リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Novel Oral Derivative UD-017, a Highly Selective CDK7 Inhibitor, Exhibits Anticancer Activity by Inducing Cell- Cycle Arrest and Apoptosis in Human Colorectal Cancer」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Novel Oral Derivative UD-017, a Highly Selective CDK7 Inhibitor, Exhibits Anticancer Activity by Inducing Cell- Cycle Arrest and Apoptosis in Human Colorectal Cancer

阿賀 康弘 広島大学

2020.02.27

概要

Cyclin-dependent kinases (CDK) are serine/threonine kinases that regulate multiple cellular
processes19). CDK7 was identified as a component of CDK-activating kinase (CAK), which plays a
pivotal role in cell cycle progression10). CAK phosphorylates threonine residues in the activation
segment (T-loop) of downstream CDKs, including cdc-2/CDK1, CDK2, CDK4, and CDK6, and
modulates their activities25). CDK7 plays another important role in transcription via phosphorylation
of the carboxyl-terminal domain (CTD) of RNA polymerase II (RNAPII)1,5,11,16). Specifically, the
general transcription factor II (TFIIH) complex (containing CDK7 and cyclin H) phosphorylates
serine-5 of the CTD, thereby enhancing the association between the CTD and m7G RNA capping
machinery1). TFIIH also phosphorylates the serine-7 residue of the CTD and modulates pausing and
transcriptional termination5,11).
The super-enhancer, first proposed by Young et al.12) is a huge cluster of enhancers and formed by
transcription factors and co-factors. The expression of several oncogenes, including Myc, is regulated
by super-enhancers18,20). The inhibition of CDK7 downregulates super-enhancer-mediated gene
expression in cancer cells, including the expression of Myc and other oncogenes7).
To date, several CDK inhibitors (including the pan-CDK inhibitor Flavopiridol) have been tested in
clinical trials and found to be highly toxic30). ...

この論文で使われている画像

参考文献

Akhtar, M. S., Heidemann,M., Tietjen, J. R., Zhang, D.W., Chapman, R. D., Eick, D., et al.

2009. TFIIH kinase places bivalent marks on the carboxy-terminal domain of RNA

polymerase II. Molecular Cell. 34:387–393.

Ali, S., Heathcote, D. A., Kroll, S. H., Jogalekar,A. S., Scheiper, B., Patel, H., et al. 2009.

The development of a selective cyclin-dependent kinase inhibitor that shows antitumor

activity. Cancer Research. 69:6208–6215.

Beaver, J. A., Amiri-Kordestani, L., Charlab, R., Chen, W., Palmby, T., Tilley, A., et al.

2015. FDA Approval: Palbociclib for the treatment of postmenopausal patients with

estrogen receptor-positive, HER2-negative metastatic breast cancer. Clinical Cancer

Research. 21:4760–4766.

Bhullar, K. S., Lagarón, N. O., McGowan, E. M., Parmar, I., Jha, A., Hubbard, B. P., et al.

2018. Kinase-targeted cancer therapies: progress, challenges and future directions.

Molecular Cancer. 17:48.

Boeing, S., Rigault, C., Heidemann, M., Eick, D. and Meisterernst, M. 2010. RNA

polymerase II C-terminal heptarepeat domain Ser-7 phosphorylation is established in a

mediator-dependent fashion. The Journal of biological chemistry. 285:188–196.

Chen, B. J., Wu, Y. L., Tanaka, Y. and Zhang, W. 2014. Small molecules targeting c-Myc

oncogene: promising anti-cancer therapeutics. International Journal of Biological Sciences.

10:1084–1096.

Chipumuro, E., Marco, E., Christensen, C. L., Kwiatkowski, N., Zhang T., Hatheway,C. M.,

et al. 2014. CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in

MYCN-driven cancer. Cell 159:1126–1139.

