Role of oncogenic KRAS in the prognosis, diagnosis and treatment of colorectal cancer

Gongmin Zhu, Lijiao Pei, Hongwei Xia, Qiulin Tang, Feng Bi, Gongmin Zhu, Lijiao Pei, Hongwei Xia, Qiulin Tang, Feng Bi

Abstract

Colorectal cancer (CRC) is a heterogeneous disease at the cellular and molecular levels. Kirsten rat sarcoma (KRAS) is a commonly mutated oncogene in CRC, with mutations in approximately 40% of all CRC cases; its mutations result in constitutive activation of the KRAS protein, which acts as a molecular switch to persistently stimulate downstream signaling pathways, including cell proliferation and survival, thereby leading to tumorigenesis. Patients whose CRC harbors KRAS mutations have a dismal prognosis. Currently, KRAS mutation testing is a routine clinical practice before treating metastatic cases, and the approaches developed to detect KRAS mutations have exhibited favorable sensitivity and accuracy. Due to the presence of KRAS mutations, this group of CRC patients requires more precise therapies. However, KRAS was historically thought to be an undruggable target until the development of KRASG12C allele-specific inhibitors. These promising inhibitors may provide novel strategies to treat KRAS-mutant CRC. Here, we provide an overview of the role of KRAS in the prognosis, diagnosis and treatment of CRC.

Keywords: Colorectal cancer; Combination therapy; G12C; KRAS; Prognosis; Targeted therapy.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
KRAS signaling pathway and relevant inhibitors of each node. After the activation of receptor tyrosine kinase, GRB2 combines with the guanine nucleotide exchange factor SOS and then interacts with KRAS protein that is attached to the cell membrane, thereby promoting KRAS activation. Intrinsic KRAS GTP-GDP cycling is regulated by GEFs and GAPs. Once KRAS is mutated, this cycle is disrupted, allowing mutant KRAS protein to accumulate in an active state and thereby persistently activating downstream MAPK and PI3K signaling cascade, resulting in cell proliferation and survival. Various KRAS inhibitors listed in the box were developed to target each node of the KRAS signaling pathway and then evaluated in preclinical or clinical studies. Created with BioRender.com
Fig. 2
Fig. 2
Frequency and distribution of KRAS mutations in CRC and the biochemical features of mutant KRAS proteins. a Percentage of KRAS mutation in CRC and the diversity of KRAS alleles. Data acquired from The Cancer Genome Atlas (pan-Cancer) from cBioPortal. b Overview of generalized biochemical change of hydrolysis and guanine exchange following mutations in codons 12 (orange), 13 (purple) or, 61 (blue). The dashed line indicates hydrolysis and the solid line indicates guanine exchange, with the thicker line indicating faster rates and vice versa for slower rates. Created with BioRender.com. WT, wild type; MUT, mutant type
Fig. 3
Fig. 3
Chemical structures of KRASG12C covalent inhibitors with their initial publication date. AMG 510 has received accelerated approval from the U.S. FDA for the treatment of patients with NSCLC in May 2021. MRTX849 has been granted Breakthrough Therapy Designation by FDA in June 2021

References

    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.
    1. Kennedy RD, Bylesjo M, Kerr P, Davison T, Black JM, Kay EW, Holt RJ, Proutski V, Ahdesmaki M, Farztdinov V, et al. Development and independent validation of a prognostic assay for stage II colon cancer using formalin-fixed paraffin-embedded tissue. J Clin Oncol. 2011;29:4620–4626.
    1. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990;61:759–767.
    1. Sagaert X, Vanstapel A, Verbeek S. Tumor heterogeneity in colorectal Cancer: what Do we know so far? Pathobiology. 2018;85:72–84.
    1. Sasaki N, Clevers H. Studying cellular heterogeneity and drug sensitivity in colorectal cancer using organoid technology. Curr Opin Genet Dev. 2018;52:117–122.
    1. Martini G, Dienstmann R, Ros J, Baraibar I, Cuadra-Urteaga JL, Salva F, Ciardiello D, Mulet N, Argiles G, Tabernero J, Elez E. Molecular subtypes and the evolution of treatment management in metastatic colorectal cancer. Ther Adv Med Oncol. 2020;12:1758835920936089.
    1. Dienstmann R, Connor K, Byrne AT. Precision therapy in RAS mutant colorectal Cancer. Gastroenterology. 2020;158:806–811.
    1. Roth AD, Tejpar S, Delorenzi M, Yan P, Fiocca R, Klingbiel D, Dietrich D, Biesmans B, Bodoky G, Barone C, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol. 2010;28:466–474.
    1. Dienstmann R, Mason MJ, Sinicrope FA, Phipps AI, Tejpar S, Nesbakken A, Danielsen SA, Sveen A, Buchanan DD, Clendenning M, et al. Prediction of overall survival in stage II and III colon cancer beyond TNM system: a retrospective, pooled biomarker study. Ann Oncol. 2017;28:1023–1031.
    1. Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD, Robitaille S, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359:1757–1765.
