Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

Targeting KRAS in metastatic colorectal cancer: current strategies and emerging opportunities

Authors: Manuela Porru, Luca Pompili, Carla Caruso, Annamaria Biroccio, Carlo Leonetti

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Developing drugs that target KRAS, the most frequently mutated oncogene in cancer, has not been successful despite much concerted efforts dedicated towards it in the last thirty years. Considering the key role this driver oncogene plays, the pharmacological drugging of KRAS remains a key challenge for cancer research. In this review, we highlight the emerging experimental strategies for blocking KRAS function and signaling and its direct targeting. We also report on the results in this field of research produced by our group.
Literature
1.
go back to reference Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRefPubMed Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRefPubMed
2.
go back to reference Gurpinar E, Grizzle WE, Piazza GA. NSAIDs inhibit tumorigenesis, but how? Clin Cancer Res. 2014;20:1104–13.CrossRefPubMed Gurpinar E, Grizzle WE, Piazza GA. NSAIDs inhibit tumorigenesis, but how? Clin Cancer Res. 2014;20:1104–13.CrossRefPubMed
3.
go back to reference Meyskens FL Jr, McLaren CE, Pelot D, Fujikawa-Brooks S, Carpenter PM, Hawk E, et al. Difluoromethylornithine plus sulindac for the prevention of sporadic colorectal adenomas: a randomized placebo-controlled, double-blind trial. Cancer Prev Res (Phila) 2008;1:32–38. Meyskens FL Jr, McLaren CE, Pelot D, Fujikawa-Brooks S, Carpenter PM, Hawk E, et al. Difluoromethylornithine plus sulindac for the prevention of sporadic colorectal adenomas: a randomized placebo-controlled, double-blind trial. Cancer Prev Res (Phila) 2008;1:32–38.
5.
go back to reference Cassidy J, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, et al. Randomized phase III study of capecitabine plus oxaliplatin compared with fluorouracil/folinic acid plus oxaliplatin as first-line therapy for metastatic colorectal cancer. J Clin Oncol. 2008;26(12):2006.CrossRefPubMed Cassidy J, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, et al. Randomized phase III study of capecitabine plus oxaliplatin compared with fluorouracil/folinic acid plus oxaliplatin as first-line therapy for metastatic colorectal cancer. J Clin Oncol. 2008;26(12):2006.CrossRefPubMed
6.
go back to reference Falcone A, Ricci S, Brunetti I, Pfanner E, Allegrini G, Barbara C, et al. Phase III trial of infusional fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) compared with infusional fluorouracil, leucovorin, and irinotecan (FOLFIRI) as first-line treatment for metastatic colorectal cancer: the Gruppo Oncologico Nord Ovest. J Clin Oncol. 2007;25:1670–6.CrossRefPubMed Falcone A, Ricci S, Brunetti I, Pfanner E, Allegrini G, Barbara C, et al. Phase III trial of infusional fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) compared with infusional fluorouracil, leucovorin, and irinotecan (FOLFIRI) as first-line treatment for metastatic colorectal cancer: the Gruppo Oncologico Nord Ovest. J Clin Oncol. 2007;25:1670–6.CrossRefPubMed
7.
go back to reference Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27:1386–422.CrossRefPubMed Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27:1386–422.CrossRefPubMed
12.
go back to reference Tol J, Nagtegaal ID, Punt CJ. BRAF mutation in metastatic colorectal cancer. N Engl J Med. 2009;361:98–9.CrossRefPubMed Tol J, Nagtegaal ID, Punt CJ. BRAF mutation in metastatic colorectal cancer. N Engl J Med. 2009;361:98–9.CrossRefPubMed
13.
go back to reference Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337–45.CrossRefPubMed Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337–45.CrossRefPubMed
14.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
15.
go back to reference Siravegna G, Mussolin B, Buscarino M, Corti G, Cassingena A, Crisafulli G, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med. 2015;21:795–801.CrossRefPubMedPubMedCentral Siravegna G, Mussolin B, Buscarino M, Corti G, Cassingena A, Crisafulli G, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med. 2015;21:795–801.CrossRefPubMedPubMedCentral
16.
go back to reference Russo M, Siravegna G, Blaszkowsky LS, Corti G, Crisafulli G, Ahronian LG, et al. Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer. Cancer Discov. 2016;6:147–53.CrossRefPubMed Russo M, Siravegna G, Blaszkowsky LS, Corti G, Crisafulli G, Ahronian LG, et al. Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer. Cancer Discov. 2016;6:147–53.CrossRefPubMed
17.