Christensen, C. L., Kwiatkowski, N., Abraham, B. J., Carretero, J., Al-Shahrour, F., Zhang,

T., et al. 2014. Targeting transcriptional addictions in small cell lung cancer with a covalent

CDK7 inhibitor. Cancer Cell. 26:909–922.

Dickler, M. N., Tolaney, S. M., Rugo, H. S., Cortés, J., Diéras, V., Patt, D., et al. 2017.

MONARCH 1, A phase II study of Abemaciclib, a CDK4 and CDK6 inhibitor, as a single

agent, in patients with refractory HR+/HER2- metastatic breast cancer. Clinical Cancer

Research. 23:5218–5224.

10 Fisher, R. P. and Morgan, D. O. 1994. A novel cyclin associates with M015/CDK7 to form

the CDK-activating kinase. Cell 78:713–724.

11 Glover-Cutter, K., Larochelle, S., Erickson, B., Zhang, C., Shokat, K., Fisher, R. P.,et al.

2009. TFIIH-associated Cdk7 kinase functions in phosphorylation of C-terminal domain

Ser7 residues, promoter-proximal pausing, and termination by RNA polymerase II.

Molecular and Cellular Biology. 29: 5455–5564.

12 Hnisz, D., Abraham, B. J., Lee, T. I., Lau, A., Saint-André, V., Sigova, A. A., et al. 2013.

Super-enhancers in the control of cell identity and disease. Cell. 155:934–947.

13 Hu, S., Marineau, J. J., Rajagopal, N., Hamman, K. B., Choi, Y. J., Schmidt, DR., et al.

2019. Discovery and Characterization of SY-1365, a Selective, Covalent Inhibitor of CDK7.

Cancer Researsh 79:3479–3491.

14 Kalan, S., Amat, R., Schachter, M.M., Kwiatkowski, N., Abraham, B.J., Liang, Y., et al.

2017. Activation of the p53 Transcriptional Program Sensitizes Cancer Cells to Cdk7

Inhibitors. Cell Report. 21:467-481.

15 Kelso, T.W., Baumgart, K., Eickhoff, J., Albert, T., Antrecht, C., Lemcke, S., et al. 2014.

Cyclin-dependent kinase 7 controls mRNA synthesis by affecting stability of preinitiation

complexes, leading to altered gene expression, cell cycle progression, and survival of tumor

cells. Molecular and Cellular Biology. 34:3675-3688.

16 Komarnitsky, P., Cho, E. J. and Buratowski, S. 2000. Different phosphorylated forms of

RNA polymerase II and associated mRNA processing factors during transcription. Genes &

Development. 14:2452–2460.

17 Kwiatkowski, N., Zhang, T., Rahl, P. B., Abraham, B. J., Reddy, J., Ficarro, S. B., et al.

2014. Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature.

511:616–620.

18 Lovén, J., Hoke, H. A., Lin, C. Y., Lau, A., Orlando, D. A., Vakoc, C.R., et al. 2013.

Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell. 153:320–

334.

19 Malumbres, M. 2014. Cyclin-dependent kinases. Genome Biology. 15:122–131.

20 Mansour, M. R., Abraham, B. J., Anders, L., Berezovskaya, A., Gutierrez, A., Durbin, A. D.,

et al. 2014. Oncogene regulation. An oncogenic super-enhancer formed through somatic

mutation of a noncoding intergenic element. Science 346:1373–1377.

21 Min, A., Kim, J.E., Kim, Y.J., Lim, J.M., Kim, S., Kim, J.W., et al. 2018. Cyclin E

overexpression confers resistance to the CDK4/6 specific inhibitor palbociclib in gastric

cancer cells. Cancer Letters. 430:123-132

22 Pandey, K., An, H.J., Kim, S.K., Lee, S.A., Kim, S., Lim, S.M., et al. 2019. Molecular

mechanisms of resistance to CDK4/6 inhibitors in breast cancer: A review. International

Journal of Cancer. 145:1179-1188.