    1. Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, Juan T, Sikorski R, Suggs S, Radinsky R, et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26:1626–1634.
    1. Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, Falchook GS, Price TJ, Sacher A, Denlinger CS, et al. KRAS(G12C) inhibition with Sotorasib in advanced solid tumors. N Engl J Med. 2020;383:1207–1217.
    1. Chen Z, Otto JC, Bergo MO, Young SG, Casey PJ. The C-terminal polylysine region and methylation of K-Ras are critical for the interaction between K-Ras and microtubules. J Biol Chem. 2000;275:41251–41257.
    1. Pells S, Divjak M, Romanowski P, Impey H, Hawkins NJ, Clarke AR, Hooper ML, Williamson DJ. Developmentally-regulated expression of murine K-ras isoforms. Oncogene. 1997;15:1781–1786.
    1. Tsai FD, Lopes MS, Zhou M, Court H, Ponce O, Fiordalisi JJ, Gierut JJ, Cox AD, Haigis KM, Philips MR. K-Ras4A splice variant is widely expressed in cancer and uses a hybrid membrane-targeting motif. Proc Natl Acad Sci U S A. 2015;112:779–784.
    1. Chen W-C, To MD, Westcott PMK, Delrosario R, Kim I-J, Philips M, Tran Q, Bayani N, Balmain A. Regulation of KRAS4A/B splicing in cancer stem cells by the RBM39 splicing complex. Preprint at bioRxiv. 2019. 10.1101/646125.
    1. Bourne HR, Sanders DA, McCormick F. The GTPase superfamily: conserved structure and molecular mechanism. Nature. 1991;349:117–127.
    1. Hancock JF, Parton RG. Ras plasma membrane signalling platforms. Biochem J. 2005;389:1–11.
    1. Vögler O, Barceló JM, Ribas C, Escribá PV. Membrane interactions of G proteins and other related proteins. Biochim Biophys Acta. 2008;1778:1640–1652.
    1. Simanshu DK, Nissley DV, McCormick F. RAS proteins and their regulators in human disease. Cell. 2017;170:17–33.
    1. Cherfils J, Zeghouf M. Regulation of small GTPases by GEFs, GAPs, and GDIs. Physiol Rev. 2013;93:269–309.
    1. Li Z, Chen Y, Wang D, Wang G, He L, Suo J. Detection of KRAS mutations and their associations with clinicopathological features and survival in Chinese colorectal cancer patients. J Int Med Res. 2012;40:1589–1598.
    1. Cercek A, Braghiroli MI, Chou JF, Hechtman JF, Kemeny N, Saltz L, Capanu M, Yaeger R. Clinical features and outcomes of patients with colorectal cancers harboring NRAS mutations. Clin Cancer Res. 2017;23:4753–4760.
    1. Haigis KM. KRAS alleles: the devil is in the detail. Trends Cancer. 2017;3:686–697.
    1. Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and structural analysis of common Cancer-associated KRAS mutations. Mol Cancer Res. 2015;13:1325–1335.
    1. Smith MJ, Neel BG, Ikura M. NMR-based functional profiling of RASopathies and oncogenic RAS mutations. Proc Natl Acad Sci U S A. 2013;110:4574–4579.
    1. Imamura Y, Morikawa T, Liao X, Lochhead P, Kuchiba A, Yamauchi M, Qian ZR, Nishihara R, Meyerhardt JA, Haigis KM, et al. Specific mutations in KRAS codons 12 and 13, and patient prognosis in 1075 BRAF wild-type colorectal cancers. Clin Cancer Res. 2012;18:4753–4763.
    1. Jones RP, Sutton PA, Evans JP, Clifford R, McAvoy A, Lewis J, Rousseau A, Mountford R, McWhirter D, Malik HZ. Specific mutations in KRAS codon 12 are associated with worse overall survival in patients with advanced and recurrent colorectal cancer. Br J Cancer. 2017;116:923–929.
    1. Modest DP, Ricard I, Heinemann V, Hegewisch-Becker S, Schmiegel W, Porschen R, Stintzing S, Graeven U, Arnold D, von Weikersthal LF, et al. Outcome according to KRAS-, NRAS- and BRAF-mutation as well as KRAS mutation variants: pooled analysis of five randomized trials in metastatic colorectal cancer by the AIO colorectal cancer study group. Ann Oncol. 2016;27:1746–1753.
    1. Camaj P, Primo S, Wang Y, Heinemann V, Zhao Y, Laubender RP, Stintzing S, Giessen-Jung C, Jung A, Gamba S, et al. KRAS exon 2 mutations influence activity of regorafenib in an SW48-based disease model of colorectal cancer. Future Oncol. 2015;11:1919–1929.
    1. Zocche DM, Ramirez C, Fontao FM, Costa LD, Redal MA. Global impact of KRAS mutation patterns in FOLFOX treated metastatic colorectal cancer. Front Genet. 2015;6:116.
