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 (2.4 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

Covalent inhibitor targets KRasG12C: A new paradigm for drugging the undruggable and challenges ahead

Hui-yu Lia,b,1Wei-liang Qia,b,c,1Yu-xiang Wanga,( )Ling-hua Menga,b( )
Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
University of Chinese Academy of Sciences, Beijing 100049, China
College of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, China

1 These authors contributed equally to this work.

Peer review under responsibility of Chongqing Medical University.

Show Author Information

Abstract

KRAS is one of the most commonly mutated oncogenes in cancers and therapeutics directly targeting the KRas have been challenging. Among the different known mutants, KRasG12C has been proved to be successfully targeted recently. Several covalent inhibitors selectively targeting KRasG12C have shown promising efficacy against cancers harboring KRASG12C mutation in clinical trials and AMG510 (sotorasib) has been approved for the treatment of KRASG12C-mutated locally advanced or metastatic non-small cell lung cancer. However, the overall responsive rate of KRasG12C inhibitors was around 50% in patients with non-small cell lung cancer and the efficacy in patients with colorectal cancer or appendiceal cancer appears to be less desirable. It is of great importance to discover biomarkers to distinguish patients who are likely benefitted. Moreover, adaptive resistance would occur inevitably with the persistent administration like other molecularly targeted therapies. Several combinatorial regimens have been studied in an effort to potentiate the efficacy of KRasG12C inhibitors in preclinical settings. This review summarized the recent progress of covalent KRasG12C inhibitors with a focus on identifying biomarkers to predict or monitor the efficacy and proposing rational drug combinations based on elucidation of the mechanisms of drug resistance.

References

1

Colicelli J. Human RAS superfamily proteins and related GTPases. Sci STKE. 2004;2004(250):RE13.

2

Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer. 2003;3(6):459-465.

3

Khan I, Rhett JM, O’Bryan JP. Therapeutic targeting of RAS: new hope for drugging the "undruggable". Biochim Biophys Acta Mol Cell Res. 2020;1867(2):118570.

4

Liu P, Wang Y, Li X. Targeting the untargetable KRAS in cancer therapy. Acta Pharm Sin B. 2019;9(5):871-879.

5

Bos JL, Rehmann H, Wittinghofer A. GEFs and GAPs: critical elements in the control of small G proteins. Cell. 2007;129(5):865-877.

6

Nagasaka M, Li Y, Sukari A, et al. KRAS G12C Game of Thrones, which direct KRAS inhibitor will claim the iron throne? Cancer Treat Rev. 2020;84: 101974.

7

Banno K, Yanokura M, Iida M, et al. Carcinogenic mechanisms of endometrial cancer: involvement of genetics and epigenetics. J Obstet Gynaecol Res. 2014;40(8):1957-1967.

8

Lu S, Jang H, Gu S, et al. Drugging Ras GTPase: a comprehensive mechanistic and signaling structural view. Chem Soc Rev. 2016;45(18):4929-4952.

9

Koera K, Nakamura K, Nakao K, et al. K-Ras is essential for the development of the mouse embryo. Oncogene. 1997;15(10):1151-1159.

10

Nakamura K, Ichise H, Nakao K, et al. Partial functional overlap of the three ras genes in mouse embryonic development. Oncogene. 2008;27(21):2961-2968.

11

Potenza N, Vecchione C, Notte A, et al. Replacement of K-Ras with H-Ras supports normal embryonic development despite inducing cardiovascular pathology in adult mice. EMBO Rep. 2005;6(5):432-437.

12

Wilson CY, Tolias P. Recent advances in cancer drug discovery targeting RAS. Drug Discov Today. 2016;21(12):1915-1919.

13

Biankin AV, Waddell N, Kassahn KS, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491(7424):399-405.

14

Neumann J, Zeindl-Eberhart E, Kirchner T, et al. Frequency and type of KRAS mutations in routine diagnostic analysis of metastatic colorectal cancer. Pathol Res Pract. 2009;205(12):858-862.

15

Lanman BA, Allen JR, Allen JG, et al. Discovery of a covalent inhibitor of KRAS G12C (AMG 510) for the treatment of solid tumors. J Med Chem. 2019;63(1):52-65.

16

Rodenhuis S. Ras and human tumors. Semin Cancer Biol. 1992; 3(4):241-247.

17

Croce CM. Oncogenes and cancer. N Engl J Med. 2008;358(5):502-511.

18

Khosravi-Far R, Der CJ. The Ras signal transduction pathway. Cancer Metastasis Rev. 1994;13(1):67-89.

19

Hansen R, Peters U, Babbar A, et al. The reactivity-driven biochemical mechanism of covalent KRAS G12C inhibitors. Nat Struct Mol Biol. 2018;25(6):454-462.