23 Patel,H., Abduljabbar, R., Lai, C. F., Periyasamy, M., Harrod, A., Gemma, C., et al. 2016.

Expression of CDK7, cyclin H, and MAT1 is elevated in breast cancer and is prognostic in

estrogen receptor-positive breast cancer. Clinical Cancer Research. 22:5929–5938.

24 Patel, H, Periyasamy, M., Sava, G. P, Bondke, A., Slafer, B. W., Kroll, S. H. B., et al. 2018.

ICEC0942, an Orally Bioavailable Selective Inhibitor of CDK7 for Cancer Treatment.

Molecular Cancer Therapeutics. 17:1156–1166.

25 Schachter, M. M., Merrick,K. A., Larochelle, S., Hirschi, A., Zhang, C., Shokat, K. M., et al.

2013. A Cdk7-Cdk4 T-Loop Phosphorylation Cascade Promotes G1 Progression. Molecular

Cell. 50:250–260.

26 Tripathy, D., Bardia, A. and Sellers, W. R. 2017. Ribociclib (LEE011): Mechanism of action

and clinical impact of this selective cyclin-dependent kinase 4/6 inhibitor in various solid

tumors. Clinical Cancer Research. 23:3251–3262.

27 Wang, B. Y., Liu, Q. Y., Cao, J., Chen, J. W. and Liu, Z. S. 2016. Selective CDK7 inhibition

with BS-181 suppresses cell proliferation and induces cell cycle arrest and apoptosis in

gastric cancer. Drug Design, Development and Therapy. 10:1181–1189.

28 Wang, Q., Li, M., Zhang, X., Huang, H., Huang, J., Ke, J., et al. 2016. Upregulation of

CDK7 in gastric cancer cell promotes tumor cell proliferation and predicts poor prognosis.

Experimental and Molecular Pathology. 100: 514–521.

29 Wang, X. N, Su, X. X, Cheng, S. Q, Sun, Z. Y, Huang, Z. S and Ou, T. M. 2019. MYC

modulators in cancer: a patent review. Expert Opinion on Therapeutic Patents. 29:353–367.

30 Wiernik P. H. 2016. Alvocidib (flavopiridol) for the treatment of chronic lymphocytic

leukemia. Expert Opinion on Investigational Drugs. 25:729–734.

31 Zhang, J., Yang, X., Wang, Y., Shi, H., Guan, C., Yao, L., et al. 2013. Low expression of

cyclinH and cyclin-dependent kinase 7 can decrease the proliferation of human esophageal

squamous cell carcinoma. Digestive Diseases and Sciences. 58:2028–2037.

32 Zhang, Y.X., Sicinska, E., Czaplinski, J.T., Remillard, S.P., Moss, S., Wang, Y., et al. 2014.

Antiproliferative effects of CDK4/6 inhibition in CDK4-amplified human liposarcoma in

vitro and in vivo. Molecular Cancer Therapeutics. 13:2184-2193.

33 Zhang, Z., Peng, H., Wang, X., Yin, X., Ma, P., Jing, Y., et al. 2017. Preclinical Efficacy and

Molecular Mechanism of Targeting CDK7-Dependent Transcriptional Addiction in Ovarian

Cancer. Molecular Cancer Therapeutics. 16:1739–1750.

34 Zhou, Y., Lu, L., Jiang, G., Chen, Z., Li, J., An, P., et al. 2019. Targeting CDK7 increases the

stability of Snail to promote the dissemination of colorectal cancer. Cell Death &

Differentiation. 26:1442-1452.

FIGURE and TABLE LEGENDS

Figure 1. In vitro properties of UD-017 as a CDK7 inhibitor.

(A) UD-017 selectivity for various kinases. Specificity of UD-017 (1000 nM) was tested against a

panel of 313 kinases. IC50 values were calculated for kinases that showed more than 50% inhibition at

1000 nM of UD-017. (B) Selectivity of UD-017 for various CDK subtypes. (C) GI50 values of UD017 against colorectal cancer cell lines.