    1. Jonker DJ, O'Callaghan CJ, Karapetis CS, Zalcberg JR, Tu D, Au HJ, Berry SR, Krahn M, Price T, Simes RJ, et al. Cetuximab for the treatment of colorectal cancer. N Engl J Med. 2007;357:2040–2048.
    1. Loupakis F, Ruzzo A, Cremolini C, Vincenzi B, Salvatore L, Santini D, Masi G, Stasi I, Canestrari E, Rulli E, et al. KRAS codon 61, 146 and BRAF mutations predict resistance to cetuximab plus irinotecan in KRAS codon 12 and 13 wild-type metastatic colorectal cancer. Br J Cancer. 2009;101:715–721.
    1. Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023–1034.
    1. Allegra CJ, Rumble RB, Hamilton SR, Mangu PB, Roach N, Hantel A, Schilsky RL. Extended RAS gene mutation testing in metastatic colorectal carcinoma to predict response to anti-epidermal growth factor receptor monoclonal antibody therapy: American Society of Clinical Oncology provisional clinical opinion update 2015. J Clin Oncol. 2016;34:179–185.
    1. De Roock W, Jonker DJ, Di Nicolantonio F, Sartore-Bianchi A, Tu D, Siena S, Lamba S, Arena S, Frattini M, Piessevaux H, et al. Association of KRAS p.G13D mutation with outcome in patients with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab. JAMA. 2010;304:1812–1820.
    1. Tejpar S, Celik I, Schlichting M, Sartorius U, Bokemeyer C, Van Cutsem E. Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab. J Clin Oncol. 2012;30:3570–3577.
    1. De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent-Puig P, et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 2010;11:753–762.
    1. Bozzetti C, Negri FV, Azzoni C, Naldi N, Nizzoli R, Bortesi B, Zobbi V, Bottarelli L, Tiseo M, Silini EM, Ardizzoni A. Epidermal growth factor receptor and Kras gene expression: reliability of mutational analysis on cytological samples. Diagn Cytopathol. 2013;41:595–598.
    1. Mohamed Suhaimi NA, Foong YM, Lee DY, Phyo WM, Cima I, Lee EX, Goh WL, Lim WY, Chia KS, Kong SL, et al. Non-invasive sensitive detection of KRAS and BRAF mutation in circulating tumor cells of colorectal cancer patients. Mol Oncol. 2015;9:850–860.
    1. Isler JA, Vesterqvist OE, Burczynski ME. Analytical validation of genotyping assays in the biomarker laboratory. Pharmacogenomics. 2007;8:353–368.
    1. Linardou H, Briasoulis E, Dahabreh IJ, Mountzios G, Papadimitriou C, Papadopoulos S, Bafaloukos D, Kosmidis P, Murray S. All about KRAS for clinical oncology practice: gene profile, clinical implications and laboratory recommendations for somatic mutational testing in colorectal cancer. Cancer Treat Rev. 2011;37:221–233.
    1. Cross J. DxS Ltd. Pharmacogenomics. 2008;9:463–467.
    1. Tol J, Koopman M, Cats A, Rodenburg CJ, Creemers GJ, Schrama JG, Erdkamp FL, Vos AH, van Groeningen CJ, Sinnige HA, et al. Chemotherapy, bevacizumab, and cetuximab in metastatic colorectal cancer. N Engl J Med. 2009;360:563–572.
    1. Fariña Sarasqueta A, Moerland E, de Bruyne H, de Graaf H, Vrancken T, van Lijnschoten G, van den Brule AJ. SNaPshot and StripAssay as valuable alternatives to direct sequencing for KRAS mutation detection in colon cancer routine diagnostics. J Mol Diagn. 2011;13:199–205.
    1. Gonzalez de Castro D, Angulo B, Gomez B, Mair D, Martinez R, Suarez-Gauthier A, Shieh F, Velez M, Brophy VH, Lawrence HJ, Lopez-Rios F. A comparison of three methods for detecting KRAS mutations in formalin-fixed colorectal cancer specimens. Br J Cancer. 2012;107:345–351.
    1. Harlé A, Filhine-Tresarrieu P, Husson M, Boidot R, Rouyer M, Dubois C, Leroux A, Merlin JL. Rare RAS mutations in metastatic colorectal Cancer detected during routine RAS genotyping using next generation sequencing. Target Oncol. 2016;11:363–370.
    1. Dong L, Wang S, Fu B, Wang J. Evaluation of droplet digital PCR and next generation sequencing for characterizing DNA reference material for KRAS mutation detection. Sci Rep. 2018;8:9650.
    1. Taly V, Pekin D, Benhaim L, Kotsopoulos SK, Le Corre D, Li X, Atochin I, Link DR, Griffiths AD, Pallier K, et al. Multiplex picodroplet digital PCR to detect KRAS mutations in circulating DNA from the plasma of colorectal cancer patients. Clin Chem. 2013;59:1722–1731.
    1. García-Foncillas J, Alba E, Aranda E, Díaz-Rubio E, López-López R, Tabernero J, Vivancos A. Incorporating BEAMing technology as a liquid biopsy into clinical practice for the management of colorectal cancer patients: an expert taskforce review. Ann Oncol. 2017;28:2943–2949.
