AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1.8 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Efficacy of rigosertib, a small molecular RAS signaling disrupter for the treatment of KRAS-mutant colorectal cancer

Xinyi Zhou1,*Qian Xiao1,* ( )Dongliang Fu1Haochen Zhang2Yang Tang1Jinjie He1Yeting Hu1Xiangxing Kong1Fei Teng3Xiangrui Liu4Ying Yuan2Kefeng Ding1,5 ( )
Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
Hangzhou Oncocare Co Ltd, Hangzhou 310009, China
Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
Cancer Center, Zhejiang University, Hangzhou 310009, China

*These authors contributed equally to this work.

Show Author Information

Abstract

Objective

Mutant KRAS, the principal isoform of RAS, plays a pivotal role in the oncogenesis of colorectal cancer by constitutively activating the RAF/MEK/ERK and PI3K/AKT pathways. Effective targeted therapies are urgently needed. We investigated whether rigosertib, a benzyl styryl sulfone RAS signaling disruptor, could selectively kill KRAS-mutant colorectal cancer cells.

Methods

CCK-8 was used to determine the cell viability. Patient-derived tumor and cancer cell xenograft models were used to detect the inhibitory efficacy of rigosertib. Flow cytometry was used to evaluate the apoptosis and cell cycle progression. Apoptosis and cell cycle arrest markers were detected by Western blot. DCFH-DA was used to determine the reactive oxygen species. Immunohistochemistry staining and Western blot were performed to characterize RAS signaling markers in colorectal cancer tissues and cells.

Results

Rigosertib (RGS) exhibited a cytotoxic effect against colorectal cancer cells, which was greater in KRAS-mutant cells. Furthermore, RGS induced mitotic arrest and oxidative stress-dependent apoptosis in KRAS-mutant DLD1 and HCT116 cells. Besides, RGS disrupted RAS signaling, and the inhibition of RAS/MEK/ERK was independent of cellular oxidative stress. Using patient-derived xenograft models, the response and tumor inhibition of RGS were significantly higher in the KRAS-mutant subgroup, while p-MEK, p-ERK, and p-AKT levels of RGS-treated tumors were significantly decreased. Finally, in a KRAS-mutant, chemotherapy-resistant patient-derived xenograft model, RGS showed a stronger therapeutic effect than the combination standard therapy involving fluoropyrimidine + oxaliplatin/irinotecan + bevacizumab.

Conclusions

These data showed that targeting RAS signaling using RGS could be a therapeutic treatment for KRAS-mutant colorectal cancer patients.

Electronic Supplementary Material

Download File(s)
cbm-19-2-213_ESM.pdf (1.6 MB)

References

1

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016; 66: 115-32.

2

Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, et al. Colorectal cancer statistics, 2017. CA Cancer J Clin. 2017; 67: 177-93.

3

Van Cutsem E, Peeters M, Siena S, Humblet Y, Hendlisz A, Neyns B, et al. Open-label phase Ⅲ trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. J Clin Oncol. 2007; 25: 1658-64.

4

Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS oncogenes: weaving a tumorigenic web. Nat Rev Cancer. 2011; 11: 761-74.

5

Simanshu DK, Nissley DV, McCormick F. RAS proteins and their regulators in human disease. Cell. 2017; 170: 17-33.

6

Matikas A, Mistriotis D, Georgoulias V, Kotsakis A. Targeting KRAS mutated non-small cell lung cancer: a history of failures and a future of hope for a diverse entity. Crit Rev Oncol Hematol. 2017; 110: 1-12.

7

Russo M, Di Nicolantonio F, Bardelli A. Climbing RAS, the everest of oncogenes. Cancer Discov. 2014; 4: 19-21.

8

Reddy MV, Venkatapuram P, Mallireddigari MR, Pallela VR, Cosenza SC, Robell KA, et al. Discovery of a clinical stage multi-kinase inhibitor sodium (E)-2-{2-methoxy-5-[(2′,4′,6′-trimethoxystyrylsulfonyl)methyl]phenylamino}acetate (ON 01910. Na): synthesis, structure-activity relationship, and biological activity. J Med Chem. 2011; 54: 6254-76.