20

Balmain A, Pragnell IB. Mouse skin carcinomas induced in vivo by chemical carcinogens have a transforming Harvey-ras oncogene. Nature. 1983;303(5912):72-74.

21

Sukumar S, Notario V, Martin-Zanca D, et al. Induction of mammary carcinomas in rats by nitroso-methylurea involves malignant activation of H-ras-1 locus by single point mutations. Nature. 1983;306(5944):658-661.

22

Guerrero I, Calzada P, Mayer A, et al. A molecular approach to leukemogenesis: mouse lymphomas contain an activated c-ras oncogene. Proc Natl Acad Sci U S A. 1984;81(1):202-205.

23

Lim KH, Counter CM. Reduction in the requirement of oncogenic Ras signaling to activation of PI3K/AKT pathway during tumor maintenance. Cancer Cell. 2005;8(5):381-392.

24

Brummelkamp TR, Bernards R, Agami R. Stable suppression of tumorigenicity by virus-mediated RNA interference. Cancer Cell. 2002;2(3):243-247.

25

Ji H, Ramsey MR, Hayes DN, et al. LKB1 modulates lung cancer differentiation and metastasis. Nature. 2007; 448(7155):807-810.

26

Hingorani SR, Wang L, Multani AS, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7(5):469-483.

27

Haigis KM, Kendall KR, Wang Y, et al. Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat Genet. 2008;40(5):600-608.

28
Chen WC, To MD, Westcott PMK, et al. Regulation of KRAS4A/B splicing in cancer stem cells by the RBM39 splicing complex. bioRxiv. 2019; Preprint at https://doi.org/10.1101/646125.
29

Ryan MB, Corcoran RB. Therapeutic strategies to target RAS-mutant cancers. Nat Rev Clin Oncol. 2018;15(11):709-720.

30

van Maldegem F, Downward J. Mutant KRAS at the heart of tumor immune evasion. Immunity. 2020;52(1):14-16.

31

Cox AD, Fesik SW, Kimmelman AC, et al. Drugging the undruggable RAS: mission possible? Nat Rev Drug Discov. 2014; 13(11):828-851.

32

Gibbs JB, Graham SL, Hartman GD, et al. Farnesyltransferase inhibitors versus Ras inhibitors. Curr Opin Chem Biol. 1997; 1(2):197-203.

33

Sanclemente M, Francoz S, Esteban-Burgos L, et al. c-RAF ablation induces regression of advanced Kras/Trp53 mutant lung adenocarcinomas by a mechanism independent of MAPK signaling. Cancer Cell. 2018;33(2):217-228.

34

Jones GG, Del Río IB, Sari S, et al. SHOC2 phosphatase-dependent RAF dimerization mediates resistance to MEK inhibition in RAS-mutant cancers. Nat Commun. 2019;10(1):2532.

35

Blasco MT, Navas C, Martín-Serrano G, et al. Complete regression of advanced pancreatic ductal adenocarcinomas upon combined inhibition of EGFR and C-RAF. Cancer Cell. 2019;35(4):573-587.

36

Luo J, Emanuele MJ, Li D, et al. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell. 2009;137(5):835-848.

37

te Heesen H, Gerwert K, Schlitter J. Role of the arginine finger in Ras.RasGAP revealed by QM/MM calculations. FEBS Lett. 2007;581(29):5677-5684.

38

Hatakeyama K, Harada T, Kagamiyama H. IMP dehydrogenase inhibitors reduce intracellular tetrahydrobiopterin levels through reduction of intracellular GTP levels. Indications of the regulation of GTP cyclohydrolase I activity by restriction of GTP availability in the cells. J Biol Chem. 1992;267(29):20734-20739.

39

Nickerson S, Joy ST, Arora PS, et al. An orthosteric inhibitor of the RAS-SOS interaction. Enzymes. 2013;34(Pt. B):25-39.

40

Shima F, Yoshikawa Y, Ye M, et al. In silico discovery of small-molecule Ras inhibitors that display antitumor activity by blocking the Ras-effector interaction. Proc Natl Acad Sci U S A. 2013;110(20):8182-8187.

41

Patricelli MP, Janes MR, Li LS, et al. Selective inhibition of oncogenic KRAS output with small molecules targeting the inactive state. Cancer Discov. 2016;6(3):316-329.

42

Canon J, Rex K, Saiki AY, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019; 575(7781):217-223.

43

Hallin J, Engstrom LD, Hargis L, 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(1):54-71.

44

Misale S, Fatherree JP, Cortez E, et al. KRAS G12C NSCLC models are sensitive to direct targeting of KRAS in combination with PI3K inhibition. Clin Cancer Res. 2019;25(2):796-807.

45

Ni D, Li X, He X, et al. Drugging K-Ras(G12C) through covalent inhibitors: mission possible? Pharmacol Ther. 2019;202: 1-17.