Figure 2. Mechanism underlying the antiproliferative effects of UD-017 in HCT-116 cells.

(A) Effect of UD-017 on the phosphorylation of CDKs and Rb protein in HCT-116 cells at 8 and 24

hrs after treatment. CDK1, pCDK1, CDK2, pCDK2: 34 kDa. Rb, pRb: 110kDa. GAPDH: 37kDa. (B)

Effect of UD-017 on cell cycle progression in HCT-116 cells at 8 hrs after treatment (n = 2). Band

densities were quantified and normalized to DMSO control.

Figure 3. Induction of apoptosis by UD-017 in HCT-116 cells.

(A) Effect of UD-017 on phosphorylation of RNAPII in HCT-116 cells at 1 to 72 hrs after treatment.

RNAPII, pRNAPII: 240 kDa. (B) Effect of UD-017 on Myc expression in HCT-116 cells at 1 to 72

hrs after treatment. c-Myc: 57-65 kDa. (C) Expression of apoptosis-related proteins at 72 hrs after

treatment with UD-017 in HCT-116 cells. XIAP: 53 kDa, PARP: 116 kDa, cleaved PARP: 89 kDa.

(D) Apoptosis induction in HCT-116 cells at 72 hrs after treatment with UD-017 (n = 3–6). Band

densities were quantified and normalized to DMSO control. Data are presented as mean ± SD.

Dunnett’s multiple test using EXSUS, ***: P < 0.001.

Figure 4. Anticancer activity of UD-017 in HCT-116 xenograft mouse model.

(A) In vivo anticancer activity of UD-017 after 14 days of treatment. (B) Body weight change after

UD-017 treatment. Data are presented as mean ± SE (n = 6). Dunnett’s multiple test using EXSUS,

*p < 0.05, **p < 0.01, ***p < 0.001 versus vehicle group.

Figure 5. Mechanism underlying anticancer activity of UD-017 in HCT-116 xenograft mouse model.

(A) Phosphorylation of Rb protein at 2 hrs after administration of a single dose of UD-017 (100

mg/kg). Rb, pRb: 110kDa. NC: not calculated. (B) Phosphorylation of RNAPII protein, expression

of c-Myc, and PARP cleavage at 2 hrs after administration of a daily single dose of UD-017 (25, 50,

100 mg/kg). RNAPII, pRNAPII: 240kDa. c-Myc: 57-65kDa. PARP: 116kDa, cleaved PARP: 89kDa.

Band densities were quantified and ratios were calculated.

Figure 6. Combination treatment of HCT-116 xenograft mouse model with UD-017 and 5-FU.

(A) In vivo anticancer activity of UD-017 with 5-FU after 14 days of treatment. (B) Body weight

change after UD-017 with 5-FU treatment. Data are presented as mean ± SE (n = 6). Dunnett’s

multiple test using EXSUS, *p < 0.05, UD-017 50 mg/kg + 5-FU 15 mg/kg group versus UD-017 50

mg/kg group.

Figure 7. Schematic of proposed mechanism of action of UD-017 in HCT-116 cells.

UD-017 suppresses phosphorylation of CDK1, 2, and 4/6, which is followed by cell cycle arrest.

UD-017 also prevents phosphorylation of RNAPII and reduces the transcription of c-Myc, which

plays an important role in cell survival. Apoptosis is induced by the activation of apoptosis-related

proteins.

Figure 1

(A)㻌

(B)㻌

(C)㻌

Figure 2

(A)

(B)

Figure 3

(A)㻌

(B)㻌

(C)㻌

(D)㻌

Figure 4

(A)㻌

(B)㻌

Figure 5

(A)㻌

(B)㻌

Figure 6

(A)㻌

(B)㻌

Figure 7

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る