    1. Timar J, Kashofer K. Molecular epidemiology and diagnostics of KRAS mutations in human cancer. Cancer Metastasis Rev. 2020;39:1029–1038.
    1. Thierry AR, Mouliere F, El Messaoudi S, Mollevi C, Lopez-Crapez E, Rolet F, Gillet B, Gongora C, Dechelotte P, Robert B, et al. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat Med. 2014;20:430–435.
    1. Van Cutsem E, Köhne CH, Láng I, Folprecht G, Nowacki MP, Cascinu S, Shchepotin I, Maurel J, Cunningham D, Tejpar S, et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol. 2011;29:2011–2019.
    1. Normanno N, Cervantes A, Ciardiello F, De Luca A, Pinto C. The liquid biopsy in the management of colorectal cancer patients: current applications and future scenarios. Cancer Treat Rev. 2018;70:1–8.
    1. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, Thornton K, Agrawal N, Sokoll L, Szabo SA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14:985–990.
    1. Garcia J, Forestier J, Dusserre E, Wozny AS, Geiguer F, Merle P, Tissot C, Ferraro-Peyret C, Jones FS, Edelstein DL, et al. Cross-platform comparison for the detection of RAS mutations in cfDNA (ddPCR biorad detection assay, BEAMing assay, and NGS strategy) Oncotarget. 2018;9:21122–21131.
    1. Tabernero J, Lenz HJ, Siena S, Sobrero A, Falcone A, Ychou M, Humblet Y, Bouché O, Mineur L, Barone C, et al. Analysis of circulating DNA and protein biomarkers to predict the clinical activity of regorafenib and assess prognosis in patients with metastatic colorectal cancer: a retrospective, exploratory analysis of the CORRECT trial. Lancet Oncol. 2015;16:937–948.
    1. Papke B, Der CJ. Drugging RAS: know the enemy. Science. 2017;355:1158–1163.
    1. Ostrem JM, Shokat KM. Direct small-molecule inhibitors of KRAS: from structural insights to mechanism-based design. Nat Rev Drug Discov. 2016;15:771–785.
    1. Passiglia F, Malapelle U, Del Re M, Righi L, Pagni F, Furlan D, Danesi R, Troncone G, Novello S. KRAS inhibition in non-small cell lung cancer: past failures, new findings and upcoming challenges. Eur J Cancer. 2020;137:57–68.
    1. Nagasaka M, Li Y, Sukari A, Ou SI, Al-Hallak MN, Azmi AS. KRAS G12C game of thrones, which direct KRAS inhibitor will claim the iron throne? Cancer Treat Rev. 2020;84:101974.
    1. Serna-Blasco R, Sanz-Álvarez M, Aguilera Ó, García-Foncillas J. Targeting the RAS-dependent chemoresistance: the Warburg connection. Semin Cancer Biol. 2019;54:80–90.
    1. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503:548–551.
    1. Patricelli MP, Janes MR, Li LS, Hansen R, Peters U, Kessler LV, Chen Y, Kucharski JM, Feng J, Ely T, et al. Selective inhibition of oncogenic KRAS output with small molecules targeting the inactive state. Cancer Discov. 2016;6:316–329.
    1. Janes MR, Zhang J, Li LS, Hansen R, Peters U, Guo X, Chen Y, Babbar A, Firdaus SJ, Darjania L, et al. Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell. 2018;172:578–589.
    1. Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, Gaida K, Holt T, Knutson CG, Koppada N, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019;575:217–223.
    1. Hallin J, Engstrom LD, Hargis L, Calinisan A, Aranda R, Briere DM, Sudhakar N, Bowcut V, Baer BR, Ballard JA, 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.
    1. Christensen JG, Olson P, Briere T, Wiel C, Bergo MO. Targeting Kras(g12c) -mutant cancer with a mutation-specific inhibitor. J Intern Med. 2020;288:183–191.
    1. FDA Approves LUMAKRAS™ (Sotorasib), The First And Only Targeted Treatment For Patients With KRAS G12C-Mutated Locally Advanced Or Metastatic Non-Small Cell Lung Cancer. 2021 .
    1. Gentile DR, Rathinaswamy MK, Jenkins ML, Moss SM, Siempelkamp BD, Renslo AR, Burke JE, Shokat KM. Ras Binder Induces a Modified Switch-II Pocket in GTP and GDP States. Cell Chem Biol. 2017;24:1455–1466.
    1. Amodio V, Yaeger R, Arcella P, Cancelliere C, Lamba S, Lorenzato A, Arena S, Montone M, Mussolin B, Bian Y, et al. EGFR blockade reverts resistance to KRAS(G12C) inhibition in colorectal Cancer. Cancer Discov. 2020;10:1129–1139.
    1. Mirati Therapeutics' Adagrasib Receives Breakthrough Therapy Designation from U.S. Food and Drug Administration for Patients with Advanced Non-Small Cell Lung Cancer Harboring the KRASG12C Mutation. 2021. .