46

Molina-Arcas M, Moore C, Rana S, et al. Development of combination therapies to maximize the impact of KRAS-G12C inhibitors in lung cancer. Sci Transl Med. 2019;11(510):eaaw7999.

47

Govindan R, Fakih MG, Price TJ, et al. Phase 1 trial evaluating safety, efficacy, and PK of AMG 510, a novel KRASG12C inhibitor, in non-small cell lung cancer. J Thorac Oncol. 2019;14(11):S1191-S1192.

48
Janne PA, Papadopoulous K, Ou SI, et al. A Phase 1 clinical trial evaluating the pharmacokinetics (PK), safety, and clinical activity of MRTX849, a mutant-selective small molecule KRAS G12C inhibitor, in advanced solid tumors. International Conference on Molecular Targets and Cancer Therapeutics. 2019;October 26-30.
49

Hata AN, Shaw AT. Resistance looms for KRAS(G12C) inhibitors. Nat Med. 2020;26(2):169-170.

50

Awad MM, Liu S, Rybkin Ⅱ, et al. Acquired resistance to KRAS(G12C) inhibition in cancer. N Engl J Med. 2021;384(25):2382-2393.

51

Downward J. RAS synthetic lethal screens revisited: still seeking the elusive prize? Clin Cancer Res. 2015;21(8):1802-1809.

52

Ostrem JM, Peters U, Sos ML, et al. K-Ras (G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503(7477):548-551.

53
Newswire P. Amgen Announces New Clinical Data Evaluating Novel Investigational KRAS(G12C) Inhibitor in Larger Patient Group at WCLC 2019; 2019. https://www.amgen.com/.
54

Lito P, Solomon M, Li LS, et al. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science. 2016;351(6273):604-608.

55

Hobbs GA, Wittinghofer A, Der CJ. Selective targeting of the KRAS G12C mutant: kicking KRAS when it’s down. Cancer Cell. 2016;29(3):251-253.

56

Janes MR, Zhang J, Li LS, et al. Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell. 2018;172(3):578-589.

57

Jiao D, Yang S. Overcoming resistance to drugs targeting KRAS(G12C) mutation. Innovation. 2020;1(2):100035.

58

Rex K, Saiki AY, Sun JR, et al. In vivo characterization of AMG 510-a potent and selective KRASG12C covalent small molecule inhibitor in preclinical KRASG12C cancer models. Cancer Res. 2019;79(13 Supplement):3090.

59

Hong DS, Fakih MG, Strickler JH, et al. KRAS(G12C) inhibition with sotorasib in advanced solid tumors. N Engl J Med. 2020;383(13):1207-1217.

60
FDA Approves LUMAKRASTM (Sotorasib), the First and Only Targeted Treatment for Patients with KRAS G12C-Mutated Locally Advanced or Metastatic Non-small Cell Lung Cancer; 2021. https://www.amgen.com/newsroom/press-releases/2021/05/fda-approves-lumakras-sotorasib-the-first-and-onlytargeted-treatment-for-patients-with-kras-g12cmutatedlocally-advanced-or-metastatic-nonsmall-cell-lung-cancer.
61

Christensen JG, Olson P, Briere T, et al. Targeting Kras(g12c)-mutant cancer with a mutation-specific inhibitor. J Intern Med. 2020;288(2):183-191.

62

Jänne PA, Rybkin Ⅱ, Spira AI, et al. Activity and safety of adagrasib (MRTX849) in advanced/metastatic non-small-cell lung cancer (NSCLC) harboring KRAS G12C mutation. Eur J Canc. 2020;138(S2):S1-S2.

63

Johnson ML, Ou SHI, Barve M, et al. Activity and safety of adagrasib (MRTX849) in patients with colorectal cancer (CRC) and other solid tumors harboring a KRAS G12C mutation. Eur J Canc. 2020;138(S2):S2.

64
Mirati Therapeutics Reports Investigational Adagrasib (MRTX849) Preliminary Data Demonstrating Tolerability and Durable Anti-tumor Activity as Well as Initial MRTX1133 Preclinical Data; 2020. https://ir.mirati.com/news-releases/news-details/2020/Mirati-Therapeutics-Reports-Investigational-Adagrasib-MRTX849-Preliminary-Data-Demonstrating-Tolerability-and-Durable-Anti-Tumor-Activity-as-wellas-Initial-MRTX1133-Preclinical-Data/default.aspx.
65

McCormick F. Progress in targeting RAS with small molecule drugs. Biochem J. 2019;476(2):365-374.

66

Klempner SJ, Hata AN. Can the help match the hype? KRAS(G12C)-specific inhibitors and beyond. Cancer Discov. 2020;10(1):20-22.

