, Fukuhara M, Yamaura T, Mutoh S, Okabe N, Yaginuma H, Hasegawa T, Yonechi A, Osugi J, Hoshino M, Kimura T. Multiple therapeutic peptide vaccines consisting of combined novel cancer testis antigens and anti-angiogenic peptides for patients with non-small cell lung cancer. Journal of Translational Medicine. 2013 Dec;11(1):1-0.

49. Mizukami Y, Kono K, Daigo Y, Takano A, Tsunoda T, Kawaguchi Y, et al. Detection of novel cancer-testis antigen-specific T-cell responses in TIL, regional lymph nodes, and PBL in patients with esophageal squamous cell carcinoma. Cancer Science. 2008 Jul;99(7):1448-54.

50. Kono K, Iinuma H, Akutsu Y, Tanaka H, Hayashi N, Uchikado Y, et al. Multicenter, phase II clinical trial of cancer vaccination for advanced esophageal cancer with three peptides derived from novel cancer-testis antigens. Journal of Translational Medicine. 2012 Dec;10(1):1-9.

51. Aruga A, Takeshita N, Kotera Y, Okuyama R, Matsushita N, Ohta T, et al. Long-term vaccination with multiple peptides derived from cancer-testis antigens can maintain a specific T-cell response and achieve disease stability in advanced biliary tract cancer. Clinical Cancer Research. 2013 Apr 15;19(8):2224-31.

52. Wang S, Zhang M, Liang D, Sun W, Zhang C, Jiang M, et al. Molecular design and anticancer activities of smallmolecule monopolar spindle 1 inhibitors: A Medicinal chemistry perspective. European Journal of Medicinal Chemistry. 2019 Aug 1;175:247-68.

53. Schulze VK, Klar U, Kosemund D, Wengner AM, Siemeister G, Stockigt D, et al. Treating cancer by spindle assembly checkpoint abrogation: Discovery of two clinical candidates, BAY 1161909 and BAY 1217389, targeting MPS1 kinase. Journal of Medicinal Chemistry. 2020 Apr 27;63(15):8025-42.

54. Uitdehaag JC, de Man J, Willemsen-Seegers N, Prinsen MB, Libouban MA, Sterrenburg JG, et al. Target residence time-guided optimization on TTK kinase results in inhibitors with potent anti-proliferative activity. Journal of Molecular Biology. 2017 Jul 7;429(14):2211-30.

55. Anderhub SJ, Mak GW, Gurden MD, Faisal A, Drosopoulos K, Walsh K, et al. High proliferation rate and a compromised spindle assembly checkpoint confers sensitivity to the MPS1 inhibitor BOS172722 in triplenegative breast cancers. Molecular Cancer Therapeutics. 2019 Oct 1;18(10):1696-707.

56. Woodward HL, Innocenti P, Cheung KM, Hayes A, Roberts J, Henley AT, et al. Introduction of a Methyl Group Curbs Metabolism of Pyrido [3, 4-d] pyrimidine Monopolar Spindle 1 (MPS1) Inhibitors and Enables the Discovery of the Phase 1 Clinical Candidate N 2-(2-Ethoxy- 4-(4-methyl-4 H-1, 2, 4-triazol-3-yl) phenyl)-6-methyl-N 8-neopentylpyrido [3, 4-d] pyrimidine-2, 8-diamine (BOS172722). Journal of Medicinal Chemistry. 2018 Sep 10;61(18):8226-40.

57. Liu Y, Laufer R, Patel NK, Ng G, Sampson PB, Li SW, et al. Discovery of pyrazolo [1, 5-a] pyrimidine TTK inhibitors: CFI-402257 is a potent, selective, bioavailable anticancer agent. ACS Medicinal Chemistry Letters. 2016 Jul 14;7(7):671-5.

58. Mason JM, Wei X, Fletcher GC, Kiarash R, Brokx R, Hodgson R, et al. Functional characterization of CFI- 402257, a potent and selective Mps1/TTK kinase inhibitor, for the treatment of cancer. Proceedings of the National Academy of Sciences. 2017 Mar 21;114(12):3127-32.

59. Szymiczek A, Carbone M, Pastorino S, Napolitano A, Tanji M, Minaai M, et al. Inhibition of the spindle assembly checkpoint kinase Mps-1 as a novel therapeutic strategy in malignant mesothelioma. Oncogene. 2017 Nov;36(46):6501-7.

60. Zheng L, Chen Z, Kawakami M, Chen Y, Roszik J, Mustachio LM, et al. Tyrosine threonine kinase inhibition eliminates lung cancers by augmenting apoptosis and polyploidy. Molecular Cancer Therapeutics. 2019 Oct 1;18(10):1775-86.

61. Gurden MD, Westwood IM, Faisal A, Naud S, Cheung KM, McAndrew C, et al. Naturally occurring mutations in the MPS1 gene predispose cells to kinase inhibitor drug resistance. Cancer Research. 2015 Aug 15;75(16):3340-54.

62. Martinez R, Blasina A, Hallin JF, Hu W, Rymer I, Fan J, Hoffman RL, Murphy S, Marx M, Yanochko G, Trajkovic D. Mitotic checkpoint kinase Mps1 has a role in normal physiology which impacts clinical utility. PLoS One. 2015 Sep 23;10(9):e0138616.

63. Maachani UB, Tandle A, Shankavaram U, Kramp T, Camphausen KA. Modulation of miR-21 signaling by MPS1 in human glioblastoma. Oncotarget. 2016 Aug 16;7(33):52912.

64. Chen S, Wang Y, Ni C, Meng G, Sheng X. HLF/ miR-132/TTK axis regulates cell proliferation, metastasis and radiosensitivity of glioma cells. Biomedicine & Pharmacotherapy. 2016 Oct 1;83:898-904.

65. Chandler BC, Speers C. A novel role for TTK in homologous recombination: implications for breast cancer radiosensitivity. Molecular & Cellular Oncology. 2020 May 3;7(3):1737771.

Author Information X