go back to reference Pietrantonio F, Vernieri C, Siravegna G, Mennitto A, Berenato R, Perrone F, et al. Heterogeneity of acquired resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer. Clin Cancer Res. 2017;23:2414–22.CrossRefPubMed Pietrantonio F, Vernieri C, Siravegna G, Mennitto A, Berenato R, Perrone F, et al. Heterogeneity of acquired resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer. Clin Cancer Res. 2017;23:2414–22.CrossRefPubMed
18.
go back to reference Cross DA, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, et al. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov. 2014;4:1046–61.CrossRefPubMedPubMedCentral Cross DA, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, et al. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov. 2014;4:1046–61.CrossRefPubMedPubMedCentral
21.
go back to reference Papke B, Murarka S, Vogel HA, Martín-Gago P, Kovacevic M, Truxius DC, et al. Identification of pyrazolopyridazinones as PDEδ inhibitors. Nat Commun. 2016;7:11360.CrossRefPubMedPubMedCentral Papke B, Murarka S, Vogel HA, Martín-Gago P, Kovacevic M, Truxius DC, et al. Identification of pyrazolopyridazinones as PDEδ inhibitors. Nat Commun. 2016;7:11360.CrossRefPubMedPubMedCentral
22.
go back to reference G Z, Schultz-Fademrecht C, Küchler P, Murarka S, Ismail S, Triola G, et al. Structure guided design and kinetic analysis of highly potent benzimidazole inhibitors targeting the PDEδprenyl binding site. J Med Chem. 2014;57:5435–48.CrossRef G Z, Schultz-Fademrecht C, Küchler P, Murarka S, Ismail S, Triola G, et al. Structure guided design and kinetic analysis of highly potent benzimidazole inhibitors targeting the PDEδprenyl binding site. J Med Chem. 2014;57:5435–48.CrossRef
23.
go back to reference Heidorn SJ1, Milagre C, Whittaker S, Nourry A, Niculescu-Duvas I, Dhomen N, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell 2010;140:209–221. Heidorn SJ1, Milagre C, Whittaker S, Nourry A, Niculescu-Duvas I, Dhomen N, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell 2010;140:209–221.
24.
go back to reference Peng SB, Henry JR, Kaufman MD, Lu WP, Smith BD, Vogeti S, 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;28:384–98.CrossRefPubMed Peng SB, Henry JR, Kaufman MD, Lu WP, Smith BD, Vogeti S, 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;28:384–98.CrossRefPubMed
25.
go back to reference Vakana E, Pratt S, Blosser W, Dowless M, Simpson N, Yuan XJ, et al. LY3009120, a panRAF inhibitor, has significant anti-tumor activity in BRAF and KRAS mutant preclinical models of colorectal cancer. Oncotarget. 2017;8:9251–66.CrossRefPubMed Vakana E, Pratt S, Blosser W, Dowless M, Simpson N, Yuan XJ, et al. LY3009120, a panRAF inhibitor, has significant anti-tumor activity in BRAF and KRAS mutant preclinical models of colorectal cancer. Oncotarget. 2017;8:9251–66.CrossRefPubMed
26.
go back to reference Zhang C, Spevak W, Zhang Y, Burton EA, Ma Y, Habets G, et al. RAF inhibitors that evade paradoxical MAPK pathway activation. Nature. 2015;526:583–6.CrossRefPubMed Zhang C, Spevak W, Zhang Y, Burton EA, Ma Y, Habets G, et al. RAF inhibitors that evade paradoxical MAPK pathway activation. Nature. 2015;526:583–6.CrossRefPubMed
27.
go back to reference Tutuka CSA, Andrews MC, Mariadason JM, Ioannidis P, Hudson C, Cebon J, et al. PLX8394, a new generation BRAF inhibitor, selectively inhibits BRAF in colonic adenocarcinoma cells and prevents paradoxical MAPK pathway activation. Mol Cancer. 2017;16:112.CrossRefPubMedPubMedCentral Tutuka CSA, Andrews MC, Mariadason JM, Ioannidis P, Hudson C, Cebon J, et al. PLX8394, a new generation BRAF inhibitor, selectively inhibits BRAF in colonic adenocarcinoma cells and prevents paradoxical MAPK pathway activation. Mol Cancer. 2017;16:112.CrossRefPubMedPubMedCentral
28.