    1. Mirati Therapeutics Reports Investigational Adagrasib (MRTX849) Preliminary Data Demonstrating Tolerability and Durable Anti-Tumor Activity as well as Initial MRTX1133 Preclinical Data. 2020. .
    1. Lito P, Solomon M, Li LS, Hansen R, Rosen N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science. 2016;351:604–608.
    1. Maurer T, Garrenton LS, Oh A, Pitts K, Anderson DJ, Skelton NJ, Fauber BP, Pan B, Malek S, Stokoe D, et al. Small-molecule ligands bind to a distinct pocket in Ras and inhibit SOS-mediated nucleotide exchange activity. Proc Natl Acad Sci U S A. 2012;109:5299–5304.
    1. Sun Q, Burke JP, Phan J, Burns MC, Olejniczak ET, Waterson AG, Lee T, Rossanese OW, Fesik SW. Discovery of small molecules that bind to K-Ras and inhibit Sos-mediated activation. Angew Chem Int Ed Eng. 2012;51:6140–6143.
    1. Shima F, Yoshikawa Y, Ye M, Araki M, Matsumoto S, Liao J, Hu L, Sugimoto T, Ijiri Y, Takeda A, 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;110:8182–8187.
    1. Burns MC, Sun Q, Daniels RN, Camper D, Kennedy JP, Phan J, Olejniczak ET, Lee T, Waterson AG, Rossanese OW, Fesik SW. Approach for targeting Ras with small molecules that activate SOS-mediated nucleotide exchange. Proc Natl Acad Sci U S A. 2014;111:3401–3406.
    1. Hillig RC, Sautier B, Schroeder J, Moosmayer D, Hilpmann A, Stegmann CM, Werbeck ND, Briem H, Boemer U, Weiske J, et al. Discovery of potent SOS1 inhibitors that block RAS activation via disruption of the RAS-SOS1 interaction. Proc Natl Acad Sci U S A. 2019;116:2551–2560.
    1. Hofmann MH, Gmachl M, Ramharter J, Savarese F, Gerlach D, Marszalek JR, Sanderson MP, Kessler D, Trapani F, Arnhof H, et al. BI-3406, a potent and selective SOS1-KRAS interaction inhibitor, is effective in KRAS-driven cancers through combined MEK inhibition. Cancer Discov. 2021;11:142–157.
    1. Li W, Nishimura R, Kashishian A, Batzer AG, Kim WJ, Cooper JA, Schlessinger J. A new function for a phosphotyrosine phosphatase: linking GRB2-Sos to a receptor tyrosine kinase. Mol Cell Biol. 1994;14:509–517.
    1. Bennett AM, Tang TL, Sugimoto S, Walsh CT, Neel BG. Protein-tyrosine-phosphatase SHPTP2 couples platelet-derived growth factor receptor beta to Ras. Proc Natl Acad Sci U S A. 1994;91:7335–7339.
    1. Chen X, Zou F, Hu Z, Du G, Yu P, Wang W, Wang H, Ye L, Tian J. PCC0208023, a potent SHP2 allosteric inhibitor, imparts an antitumor effect against KRAS mutant colorectal cancer. Toxicol Appl Pharmacol. 2020;398:115019.
    1. Wong GS, Zhou J, Liu JB, Wu Z, Xu X, Li T, Xu D, Schumacher SE, Puschhof J, McFarland J, et al. Targeting wild-type KRAS-amplified gastroesophageal cancer through combined MEK and SHP2 inhibition. Nat Med. 2018;24:968–977.
    1. Fedele C, Ran H, Diskin B, Wei W, Jen J, Geer MJ, Araki K, Ozerdem U, Simeone DM, Miller G, et al. SHP2 inhibition prevents adaptive resistance to MEK inhibitors in multiple Cancer models. Cancer Discov. 2018;8:1237–1249.
    1. Lu H, Liu C, Velazquez R, Wang H, Dunkl LM, Kazic-Legueux M, Haberkorn A, Billy E, Manchado E, Brachmann SM, et al. SHP2 inhibition overcomes RTK-mediated pathway reactivation in KRAS-mutant tumors treated with MEK inhibitors. Mol Cancer Ther. 2019;18:1323–1334.
    1. Liu C, Lu H, Wang H, Loo A, Zhang X, Yang G, Kowal C, Delach S, Wang Y, Goldoni S, et al. Combinations with allosteric SHP2 inhibitor TNO155 to block receptor tyrosine kinase signaling. Clin Cancer Res. 2021;27:342–354.
    1. Quintana E, Schulze CJ, Myers DR, Choy TJ, Mordec K, Wildes D, Shifrin NT, Belwafa A, Koltun ES, Gill AL, et al. Allosteric inhibition of SHP2 stimulates antitumor immunity by transforming the immunosuppressive environment. Cancer Res. 2020;80:2889–2902.