9

Dai YH, Hung LY, Chen RY, Lai CH, Chang KC. ON 01910.Na inhibits growth of diffuse large B-cell lymphoma by cytoplasmic sequestration of sumoylated C-MYB/TRAF6 complex. Transl Res. 2016; 175: 129-43.

10

Oussenko IA, Holland JF, Reddy EP, Ohnuma T. Effect of ON 01910.Na, an anticancer mitotic inhibitor, on cell-cycle progression correlates with RanGAP1 hyperphosphorylation. Cancer Res. 2011; 71: 4968-76.

11

Athuluri-Divakar SK, Vasquez-Del Carpio R, Dutta K, Baker SJ, Cosenza SC, Basu I, et al. A small molecule RAS-mimetic disrupts RAS association with effector proteins to block signaling. Cell. 2016; 165: 643-55.

12

Ritt DA, Abreu-Blanco MT, Bindu L, Durrant DE, Zhou M, Specht SI, et al. Inhibition of Ras/Raf/MEK/ERK pathway signaling by a stress-induced phospho-regulatory circuit. Mol Cell. 2016; 64: 875-87.

13

Foster SA, Malek S. The RAS/MAPK Axis Gets Stressed Out. Mol Cell. 2016; 64: 854-5.

14

Boutin AT, Liao WT, Wang M, Hwang SS, Karpinets TV, Cheung H, et al. Oncogenic Kras drives invasion and maintains metastases in colorectal cancer. Genes Dev. 2017; 31: 370-82.

15

Vartanian S, Bentley C, Brauer MJ, Li L, Shirasawa S, Sasazuki T, et al. Identification of mutant K-Ras-dependent phenotypes using a panel of isogenic cell lines. J Biol Chem. 2013; 288: 2403-13.

16

Shirasawa S, Furuse M, Yokoyama N, Sasazuki T. Altered growth of human colon cancer cell lines disrupted at activated Ki-ras. Science. 1993; 260: 85-8.

17

Huang D, Sun W, Zhou Y, Li P, Chen F, Chen H, et al. Mutations of key driver genes in colorectal cancer progression and metastasis. Cancer Metastasis Rev. 2018; 37: 173-87.

18

Liu Y, He J, Xu J, Li J, Jiao Y, Bei D, et al. Neuroendocrine differentiation is predictive of poor survival in patients with stage Ⅱ colorectal cancer. Oncol Lett. 2017; 13: 2230-6.

19

Remmele W, Stegner HE. Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue. Pathologe. 1987; 8: 138-40.

20

Fulda S, Debatin KM. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene. 2006; 25: 4798-811.

21

Hyoda T, Tsujioka T, Nakahara T, Suemori S, Okamoto S, Kataoka M, et al. Rigosertib induces cell death of a myelodysplastic syndrome-derived cell line by DNA damage-induced G2/M arrest. Cancer Sci. 2015; 106: 287-93.

22

Amodio V, Yaeger R, Arcella P, Cancelliere C, Lamba S, Lorenzato A, et al. EGFR blockade reverts resistance to KRAS(G12C) inhibition in colorectal cancer. Cancer Discov. 2020; 10: 1129-39.

23

Chapman CM, Sun X, Roschewski M, Aue G, Farooqui M, Stennett L, et al. ON 01910.Na is selectively cytotoxic for chronic lymphocytic leukemia cells through a dual mechanism of action involving PI3K/AKT inhibition and induction of oxidative stress. Clin Cancer Res. 2012; 18: 1979-91.

24

Montor WR, Salas A, Melo FHM. Receptor tyrosine kinases and downstream pathways as druggable targets for cancer treatment: the current arsenal of inhibitors. Mol Cancer. 2018; 17: 55.

25

Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019; 575: 217-23.

26

Hallin J, Engstrom LD, Hargis L, Calinisan A, Aranda R, Briere DM, et al. The KRAS(G12C) inhibitor MRTX849 provides insight toward therapeutic susceptibility of KRAS-mutant cancers in mouse models and patients. Cancer Discov. 2020; 10: 54-71.

27

Yang Q, Huo S, Sui Y, Du Z, Zhao H, Liu Y, et al. Mutation status and immunohistochemical correlation of KRAS, NRAS, and BRAF in 260 Chinese colorectal and gastric cancers. Front Oncol. 2018; 8: 487.

28

Holkova B, Zingone A, Kmieciak M, Bose P, Badros AZ, Voorhees PM, et al. A phase Ⅱ trial of AZD6244 (selumetinib, ARRY-142886), an oral MEK1/2 inhibitor, in relapsed/refractory multiple myeloma. Clin Cancer Res. 2016; 22: 1067-75.

29

Ganesan P, Janku F, Naing A, Hong DS, Tsimberidou AM, Falchook GS, et al. Target-based therapeutic matching in early-phase clinical trials in patients with advanced colorectal cancer and PIK3CA mutations. Mol Cancer Ther. 2013; 12: 2857-63.

30

Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med. 2008; 14: 1351-6.

31

Schram AM, Gandhi L, Mita MM, Damstrup L, Campana F, Hidalgo M, et al. A phase Ib dose-escalation and expansion study of the oral MEK inhibitor pimasertib and PI3K/MTOR inhibitor voxtalisib in patients with advanced solid tumours. Br J Cancer. 2018; 119: 1471-6.

32

Gumireddy K, Reddy MV, Cosenza SC, Boominathan R, Baker SJ, Papathi N, et al. ON01910, a non-ATP-competitive small molecule inhibitor of Plk1, is a potent anticancer agent. Cancer Cell. 2005; 7: 275-86.

33

Steegmaier M, Hoffmann M, Baum A, Lénárt P, Petronczki M, Krssák M, et al. BI 2536, a potent and selective inhibitor of pololike kinase 1, inhibits tumor growth in vivo. Curr Biol. 2007; 17: 316-22.

34

Prasad A, Park IW, Allen H, Zhang X, Reddy MV, Boominathan R, et al. Styryl sulfonyl compounds inhibit translation of cyclin D1 in mantle cell lymphoma cells. Oncogene. 2009; 28: 1518-28.

35

Jiang CC, Lai F, Thorne RF, Yang F, Liu H, Hersey P, et al. MEKindependent survival of B-RAFV600E melanoma cells selected for resistance to apoptosis induced by the RAF inhibitor PLX4720. Clin Cancer Res. 2011; 17: 721-30.

36

Liu Z, Wang M, Wang H, Fang L, Gou S. Platinum-based modification of styrylbenzylsulfones as multifunctional antitumor agents: targeting the RAS/RAF pathway, enhancing antitumor activity, and overcoming multidrug resistance. J Med Chem. 2020; 63: 186-204.

37

Jost M, Chen Y, Gilbert LA, Horlbeck MA, Krenning L, Menchon G, et al. Combined CRISPRi/a-based chemical genetic screens reveal that rigosertib is a microtubule-destabilizing agent. Mol Cell. 2017; 68: 210-223.e6.

Cancer Biology & Medicine
Pages 213-228
Cite this article:
Zhou X, Xiao Q, Fu D, et al. Efficacy of rigosertib, a small molecular RAS signaling disrupter for the treatment of KRAS-mutant colorectal cancer. Cancer Biology & Medicine, 2022, 19(2): 213-228. https://doi.org/10.20892/j.issn.2095-3941.2020.0532

48

Views

0

Downloads

2

Crossref

9

Web of Science

7

Scopus

Altmetrics

Received: 07 September 2020
Accepted: 13 January 2021
Published: 29 March 2022
©2022 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return