go back to reference Maurer T, Garrenton LS, Oh A, Pitts K, Anderson DJ, Skelton NJ, 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;109:5299–304.CrossRefPubMedPubMedCentral Maurer T, Garrenton LS, Oh A, Pitts K, Anderson DJ, Skelton NJ, 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;109:5299–304.CrossRefPubMedPubMedCentral
29.
go back to reference Sun Q, Burke JP, Phan J, Burns MC, Olejniczak ET, Waterson AG, et al. Discovery of small molecules that bind to K-Ras and inhibit Sos-mediated activation. Angew Chem Int Ed Engl. 2012;51:6140–3.CrossRefPubMedPubMedCentral Sun Q, Burke JP, Phan J, Burns MC, Olejniczak ET, Waterson AG, et al. Discovery of small molecules that bind to K-Ras and inhibit Sos-mediated activation. Angew Chem Int Ed Engl. 2012;51:6140–3.CrossRefPubMedPubMedCentral
30.
go back to reference Shima F, Yoshikawa Y, Ye M, Araki M, Matsumoto S, Liao J, 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;110:8182–7.CrossRefPubMedPubMedCentral Shima F, Yoshikawa Y, Ye M, Araki M, Matsumoto S, Liao J, 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;110:8182–7.CrossRefPubMedPubMedCentral
31.
go back to reference Wilson CY, Tolias P. Recent advances in cancer drug discovery targeting RAS. Drug Discov Today. 2016;21:1915–9.CrossRefPubMed Wilson CY, Tolias P. Recent advances in cancer drug discovery targeting RAS. Drug Discov Today. 2016;21:1915–9.CrossRefPubMed
32.
go back to reference Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503:548–51.CrossRefPubMedPubMedCentral Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503:548–51.CrossRefPubMedPubMedCentral
33.
go back to reference Patricelli MP, Janes MR, Li LS, Hansen R, Peters U, Kessler LV, et al. Selective inhibition of oncogenic KRAS output with small molecules targeting the inactive state. Cancer Discov. 2016;6:316–29.CrossRefPubMed Patricelli MP, Janes MR, Li LS, Hansen R, Peters U, Kessler LV, et al. Selective inhibition of oncogenic KRAS output with small molecules targeting the inactive state. Cancer Discov. 2016;6:316–29.CrossRefPubMed
34.
go back to reference Ross SJ, Revenko AS, Hanson LL, Ellston R, Staniszewska A, Whalley N, et al. Targeting KRAS-dependent tumors with AZD4785, a high-affinity therapeutic antisense oligonucleotide inhibitor of KRAS. Sci Transl Med. 2017;9(394):eaal5253. Ross SJ, Revenko AS, Hanson LL, Ellston R, Staniszewska A, Whalley N, et al. Targeting KRAS-dependent tumors with AZD4785, a high-affinity therapeutic antisense oligonucleotide inhibitor of KRAS. Sci Transl Med. 2017;9(394):eaal5253.
36.
go back to reference Hiraoka K, Inoue T, Taylor RD, Watanabe T, Koshikawa N, Yoda H, et al. Inhibition of KRAS codon 12 mutants using a novel DNA-alkylating pyrrole-imidazole polyamide conjugate. Nat Commun. 2015;6:6706.CrossRefPubMed Hiraoka K, Inoue T, Taylor RD, Watanabe T, Koshikawa N, Yoda H, et al. Inhibition of KRAS codon 12 mutants using a novel DNA-alkylating pyrrole-imidazole polyamide conjugate. Nat Commun. 2015;6:6706.CrossRefPubMed
37.
go back to reference Balasubramanian S, Hurley LH, Neidle S. Targeting G-quadruplexes in gene promoters: a novel anticancer strategy? Nat Rev Drug Discov. 2011;10:261–75.CrossRefPubMedPubMedCentral Balasubramanian S, Hurley LH, Neidle S. Targeting G-quadruplexes in gene promoters: a novel anticancer strategy? Nat Rev Drug Discov. 2011;10:261–75.CrossRefPubMedPubMedCentral
38.
go back to reference Chambers VS, Marsico G, Boutell JM, Di Antonio M, Smith GP, Balasubramanian S. High-throughput sequencing of DNA G-quadruplex structures in the human genome. Nat Biotechnol. 2015;33:877–81.CrossRefPubMed Chambers VS, Marsico G, Boutell JM, Di Antonio M, Smith GP, Balasubramanian S. High-throughput sequencing of DNA G-quadruplex structures in the human genome. Nat Biotechnol. 2015;33:877–81.CrossRefPubMed
39.