67

Amodio V, Yaeger R, Arcella P, et al. EGFR blockade reverts resistance to KRAS(G12C) inhibition in colorectal cancer. Cancer Discov. 2020;10(8):1129-1139.

68

Ryan MB, Fece de la Cruz F, Phat S, et al. Vertical pathway inhibition overcomes adaptive feedback resistance to KRAS(G12C) inhibition. Clin Canc Res. 2020;26(7):1633-1643.

69

Adachi Y, Ito K, Hayashi Y, et al. Epithelial-to-mesenchymal transition is a cause of both intrinsic and acquired resistance to KRAS G12C inhibitor in KRAS G12C mutant non-small cell lung cancer. Clin Cancer Res. 2020;26(22):5962-5973.

70
Results from Phase 2 CodeBreaK 100 Show LUMAKRASTM (Sotorasib) Is the First and Only KRAS G12C Inhibitor with Overall Survival Data; 2021. https://www.amgen.com/newsroom/press-releases/2021/06/results-from-phase-2-codebreak-100-show-lumakras-sotorasib-is-the-first-and-onlykras-g12c-inhibitor-with-overall-survival-data.
71

Kelsey I, Manning BD. mTORC1 status dictates tumor response to targeted therapeutics. Sci Signal. 2013;6(294):pe31.

72

Bunda S, Burrell K, Heir P, et al. Inhibition of SHP2-mediated dephosphorylation of Ras suppresses oncogenesis. Nat Commun. 2015;6:e8859.

73

Fedele C, Li S, Teng KW, et al. SHP2 inhibition diminishes KRASG12C cycling and promotes tumor microenvironment remodeling. J Exp Med. 2021;218(1):e20201414.

74

Santarpia L, Lippman SM, El-Naggar AK. Targeting the MAPK-RAS-RAF signaling pathway in cancer therapy. Expert Opin Ther Targets. 2012;16(1):103-119.

75

Seger R, Krebs EG. The MAPK signaling cascade. Faseb J. 1995; 9(9):726-735.

76

Costa-Cabral S, Brough R, Konde A, et al. CDK1 is a synthetic lethal target for KRAS mutant tumours. PloS One. 2016;11(2):e0149099.

77

Puyol M, Martin A, Dubus P, et al. A synthetic lethal interaction between K-Ras oncogenes and Cdk4 unveils a therapeutic strategy for non-small cell lung carcinoma. Cancer Cell. 2010; 18(1):63-73.

78

Santana-Codina N, Chandhoke AS, Yu Q, et al. Defining and targeting adaptations to oncogenic KRAS(G12C) inhibition using quantitative temporal proteomics. Cell Rep. 2020;30(13):4584-4599.

79

Xue JY, Zhao Y, Aronowitz J, et al. Rapid non-uniform adaptation to conformation-specific KRAS(G12C) inhibition. Nature. 2020;577(7790):421-425.

80

Bishop L, Chen Z, Omar O, et al. Targeting Aurka effectively suppresses Kras-mutant tumorigenesis through regulating P70S6K phosphorylaton in gastrointestinal cancers. Gastroenterology. 2017;152(5):S41.

81

Tanaka N, Lin JJ, Li C, et al. Clinical acquired resistance to KRAS(G12C) inhibition through a novel KRAS switch-Ⅱ pocket mutation and polyclonal alterations converging on RAS-MAPK reactivation. Cancer Discov. 2021;11(8):1913-1922.

82

Welsch ME, Kaplan A, Chambers JM, et al. Multivalent small-molecule pan-RAS inhibitors. Cell. 2017;168(5):878-889.

83

Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15(3):178-196.

84

Fischer KR, Durrans A, Lee S, et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527(7579):472-476.

85

Chen N, Fang W, Lin Z, et al. KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunol Immunother. 2017;66(9):1175-1187.

86

Weijzen S, Velders MP, Kast WM. Modulation of the immune response and tumor growth by activated Ras. Leukemia. 1999; 13(4):502-513.

87

Goebel L, Müller MP, Goody RS, et al. KRasG12C inhibitors in clinical trials: a short historical perspective. RSC Med Chem. 2020;11(7):760-770.

Genes & Diseases
Pages 403-414
Cite this article:
Li H-y, Qi W-l, Wang Y-x, et al. Covalent inhibitor targets KRasG12C: A new paradigm for drugging the undruggable and challenges ahead. Genes & Diseases, 2023, 10(2): 403-414. https://doi.org/10.1016/j.gendis.2021.08.011

285

Views

5

Downloads

7

Crossref

6

Web of Science

6

Scopus

0

CSCD

Altmetrics

Received: 06 July 2021
Revised: 12 August 2021
Accepted: 27 August 2021
Published: 28 September 2021
© 2021 The Authors.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return