    1. End DW, Smets G, Todd AV, Applegate TL, Fuery CJ, Angibaud P, Venet M, Sanz G, Poignet H, Skrzat S, et al. Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer Res. 2001;61:131–137.
    1. Whitehead RP, McCoy S, Macdonald JS, Rivkin SE, Neubauer MA, Dakhil SR, Lenz HJ, Tanaka MS, Abbruzzese JL. Phase II trial of R115777 (NSC #70818) in patients with advanced colorectal cancer: a southwest oncology group study. Investig New Drugs. 2006;24:335–341.
    1. Rao S, Cunningham D, de Gramont A, Scheithauer W, Smakal M, Humblet Y, Kourteva G, Iveson T, Andre T, Dostalova J, et al. Phase III double-blind placebo-controlled study of farnesyl transferase inhibitor R115777 in patients with refractory advanced colorectal cancer. J Clin Oncol. 2004;22:3950–3957.
    1. Winter-Vann AM, Baron RA. Wong W, dela Cruz J, York JD, Gooden DM, Bergo MO, Young SG, Toone EJ, Casey PJ: a small-molecule inhibitor of isoprenylcysteine carboxyl methyltransferase with antitumor activity in cancer cells. Proc Natl Acad Sci U S A. 2005;102:4336–4341.
    1. Marín-Ramos NI, Balabasquer M, Ortega-Nogales FJ, Torrecillas IR, Gil-Ordóñez A, Marcos-Ramiro B, Aguilar-Garrido P, Cushman I, Romero A, Medrano FJ, et al. A potent Isoprenylcysteine Carboxylmethyltransferase (ICMT) inhibitor improves survival in Ras-driven acute myeloid leukemia. J Med Chem. 2019;62:6035–6046.
    1. Chandra A, Grecco HE, Pisupati V, Perera D, Cassidy L, Skoulidis F, Ismail SA, Hedberg C, Hanzal-Bayer M, Venkitaraman AR, et al. The GDI-like solubilizing factor PDEδ sustains the spatial organization and signalling of Ras family proteins. Nat Cell Biol. 2011;14:148–158.
    1. Zimmermann G, Papke B, Ismail S, Vartak N, Chandra A, Hoffmann M, Hahn SA, Triola G, Wittinghofer A, Bastiaens PI, Waldmann H. Small molecule inhibition of the KRAS-PDEδ interaction impairs oncogenic KRAS signalling. Nature. 2013;497:638–642.
    1. Klein CH, Truxius DC, Vogel HA, Harizanova J, Murarka S, Martín-Gago P, Bastiaens PIH. PDEδ inhibition impedes the proliferation and survival of human colorectal cancer cell lines harboring oncogenic KRas. Int J Cancer. 2019;144:767–776.
    1. Martín-Gago P, Fansa EK, Klein CH, Murarka S, Janning P, Schürmann M, Metz M, Ismail S, Schultz-Fademrecht C, Baumann M, et al. A PDE6δ-KRas inhibitor Chemotype with up to seven H-bonds and Picomolar affinity that prevents efficient inhibitor release by Arl2. Angew Chem Int Ed Eng. 2017;56:2423–2428.
    1. Cheng J, Li Y, Wang X, Dong G, Sheng C. Discovery of novel PDEδ degraders for the treatment of KRAS mutant colorectal Cancer. J Med Chem. 2020;63:7892–7905.
    1. Fang JY, Richardson BC. The MAPK signalling pathways and colorectal cancer. Lancet Oncol. 2005;6:322–327.
    1. Roskoski R., Jr ERK1/2 MAP kinases: structure, function, and regulation. Pharmacol Res. 2012;66:105–143.
    1. Peng SB, Henry JR, Kaufman MD, Lu WP, Smith BD, Vogeti S, Rutkoski TJ, Wise S, Chun L, Zhang Y, et al. Inhibition of RAF isoforms and active dimers by LY3009120 leads to anti-tumor activities in RAS or BRAF mutant cancers. Cancer Cell. 2015;28:384–398.
    1. Vakana E, Pratt S, Blosser W, Dowless M, Simpson N, Yuan XJ, Jaken S, Manro J, Stephens J, Zhang Y, et al. LY3009120, a panRAF inhibitor, has significant anti-tumor activity in BRAF and KRAS mutant preclinical models of colorectal cancer. Oncotarget. 2017;8:9251–9266.
    1. Yao YM, Donoho GP, Iversen PW, Zhang Y, Van Horn RD, Forest A, Novosiadly RD, Webster YW, Ebert P, Bray S, et al. Mouse PDX trial suggests synergy of concurrent inhibition of RAF and EGFR in colorectal Cancer with BRAF or KRAS mutations. Clin Cancer Res. 2017;23:5547–5560.
    1. Sullivan RJ, Hollebecque A, Flaherty KT, Shapiro GI, Rodon Ahnert J, Millward MJ, Zhang W, Gao L, Sykes A, Willard MD, et al. A phase I study of LY3009120, a Pan-RAF inhibitor, in patients with advanced or metastatic Cancer. Mol Cancer Ther. 2020;19:460–467.
    1. Desai J, Gan H, Barrow C, Jameson M, Atkinson V, Haydon A, Millward M, Begbie S, Brown M, Markman B, et al. Phase I, open-label, dose-escalation/dose-expansion study of Lifirafenib (BGB-283), an RAF family kinase inhibitor, in patients with solid tumors. J Clin Oncol. 2020;38:2140–2150.
    1. Zimmer L, Barlesi F, Martinez-Garcia M, Dieras V, Schellens JH, Spano JP, Middleton MR, Calvo E, Paz-Ares L, Larkin J, et al. Phase I expansion and pharmacodynamic study of the oral MEK inhibitor RO4987655 (CH4987655) in selected patients with advanced cancer with RAS-RAF mutations. Clin Cancer Res. 2014;20:4251–4261.
    1. Blumenschein GR, Jr, Smit EF, Planchard D, Kim DW, Cadranel J, De Pas T, Dunphy F, Udud K, Ahn MJ, Hanna NH, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC)†. Ann Oncol. 2015;26:894–901.
    1. Friday BB, Yu C, Dy GK, Smith PD, Wang L, Thibodeau SN, Adjei AA. BRAF V600E disrupts AZD6244-induced abrogation of negative feedback pathways between extracellular signal-regulated kinase and Raf proteins. Cancer Res. 2008;68:6145–6153.
    1. Pratilas CA, Taylor BS, Ye Q, Viale A, Sander C, Solit DB. Rosen N: (V600E) BRAF is associated with disabled feedback inhibition of RAF-MEK signaling and elevated transcriptional output of the pathway. Proc Natl Acad Sci U S A. 2009;106:4519–4524.
    1. Lamba S, Russo M, Sun C, Lazzari L, Cancelliere C, Grernrum W, Lieftink C, Bernards R, Di Nicolantonio F, Bardelli A. RAF suppression synergizes with MEK inhibition in KRAS mutant cancer cells. Cell Rep. 2014;8:1475–1483.
    1. Yen I, Shanahan F, Merchant M, Orr C, Hunsaker T, Durk M, La H, Zhang X, Martin SE, Lin E, et al. Pharmacological Induction of RAS-GTP Confers RAF Inhibitor Sensitivity in KRAS Mutant Tumors. Cancer Cell. 2018;34:611–625.
    1. Xu H, Zhou S, Xia H, Yu H, Tang Q, Bi F. MEK nuclear localization promotes YAP stability via sequestering β-TrCP in KRAS mutant cancer cells. Cell Death Differ. 2019;26:2400–2415.
    1. Dai X, Xia H, Zhou S, Tang Q, Bi F. Zoledronic acid enhances the efficacy of the MEK inhibitor trametinib in KRAS mutant cancers. Cancer Lett. 2019;442:202–212.
    1. Varga A, Soria JC, Hollebecque A, LoRusso P, Bendell J, Huang SA, Wagle MC, Okrah K, Liu L, Murray E, et al. A first-in-human phase I study to evaluate the ERK1/2 inhibitor GDC-0994 in patients with advanced solid tumors. Clin Cancer Res. 2020;26:1229–1236.
    1. Merchant M, Moffat J, Schaefer G, Chan J, Wang X, Orr C, Cheng J, Hunsaker T, Shao L, Wang SJ, et al. Combined MEK and ERK inhibition overcomes therapy-mediated pathway reactivation in RAS mutant tumors. PLoS One. 2017;12:e0185862.
    1. Mallucci L, Wells V. The end of KRAS, and other, cancers? A new way forward. Drug Discov Today. 2014;19:383–387.
    1. Hoeflich KP, Merchant M, Orr C, Chan J, Den Otter D, Berry L, Kasman I, Koeppen H, Rice K, Yang NY, et al. Intermittent administration of MEK inhibitor GDC-0973 plus PI3K inhibitor GDC-0941 triggers robust apoptosis and tumor growth inhibition. Cancer Res. 2012;72:210–219.
    1. Köhler J, Zhao Y, Li J, Gokhale PC, Tiv HL, Knott AR, Wilkens MK, Soroko KM, Lin M, Ambrogio C, et al. ERK inhibitor LY3214996-based treatment strategies for RAS-driven lung Cancer. Mol Cancer Ther. 2021;20:641–654.
    1. Martinelli E, Troiani T, D'Aiuto E, Morgillo F, Vitagliano D, Capasso A, Costantino S, Ciuffreda LP, Merolla F, Vecchione L, et al. Antitumor activity of pimasertib, a selective MEK 1/2 inhibitor, in combination with PI3K/mTOR inhibitors or with multi-targeted kinase inhibitors in pimasertib-resistant human lung and colorectal cancer cells. Int J Cancer. 2013;133:2089–2101.
    1. Do K, Speranza G, Bishop R, Khin S, Rubinstein L, Kinders RJ, Datiles M, Eugeni M, Lam MH, Doyle LA, et al. Biomarker-driven phase 2 study of MK-2206 and selumetinib (AZD6244, ARRY-142886) in patients with colorectal cancer. Investig New Drugs. 2015;33:720–728.
    1. Shapiro GI, LoRusso P, Kwak E, Pandya S, Rudin CM, Kurkjian C, Cleary JM, Pilat MJ, Jones S, de Crespigny A, et al. Phase Ib study of the MEK inhibitor cobimetinib (GDC-0973) in combination with the PI3K inhibitor pictilisib (GDC-0941) in patients with advanced solid tumors. Investig New Drugs. 2020;38:419–432.
    1. Ramanathan RK, Von Hoff DD, Eskens F, Blumenschein G, Jr, Richards D, Genvresse I, Reschke S, Granvil C, Skubala A, Peña C, Mross K. Phase Ib trial of the PI3K inhibitor Copanlisib combined with the allosteric MEK inhibitor Refametinib in patients with advanced Cancer. Target Oncol. 2020;15:163–174.
    1. Tsubaki M, Takeda T, Noguchi M, Jinushi M, Seki S, Morii Y, et al. Overactivation of Akt Contributes to MEK Inhibitor Primary and Acquired Resistance in Colorectal Cancer Cells. Cancers (Basel) 2019;11:1866–1884.
    1. Wee S, Jagani Z, Xiang KX, Loo A, Dorsch M, Yao YM, Sellers WR, Lengauer C, Stegmeier F. PI3K pathway activation mediates resistance to MEK inhibitors in KRAS mutant cancers. Cancer Res. 2009;69:4286–4293.
    1. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–330.
    1. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, Bignell GR, Bolli N, Borg A, Børresen-Dale AL, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421.
    1. Patnaik A, Kang SP, Rasco D, Papadopoulos KP, Elassaiss-Schaap J, Beeram M, Drengler R, Chen C, Smith L, Espino G, et al. Phase I study of Pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. Clin Cancer Res. 2015;21:4286–4293.
    1. Liu J, Huang X, Liu H, Wei C, Ru H, Qin H, Lai H, Meng Y, Wu G, Xie W, et al. Immune landscape and prognostic immune-related genes in KRAS-mutant colorectal cancer patients. J Transl Med. 2021;19:27.
    1. Lal N, White BS, Goussous G, Pickles O, Mason MJ, Beggs AD, Taniere P, Willcox BE, Guinney J, Middleton GW. KRAS mutation and consensus molecular subtypes 2 and 3 are independently associated with reduced immune infiltration and reactivity in colorectal Cancer. Clin Cancer Res. 2018;24:224–233.
    1. Poon E, Mullins S, Watkins A, Williams GS, Koopmann JO, Di Genova G, Cumberbatch M, Veldman-Jones M, Grosskurth SE, Sah V, et al. The MEK inhibitor selumetinib complements CTLA-4 blockade by reprogramming the tumor immune microenvironment. J Immunother Cancer. 2017;5:63.
    1. Tran E, Robbins PF, Lu YC, Prickett TD, Gartner JJ, Jia L, Pasetto A, Zheng Z, Ray S, Groh EM, et al. T-cell transfer therapy targeting mutant KRAS in Cancer. N Engl J Med. 2016;375:2255–2262.
    1. Veatch JR, Jesernig BL, Kargl J, Fitzgibbon M, Lee SM, Baik C, Martins R, Houghton AM, Riddell SR. Endogenous CD4(+) T cells recognize Neoantigens in lung Cancer patients, including recurrent oncogenic KRAS and ERBB2 (Her2) driver mutations. Cancer Immunol Res. 2019;7:910–922.
    1. Wang QJ, Yu Z, Griffith K, Hanada K, Restifo NP, Yang JC. Identification of T-cell receptors targeting KRAS-mutated human tumors. Cancer Immunol Res. 2016;4:204–214.
    1. Lu Y, Bellgrau D, Dwyer-Nield LD, Malkinson AM, Duke RC, Rodell TC, Franzusoff A. Mutation-selective tumor remission with Ras-targeted, whole yeast-based immunotherapy. Cancer Res. 2004;64:5084–5088.
    1. Cohn A, Morse MA, O'Neil B, Whiting S, Coeshott C, Ferraro J, Bellgrau D, Apelian D, Rodell TC. Whole recombinant Saccharomyces cerevisiae yeast expressing Ras mutations as treatment for patients with solid tumors bearing Ras mutations: results from a phase 1 trial. J Immunother. 2018;41:141–150.
    1. Gjertsen MK, Buanes T, Rosseland AR, Bakka A, Gladhaug I, Søreide O, Eriksen JA, Møller M, Baksaas I, Lothe RA, et al. Intradermal ras peptide vaccination with granulocyte-macrophage colony-stimulating factor as adjuvant: clinical and immunological responses in patients with pancreatic adenocarcinoma. Int J Cancer. 2001;92:441–450.
    1. Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, Marisa L, Roepman P, Nyamundanda G, Angelino P, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21:1350–1356.

Source: PubMed

Подписаться