go back to reference Cogoi S, Xodo LE. G4 DNA in ras genes and its potential in cancer therapy. Biochim Biophys Acta. 2016;1859:663–74.CrossRefPubMed Cogoi S, Xodo LE. G4 DNA in ras genes and its potential in cancer therapy. Biochim Biophys Acta. 2016;1859:663–74.CrossRefPubMed
40.
go back to reference Morgan RK, Batra H, Gaerig VC, Hockings J, Brooks TA. Identification and characterization of a new G-quadruplex forming region within the kRAS promoter as a transcriptional regulator. Biochim Biophys Acta. 2016;1859:235–45.CrossRefPubMed Morgan RK, Batra H, Gaerig VC, Hockings J, Brooks TA. Identification and characterization of a new G-quadruplex forming region within the kRAS promoter as a transcriptional regulator. Biochim Biophys Acta. 2016;1859:235–45.CrossRefPubMed
41.
go back to reference Kaiser CE, Van Ert NA, Agrawal P, Chawla R, Yang D, Hurley LH. Insight into the complexity of the i-motif and G-Quadruplex DNA structures formed in the KRAS promoter and subsequent drug-induced gene repression. J Am Chem Soc. 2017;139:8522–36.CrossRefPubMed Kaiser CE, Van Ert NA, Agrawal P, Chawla R, Yang D, Hurley LH. Insight into the complexity of the i-motif and G-Quadruplex DNA structures formed in the KRAS promoter and subsequent drug-induced gene repression. J Am Chem Soc. 2017;139:8522–36.CrossRefPubMed
43.
go back to reference Lavrado J, Borralho PM, Ohnmacht SA, Castro RE, Rodrigues CM, Moreira R, et al. Synthesis, G-quadruplexstabilisation, docking studies, and effect on cancer cells of indolo[3,2-b]quinolines with one, two, or three basic side chains. Chem Med Chem 2013;8:1648–1661. Lavrado J, Borralho PM, Ohnmacht SA, Castro RE, Rodrigues CM, Moreira R, et al. Synthesis, G-quadruplexstabilisation, docking studies, and effect on cancer cells of indolo[3,2-b]quinolines with one, two, or three basic side chains. Chem Med Chem 2013;8:1648–1661.
44.
go back to reference Brito H, Martins AC, Lavrado J, Mendes E, Francisco AP, Santos SA, et al. Targeting KRAS oncogene in colon cancer cells with 7-carboxylate Indolo[3,2-b]quinoline tri-Alkylamine derivatives. PLoS One. 2015;10:e0126891.CrossRefPubMedPubMedCentral Brito H, Martins AC, Lavrado J, Mendes E, Francisco AP, Santos SA, et al. Targeting KRAS oncogene in colon cancer cells with 7-carboxylate Indolo[3,2-b]quinoline tri-Alkylamine derivatives. PLoS One. 2015;10:e0126891.CrossRefPubMedPubMedCentral
45.
go back to reference Leonetti C, Scarsella M, Riggio G, Rizzo A, Salvati E, D’Incalci M, et al. G-quadruplex ligand RHPS4 potentiates the antitumor activity of camptothecins in preclinical models of solid tumors. Clin Cancer Res. 2008;14:7284–91.CrossRefPubMed Leonetti C, Scarsella M, Riggio G, Rizzo A, Salvati E, D’Incalci M, et al. G-quadruplex ligand RHPS4 potentiates the antitumor activity of camptothecins in preclinical models of solid tumors. Clin Cancer Res. 2008;14:7284–91.CrossRefPubMed
46.
go back to reference Salvati E, Scarsella M, Porru M, Rizzo A, Iachettini S, Tentori L, et al. PARP1 is activated at telomeres upon G4 stabilization: possible target for telomere-based therapy. Oncogene. 2010;29:6280–93.CrossRefPubMed Salvati E, Scarsella M, Porru M, Rizzo A, Iachettini S, Tentori L, et al. PARP1 is activated at telomeres upon G4 stabilization: possible target for telomere-based therapy. Oncogene. 2010;29:6280–93.CrossRefPubMed
47.
go back to reference Biroccio A, Porru M, Rizzo A, Salvati E, D’Angelo C, Orlandi A, et al. DNA damage persistence as determinant of tumor sensitivity to the combination of topo I inhibitors and telomere-targeting agents. Clin Cancer Res. 2011;17:2227–36.CrossRefPubMed Biroccio A, Porru M, Rizzo A, Salvati E, D’Angelo C, Orlandi A, et al. DNA damage persistence as determinant of tumor sensitivity to the combination of topo I inhibitors and telomere-targeting agents. Clin Cancer Res. 2011;17:2227–36.CrossRefPubMed
48.
go back to reference Franceschin M, Rizzo A, Casagrande V, Salvati E, Alvino A, Altieri A, et al. Aromatic core extension in the series of N-cyclic bay-substituted perylene G-quadruplex ligands: increased telomere damage, antitumor activity, and strong selectivity for neoplastic over healthy cells. ChemMedChem. 2012;7:2144–54.CrossRefPubMed Franceschin M, Rizzo A, Casagrande V, Salvati E, Alvino A, Altieri A, et al. Aromatic core extension in the series of N-cyclic bay-substituted perylene G-quadruplex ligands: increased telomere damage, antitumor activity, and strong selectivity for neoplastic over healthy cells. ChemMedChem. 2012;7:2144–54.CrossRefPubMed
49.
go back to reference Porru M, Zizza P, Franceschin M, Leonetti C, Biroccio A. EMICORON: A multi-targeting G4 ligand with a promising preclinical profile. Biochim Biophys Acta. 2017;1861(5 Pt B):1362–70.CrossRefPubMed Porru M, Zizza P, Franceschin M, Leonetti C, Biroccio A. EMICORON: A multi-targeting G4 ligand with a promising preclinical profile. Biochim Biophys Acta. 2017;1861(5 Pt B):1362–70.CrossRefPubMed
50.
go back to reference Porru M, Artuso S, Salvati E, Bianco A, Franceschin M, Diodoro MG, et al. Targeting G-quadruplex DNA structures by EMICORON has a strong antitumor efficacy against advanced models of human colon cancer. Mol Cancer Ther. 2015;14:2541–51.CrossRefPubMed Porru M, Artuso S, Salvati E, Bianco A, Franceschin M, Diodoro MG, et al. Targeting G-quadruplex DNA structures by EMICORON has a strong antitumor efficacy against advanced models of human colon cancer. Mol Cancer Ther. 2015;14:2541–51.CrossRefPubMed
51.
go back to reference Pompili L, Porru M, Caruso C, Biroccio A, Leonetti C. Patient-derived xenografts: a relevant preclinical model for drug development. J Exp Clin Cancer Res. 2016;35:189.CrossRefPubMedPubMedCentral Pompili L, Porru M, Caruso C, Biroccio A, Leonetti C. Patient-derived xenografts: a relevant preclinical model for drug development. J Exp Clin Cancer Res. 2016;35:189.CrossRefPubMedPubMedCentral
52.
54.
go back to reference Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002;99:15524–9.CrossRefPubMedPubMedCentral Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002;99:15524–9.CrossRefPubMedPubMedCentral
55.
go back to reference Zhang GJ, Li JS, Zhou H, Xiao HX, Li Y, Zhou T. MicroRNA-106b promotes colorectal cancer cell migration and invasion by directly targeting DLC1. J Exp Clin Cancer Res. 2015;30:34–73. Zhang GJ, Li JS, Zhou H, Xiao HX, Li Y, Zhou T. MicroRNA-106b promotes colorectal cancer cell migration and invasion by directly targeting DLC1. J Exp Clin Cancer Res. 2015;30:34–73.
56.
go back to reference Chi Y, Zhou D. MicroRNAs in colorectal carcinoma from pathogenesis to therapy. J Exp Clin Cancer Res. 2016;10:35–43. Chi Y, Zhou D. MicroRNAs in colorectal carcinoma from pathogenesis to therapy. J Exp Clin Cancer Res. 2016;10:35–43.
57.
go back to reference Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, et al. RAS is regulated by the Let-7 MicroRNA family. Cell. 2005;120:635–47.CrossRefPubMed Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, et al. RAS is regulated by the Let-7 MicroRNA family. Cell. 2005;120:635–47.CrossRefPubMed
58.
go back to reference Boyerinas B, Park SM, Hau A, Murmann AE, Peter ME. The role of let-7 in cell differentiation and cancer. Endocr Relat Cancer. 2010;17:F19–36.CrossRefPubMed Boyerinas B, Park SM, Hau A, Murmann AE, Peter ME. The role of let-7 in cell differentiation and cancer. Endocr Relat Cancer. 2010;17:F19–36.CrossRefPubMed
60.
go back to reference Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko D, et al. The let-7 MicroRNA represses cell proliferation pathways in human cells. Cancer Res. 2007;67(16):7713–22.CrossRefPubMed Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko D, et al. The let-7 MicroRNA represses cell proliferation pathways in human cells. Cancer Res. 2007;67(16):7713–22.CrossRefPubMed
61.
go back to reference Rachagani S, Macha MA, Menning MS, Dey P, Pai P, Smith LM, et al. Changes in microRNA (miRNA) expression during pancreatic cancer development and progression in a genetically engineered KrasG12D; Pdx1-Cre mouse (KC) model. Oncotarget. 2015;6:40295–309.CrossRefPubMedPubMedCentral Rachagani S, Macha MA, Menning MS, Dey P, Pai P, Smith LM, et al. Changes in microRNA (miRNA) expression during pancreatic cancer development and progression in a genetically engineered KrasG12D; Pdx1-Cre mouse (KC) model. Oncotarget. 2015;6:40295–309.CrossRefPubMedPubMedCentral
62.
go back to reference Hong Y, Liang H, Uzair-Ur-Rehman WY, Zhang W, Zhou Y, et al. miR-96 promotes cell proliferation, migration and invasion by targeting PTPN9 in breast cancer. Sci Rep. 2016;6:37421.CrossRefPubMedPubMedCentral Hong Y, Liang H, Uzair-Ur-Rehman WY, Zhang W, Zhou Y, et al. miR-96 promotes cell proliferation, migration and invasion by targeting PTPN9 in breast cancer. Sci Rep. 2016;6:37421.CrossRefPubMedPubMedCentral
63.
go back to reference Yu S, Lu Z, Liu C, Meng Y, Ma Y, Zhao W, et al. miRNA-96 suppresses KRAS and functions as a tumor suppressor gene in pancreatic cancer. Cancer Res. 2010;70:6015–25.CrossRefPubMed Yu S, Lu Z, Liu C, Meng Y, Ma Y, Zhao W, et al. miRNA-96 suppresses KRAS and functions as a tumor suppressor gene in pancreatic cancer. Cancer Res. 2010;70:6015–25.CrossRefPubMed
64.
go back to reference Shin KH, Bae SD, Hong HS, Kim RH, Kang MK, Park NH. miR-181a shows tumor suppressive effect against oral squamous cell carcinoma cells by downregulating K-ras. Biochem Biophys Res Commun. 2011;404:896–902.CrossRefPubMed Shin KH, Bae SD, Hong HS, Kim RH, Kang MK, Park NH. miR-181a shows tumor suppressive effect against oral squamous cell carcinoma cells by downregulating K-ras. Biochem Biophys Res Commun. 2011;404:896–902.CrossRefPubMed
65.
go back to reference Liao WT, Ye YP, Zhang NJ, Li TT, Wang SY, Cui YM, et al. MicroRNA-30b functions as a tumour suppressor in human colorectal cancer by targeting KRAS, PIK3CD and BCL2. J Pathol. 2014;232:415–27.CrossRefPubMed Liao WT, Ye YP, Zhang NJ, Li TT, Wang SY, Cui YM, et al. MicroRNA-30b functions as a tumour suppressor in human colorectal cancer by targeting KRAS, PIK3CD and BCL2. J Pathol. 2014;232:415–27.CrossRefPubMed
66.
go back to reference Chen X, Guo X, Zhang H, Xiang Y, Chen J, Yin Y, et al. Role of miR-143 targeting KRAS in colorectal tumorigenesis. Oncogene. 2009;28:1385–92.CrossRefPubMed Chen X, Guo X, Zhang H, Xiang Y, Chen J, Yin Y, et al. Role of miR-143 targeting KRAS in colorectal tumorigenesis. Oncogene. 2009;28:1385–92.CrossRefPubMed
67.
go back to reference Zhou Y, Dang J, Chang KY, Yau E, Aza-Blanc P, Moscat J, et al. miR-1298 inhibits mutant KRAS-driven tumor growth by repressing FAK and LAMB3. Cancer Res. 2016;76:5777–87.CrossRefPubMedPubMedCentral Zhou Y, Dang J, Chang KY, Yau E, Aza-Blanc P, Moscat J, et al. miR-1298 inhibits mutant KRAS-driven tumor growth by repressing FAK and LAMB3. Cancer Res. 2016;76:5777–87.CrossRefPubMedPubMedCentral
Metadata
Title
Targeting KRAS in metastatic colorectal cancer: current strategies and emerging opportunities
Authors
Manuela Porru
Luca Pompili
Carla Caruso
Annamaria Biroccio
Carlo Leonetti
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0719-1

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine