Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1337 word(s) 1337 2021-11-29 07:27:27 |
2 The format is correct. -32 word(s) 1305 2021-11-29 08:46:02 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Sunaga, N. Targeting Oncogenic KRAS in Non-Small-Cell Lung Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/16484 (accessed on 26 April 2024).
Sunaga N. Targeting Oncogenic KRAS in Non-Small-Cell Lung Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/16484. Accessed April 26, 2024.
Sunaga, Noriaki. "Targeting Oncogenic KRAS in Non-Small-Cell Lung Cancer" Encyclopedia, https://encyclopedia.pub/entry/16484 (accessed April 26, 2024).
Sunaga, N. (2021, November 29). Targeting Oncogenic KRAS in Non-Small-Cell Lung Cancer. In Encyclopedia. https://encyclopedia.pub/entry/16484
Sunaga, Noriaki. "Targeting Oncogenic KRAS in Non-Small-Cell Lung Cancer." Encyclopedia. Web. 29 November, 2021.
Targeting Oncogenic KRAS in Non-Small-Cell Lung Cancer
Edit

v-Ki-ras2 Kirsten rat sarcoma viral oncogene (KRAS) is the most common driver in NSCLC, and targeting oncogenic KRAS is a major challenge in the treatment of non-small-cell lung cancer (NSCLC). The recent discovery of covalent KRAS G12C inhibitors offers hope for improving the prognosis of NSCLC patients, but the development of combination therapies corresponding to tumor characteristics is still required given the vast heterogeneity of KRAS-mutated NSCLC.

v-Ki-ras2 Kirsten rat sarcoma viral oncogene non-small-cell lung cancer covalent KRAS G12C inhibitor combination therapy

1. Introduction

Lung cancer is the leading cause of cancer-related death worldwide. Lung cancer is classified into two major histological subtypes: small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC), with the latter accounting for approximately 85% of all lung cancers. NSCLC consists mainly of lung adenocarcinomas (LUADs) and squamous cell lung carcinomas. The prognosis of lung cancer has been poor because the majority of lung cancer patients are initially diagnosed at an advanced stage. While recent developments in molecularly targeted drugs and immune checkpoint inhibitors have prolonged the survival of patients with advanced NSCLC [1][2], v-Ki-ras2 Kirsten rat sarcoma viral oncogene (KRAS), which is one of the most common oncogenes in NSCLC, had long been an “undruggable target” despite extensive efforts. RAS proteins are small guanosine triphosphatase (GTP)-binding proteins. The RAS family consists of three members, HRAS, NRAS and KRAS. KRAS knockout in mice is lethal, but HRAS or NRAS knockout mice are viable and develop normally [3][4], and KRAS can be replaced by HRAS during mouse embryogenesis [5]. Upon activation of receptor tyrosine kinases (RTKs), RAS is activated by SHP2 in cooperation with the adaptor protein GRB2 and SOS, which is a guanine nucleotide exchange factor (GEF) that catalyzes the transition of the guanosine diphosphate (GDP)-bound inactive form of RAS to the GTP-bound active form [6][7]. When KRAS is mutated, RAS is locked into the GTP-bound active form, which in turn constitutively activates downstream signaling pathways, such as the RAF-MEK-ERK and PI3K-AKT-mTOR pathways, conferring malignant phenotypes [8][9]. Recently, several covalent KRAS G12C inhibitors, including AMG510 (sotorasib) and MRTX849 (adagrasib), have been developed for KRAS G12C-mutant tumors [10][11][12]. Of note, the CodeBreaK100 clinical trial showed a beneficial effect of sotorasib in patients with advanced NSCLC harboring KRAS G12C mutations [13], and the FDA approved sotorasib for KRAS G12C-positive NSCLC patients who had received at least one prior systemic therapy in May 2021. On the other hand, KRAS G12C inhibitors do not provide as durable responses as tyrosine kinase inhibitors against EGFR or ALK; the median progression-free survival times of NSCLC patients treated with sotorasib and the EGFR-tyrosine kinase inhibitor (EGFR-TKI) osimertinib were 6.8 months [13] and 18.9 months [14], respectively. Furthermore, several secondary KRAS mutations and other molecular abnormalities causing resistance to covalent KRAS G12C-specific inhibitors have been reported [15][16][17]. The precise resistance mechanisms and therapeutic strategies for KRAS G12C inhibitor-resistant tumors are under investigation [18].

2. Multiple Faces of KRAS-Mutated NSCLC

The biological and clinical significance of KRAS mutations varies depending on the mutation subtype in NSCLC. For instance, G12C and G12D have higher affinities for binding to RALGDS and PI3K, respectively [19]. A recent study assessing the molecular profiles of NSCLC specimens revealed that tumors harboring KRAS G12C mutations exhibited a higher programmed death-ligand 1 (PD-L1) tumor proportion score and tumor mutational burden than those with KRAS non-G12C mutations [20]. From a clinical point of view, NSCLC patients harboring KRAS G12C or G12V mutations have a worse prognosis than those without these mutation subtypes [19]. In another study, surgically resected LUAD patients with KRAS G12C mutations had significantly shorter survival times than those with KRAS non-G12C mutations [21]. Therefore, targeting KRAS G12C seems to be an attractive therapeutic approach to improve the prognosis of NSCLC patients. In contrast, no significant difference in the overall survival of patients with metastatic NSCLC was found among different subtypes of KRAS mutations [22][23]. These inconsistent observations are likely due to the intratumoral heterogeneity of KRAS-mutated tumors [24][25], in addition to biological and immunological differences in KRAS mutation subtypes [19][26].

3. Oncogenic KRAS Regulates the Tumor Microenvironment (TME)

There is growing evidence that oncogenic KRAS is involved in tumor immune evasion by regulating the TME [27]KRAS-mutated NSCLC tumors have inflammatory characteristics; oncogenic KRAS induces several inflammatory cytokines and chemokines, including IL-6, IL-8, CXCL1 and CCL5, which influence the TME [28]. For instance, oncogenic KRAS mediates the secretion of IL-6, which drives protumor M2-type macrophage polarization along with the recruitment of myeloid-derived suppressor cells (MDSCs) in lung tumors [29]. Oncogenic KRAS also upregulates the expression of IL-8, a neutrophil chemoattractant, through MEK-ERK pathway activation in NSCLC cells [30]. Elevated serum IL-8 levels were found to be associated with an unfavorable prognosis in NSCLC patients treated with the PD-l antibody nivolumab, and increased IL-8 expression in tumors was negatively correlated with T cell markers and IFNγ-dependent gene signatures in the TME [31]. Another neutrophil chemoattractant, CXCL1, is also upregulated by KRAS mutations, and is involved in the development of lung cancer [32][33][34]. Moreover, KRAS-mutated NSCLC cells produce CCL5 [35], which plays an important role in antitumor immunity by promoting the recruitment of T cells and dendritic cells to the TME [36].
Several studies have demonstrated that KRAS-mutated NSCLC tumors exhibit PD-L1 overexpression [37], which is induced by oncogenic RAS-related pathway activation [38][39][40][41]. A previous study showed that B7-H3, T-cell immunoglobulin mucin family member 3, and indoleamine 2,3-dioxygenase-1 were highly expressed in tumor stroma-associated inflammatory cells in LUADs harboring KRAS mutations [42]. In addition to immune checkpoint molecules, regulatory T cells (Tregs) play a pivotal role in tumor immune evasion [43]. Mutant KRAS induces Tregs via secretion of IL-10 and transforming growth factor-β1 from tumor cells [44]. Le et al. found CD73 overexpression in EGFR-mutated NSCLC tumors [45]. They further demonstrated that the proportion of Tregs was decreased by coculturing these cells with conditioned medium from EGFR-mutated NSCLC cells with CD73 knockdown, and that an anti-CD73 antibody suppressed tumor growth in immunocompetent mice [45]. These observations suggest that KRAS-mutated NSCLC tumors have the ability to evade immune responses by regulating inflammatory cytokines and chemokines, immune checkpoint molecules and Tregs. Thus, targeting mutant KRAS may be an optional approach to abolish tumor immune evasion, and combination treatments including KRAS G12C inhibitors and immune checkpoint inhibitors could be effective therapeutic strategies.

4. Covalent KRAS G12C Inhibitors for KRAS-Mutated NSCLC

The recent discovery of covalent KRAS G12C inhibitors has changed “undruggable” oncogenic KRAS into a “druggable” target. In the KRAS G12C inactive (GDP-bound) form, the mutant cysteine residues adjacent to the switch II pocket are involved in the effector interaction [46]. The binding of covalent KRAS G12C inhibitors, such as sotorasib and adagrasib, to the switch pocket changes the nucleotide preference to favor GDP, thus impairing oncogenic KRAS-mediated signal transduction and leading to tumor regression in preclinical models [10][11]

5. Combined Therapies Involving Targeting of Oncogenic KRAS plus Other Targeted Drugs for KRAS-Mutated NSCLC

Oncogenic RAS upregulates downstream pathways and negatively regulates RTK signaling, whereas attenuating oncogenic RAS results in compensatory activation of RTK and wild-type RAS signaling pathways [47]. Thus, mutant RAS and wild-type RAS appear to complement each other to activate RTK-RAS signaling pathways to sustain tumor growth and survival, and the multitargeting of RAS signaling pathways could be an effective therapeutic approach. Researchers reported that the dual inhibition of KRAS and p38 inhibited tumor growth in KRAS-mutated colorectal cancer cells [48] and KRAS-mutated LUAD cells [49]. These findings suggest that blocking p38 or EGFR enhances the growth-inhibitory effect of KRAS G12C inhibitors on NSCLC tumors. In another study by Kitani et al., effective combination strategies differed between epithelial-like and mesenchymal-like KRAS-mutant NSCLC cells; a combination therapy with the MEK inhibitor trametinib plus the EGFR-TKI afatinib was effective for epithelial-like tumors, while the dual blockade of MEK and FGFR effectively suppressed mesenchymal tumor growth [50]. A recent study demonstrated that combined treatment with the KRAS G12C inhibitor ARS-1620 plus the pan-PI3K inhibitor GDC0941 consistently suppressed tumor growth in ARS-1620-resistant KRAS-mutated tumors, whereas the growth-inhibitory effects of ARS-1620 plus afatinib or cetuximab varied among KRAS-mutated cell lines [51]. Together, these observations indicate that the therapeutic efficacy of covalent KRAS G12C inhibitors varies depending on the biological properties of the treated KRAS-mutated tumors.

References

  1. Herbst, R.S.; Garon, E.B.; Kim, D.W.; Cho, B.C.; Gervais, R.; Perez-Gracia, J.L.; Han, J.Y.; Majem, M.; Forster, M.D.; Monnet, I.; et al. Five Year Survival Update From KEYNOTE-010: Pembrolizumab Versus Docetaxel for Previously Treated, Programmed Death-Ligand 1-Positive Advanced NSCLC. J. Thorac. Oncol. 2021, 16, 1718–1732.
  2. Okamoto, I.; Morita, S.; Tashiro, N.; Imamura, F.; Inoue, A.; Seto, T.; Yamamoto, N.; Ohe, Y.; Nakagawa, K.; Fukuoka, M. Real world treatment and outcomes in EGFR mutation-positive non-small cell lung cancer: Long-term follow-up of a large patient cohort. Lung Cancer 2018, 117, 14–19.
  3. Esteban, L.M.; Vicario-Abejon, C.; Fernandez-Salguero, P.; Fernandez-Medarde, A.; Swaminathan, N.; Yienger, K.; Lopez, E.; Malumbres, M.; McKay, R.; Ward, J.M.; et al. Targeted genomic disruption of H-ras and N-ras, individually or in combination, reveals the dispensability of both loci for mouse growth and development. Mol. Cell Biol. 2001, 21, 1444–1452.
  4. Koera, K.; Nakamura, K.; Nakao, K.; Miyoshi, J.; Toyoshima, K.; Hatta, T.; Otani, H.; Aiba, A.; Katsuki, M. K-ras is essential for the development of the mouse embryo. Oncogene 1997, 15, 1151–1159.
  5. Potenza, N.; Vecchione, C.; Notte, A.; De Rienzo, A.; Rosica, A.; Bauer, L.; Affuso, A.; De Felice, M.; Russo, T.; Poulet, R.; et al. Replacement of K-Ras with H-Ras supports normal embryonic development despite inducing cardiovascular pathology in adult mice. EMBO Rep. 2005, 6, 432–437.
  6. Dance, M.; Montagner, A.; Salles, J.P.; Yart, A.; Raynal, P. The molecular functions of Shp2 in the Ras/Mitogen-activated protein kinase (ERK1/2) pathway. Cell Signal. 2008, 20, 453–459.
  7. Ran, H.; Tsutsumi, R.; Araki, T.; Neel, B.G. Sticking It to Cancer with Molecular Glue for SHP2. Cancer Cell 2016, 30, 194–196.
  8. Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat. Rev. Cancer 2003, 3, 11–22.
  9. Suda, K.; Tomizawa, K.; Mitsudomi, T. Biological and clinical significance of KRAS mutations in lung cancer: An oncogenic driver that contrasts with EGFR mutation. Cancer Metastasis Rev. 2010, 29, 49–60.
  10. Canon, J.; Rex, K.; Saiki, A.Y.; Mohr, C.; Cooke, K.; Bagal, D.; Gaida, K.; Holt, T.; Knutson, C.G.; Koppada, N.; et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 2019, 575, 217–223.
  11. Hallin, J.; Engstrom, L.D.; Hargis, L.; Calinisan, A.; Aranda, R.; Briere, D.M.; Sudhakar, N.; Bowcut, V.; Baer, B.R.; Ballard, J.A.; 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.
  12. Janes, M.R.; Zhang, J.; Li, L.S.; Hansen, R.; Peters, U.; Guo, X.; Chen, Y.; Babbar, A.; Firdaus, S.J.; Darjania, L.; et al. Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell 2018, 172, 578–589.
  13. Skoulidis, F.; Li, B.T.; Dy, G.K.; Price, T.J.; Falchook, G.S.; Wolf, J.; Italiano, A.; Schuler, M.; Borghaei, H.; Barlesi, F.; et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N. Engl. J. Med. 2021, 384, 2371–2381.
  14. Soria, J.C.; Ohe, Y.; Vansteenkiste, J.; Reungwetwattana, T.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Imamura, F.; Nogami, N.; Kurata, T.; et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 113–125.
  15. Koga, T.; Suda, K.; Fujino, T.; Ohara, S.; Hamada, A.; Nishino, M.; Chiba, M.; Shimoji, M.; Takemoto, T.; Arita, T.; et al. KRAS Secondary Mutations That Confer Acquired Resistance to KRAS G12C Inhibitors, Sotorasib and Adagrasib, and Overcoming Strategies: Insights From In Vitro Experiments. J. Thorac. Oncol. 2021, 16, 1321–1332.
  16. Awad, M.M.; Liu, S.; Rybkin, I.I.; Arbour, K.C.; Dilly, J.; Zhu, V.W.; Johnson, M.L.; Heist, R.S.; Patil, T.; Riely, G.J.; et al. Acquired Resistance to KRAS(G12C) Inhibition in Cancer. N. Engl. J. Med. 2021, 384, 2382–2393.
  17. Tanaka, N.; Lin, J.J.; Li, C.; Ryan, M.B.; Zhang, J.; Kiedrowski, L.A.; Michel, A.G.; Syed, M.U.; Fella, K.A.; Sakhi, M.; 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, 1913–1922.
  18. Addeo, A.; Banna, G.L.; Friedlaender, A. KRAS G12C Mutations in NSCLC: From Target to Resistance. Cancers 2021, 13, 2541.
  19. Ihle, N.T.; Byers, L.A.; Kim, E.S.; Saintigny, P.; Lee, J.J.; Blumenschein, G.R.; Tsao, A.; Liu, S.; Larsen, J.E.; Wang, J.; et al. Effect of KRAS oncogene substitutions on protein behavior: Implications for signaling and clinical outcome. J. Natl. Cancer Inst. 2012, 104, 228–239.
  20. Judd, J.; Abdel Karim, N.; Khan, H.; Naqash, A.R.; Baca, Y.; Xiu, J.; VanderWalde, A.M.; Mamdani, H.; Raez, L.E.; Nagasaka, M.; et al. Characterization of KRAS Mutation Subtypes in Non-small Cell Lung Cancer. Mol. Cancer Ther. 2021. Epub ahead of print.
  21. Nadal, E.; Chen, G.; Prensner, J.R.; Shiratsuchi, H.; Sam, C.; Zhao, L.; Kalemkerian, G.P.; Brenner, D.; Lin, J.; Reddy, R.M.; et al. KRAS-G12C mutation is associated with poor outcome in surgically resected lung adenocarcinoma. J. Thorac. Oncol. 2014, 9, 1513–1522.
  22. Yu, H.A.; Sima, C.S.; Shen, R.; Kass, S.; Gainor, J.; Shaw, A.; Hames, M.; Iams, W.; Aston, J.; Lovly, C.M.; et al. Prognostic impact of KRAS mutation subtypes in 677 patients with metastatic lung adenocarcinomas. J. Thorac. Oncol. 2015, 10, 431–437.
  23. Scheffler, M.; Ihle, M.A.; Hein, R.; Merkelbach-Bruse, S.; Scheel, A.H.; Siemanowski, J.; Bragelmann, J.; Kron, A.; Abedpour, N.; Ueckeroth, F.; et al. K-ras Mutation Subtypes in NSCLC and Associated Co-occuring Mutations in Other Oncogenic Pathways. J. Thorac. Oncol. 2019, 14, 606–616.
  24. Chung, W.J.; Daemen, A.; Cheng, J.H.; Long, J.E.; Cooper, J.E.; Wang, B.E.; Tran, C.; Singh, M.; Gnad, F.; Modrusan, Z.; et al. Kras mutant genetically engineered mouse models of human cancers are genomically heterogeneous. Proc. Natl. Acad. Sci. USA 2017, 114, E10947–E10955.
  25. Kordiak, J.; Szemraj, J.; Grabska-Kobylecka, I.; Bialasiewicz, P.; Braun, M.; Kordek, R.; Nowak, D. Intratumor heterogeneity and tissue distribution of KRAS mutation in non-small cell lung cancer: Implications for detection of mutated KRAS oncogene in exhaled breath condensate. J. Cancer Res. Clin. Oncol. 2019, 145, 241–251.
  26. Hunter, J.C.; Manandhar, A.; Carrasco, M.A.; Gurbani, D.; Gondi, S.; Westover, K.D. Biochemical and Structural Analysis of Common Cancer-Associated KRAS Mutations. Mol. Cancer Res. 2015, 13, 1325–1335.
  27. Miura, Y.; Sunaga, N. Role of Immunotherapy for Oncogene-Driven Non-Small Cell Lung Cancer. Cancers 2018, 10, 245.
  28. Hamarsheh, S.; Gross, O.; Brummer, T.; Zeiser, R. Immune modulatory effects of oncogenic KRAS in cancer. Nat. Commun. 2020, 11, 5439.
  29. Caetano, M.S.; Zhang, H.; Cumpian, A.M.; Gong, L.; Unver, N.; Ostrin, E.J.; Daliri, S.; Chang, S.H.; Ochoa, C.E.; Hanash, S.; et al. IL6 Blockade Reprograms the Lung Tumor Microenvironment to Limit the Development and Progression of K-ras-Mutant Lung Cancer. Cancer Res. 2016, 76, 3189–3199.
  30. Sunaga, N.; Imai, H.; Shimizu, K.; Shames, D.S.; Kakegawa, S.; Girard, L.; Sato, M.; Kaira, K.; Ishizuka, T.; Gazdar, A.F.; et al. Oncogenic KRAS-induced interleukin-8 overexpression promotes cell growth and migration and contributes to aggressive phenotypes of non-small cell lung cancer. Int. J. Cancer. 2012, 130, 1733–1744.
  31. Schalper, K.A.; Carleton, M.; Zhou, M.; Chen, T.; Feng, Y.; Huang, S.P.; Walsh, A.M.; Baxi, V.; Pandya, D.; Baradet, T.; et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat. Med. 2020, 26, 688–692.
  32. Zhong, L.; Roybal, J.; Chaerkady, R.; Zhang, W.; Choi, K.; Alvarez, C.A.; Tran, H.; Creighton, C.J.; Yan, S.; Strieter, R.M.; et al. Identification of secreted proteins that mediate cell-cell interactions in an in vitro model of the lung cancer microenvironment. Cancer Res. 2008, 68, 7237–7245.
  33. Marazioti, A.; Lilis, I.; Vreka, M.; Apostolopoulou, H.; Kalogeropoulou, A.; Giopanou, I.; Giotopoulou, G.A.; Krontira, A.C.; Iliopoulou, M.; Kanellakis, N.I.; et al. Myeloid-derived interleukin-1beta drives oncogenic KRAS-NF-kappaBeta addiction in malignant pleural effusion. Nat. Commun. 2018, 9, 672.
  34. Liclican, E.L.; Walser, T.C.; Hazra, S.; Krysan, K.; Park, S.J.; Pagano, P.C.; Gardner, B.K.; Larsen, J.E.; Minna, J.D.; Dubinett, S.M. Loss of miR125a expression in a model of K-ras-dependent pulmonary premalignancy. Cancer Prev. Res. 2014, 7, 845–855.
  35. Zhu, Z.; Aref, A.R.; Cohoon, T.J.; Barbie, T.U.; Imamura, Y.; Yang, S.; Moody, S.E.; Shen, R.R.; Schinzel, A.C.; Thai, T.C.; et al. Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit. Cancer Discov. 2014, 4, 452–465.
  36. Aldinucci, D.; Borghese, C.; Casagrande, N. The CCL5/CCR5 Axis in Cancer Progression. Cancers 2020, 12, 1765.
  37. Lan, B.; Ma, C.; Zhang, C.; Chai, S.; Wang, P.; Ding, L.; Wang, K. Association between PD-L1 expression and driver gene status in non-small-cell lung cancer: A meta-analysis. Oncotarget 2018, 9, 7684–7699.
  38. Miura, Y.; Sunaga, N.; Kyoichi, K.; Tsukagoshi, Y.; Osaki, T.; Sakurai, R.; Hisada, T.; Girard, L.; Minna, J.D. Oncogenic KRAS mutations induce PD-L1 overexpression through MAPK pathway activation in non-small cell lung cancer cells. Cancer Res. 2016, 76, Abstract nr 4028.
  39. Sumimoto, H.; Takano, A.; Teramoto, K.; Daigo, Y. RAS-Mitogen-Activated Protein Kinase Signal Is Required for Enhanced PD-L1 Expression in Human Lung Cancers. PLoS ONE 2016, 11, e0166626.
  40. Coelho, M.A.; de Carne Trecesson, S.; Rana, S.; Zecchin, D.; Moore, C.; Molina-Arcas, M.; East, P.; Spencer-Dene, B.; Nye, E.; Barnouin, K.; et al. Oncogenic RAS Signaling Promotes Tumor Immunoresistance by Stabilizing PD-L1 mRNA. Immunity 2017, 47, 1083–1099.e6.
  41. Lastwika, K.J.; Wilson, W., 3rd; Li, Q.K.; Norris, J.; Xu, H.; Ghazarian, S.R.; Kitagawa, H.; Kawabata, S.; Taube, J.M.; Yao, S.; et al. Control of PD-L1 Expression by Oncogenic Activation of the AKT-mTOR Pathway in Non-Small Cell Lung Cancer. Cancer Res. 2016, 76, 227–238.
  42. Parra, E.R.; Villalobos, P.; Zhang, J.; Behrens, C.; Mino, B.; Swisher, S.; Sepesi, B.; Weissferdt, A.; Kalhor, N.; Heymach, J.V.; et al. Immunohistochemical and Image Analysis-Based Study Shows that Several Immune Checkpoints are Co-expressed in Non-Small Cell Lung Carcinoma Tumors. J. Thorac. Oncol. 2018, 13, 779–791.
  43. Li, C.; Jiang, P.; Wei, S.; Xu, X.; Wang, J. Regulatory T cells in tumor microenvironment: New mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer. 2020, 19, 116.
  44. Zdanov, S.; Mandapathil, M.; Abu Eid, R.; Adamson-Fadeyi, S.; Wilson, W.; Qian, J.; Carnie, A.; Tarasova, N.; Mkrtichyan, M.; Berzofsky, J.A.; et al. Mutant KRAS Conversion of Conventional T Cells into Regulatory T Cells. Cancer Immunol. Res. 2016, 4, 354–365.
  45. Le, X.; Negrao, M.V.; Reuben, A.; Federico, L.; Diao, L.; McGrail, D.; Nilsson, M.; Robichaux, J.; Munoz, I.G.; Patel, S.; et al. Characterization of the Immune Landscape of EGFR-Mutant NSCLC Identifies CD73/Adenosine Pathway as a Potential Therapeutic Target. J. Thorac. Oncol. 2021, 16, 583–600.
  46. Ostrem, J.M.; Peters, U.; Sos, M.L.; Wells, J.A.; Shokat, K.M. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 2013, 503, 548–551.
  47. Young, A.; Lou, D.; McCormick, F. Oncogenic and wild-type Ras play divergent roles in the regulation of mitogen-activated protein kinase signaling. Cancer Discov. 2013, 3, 112–123.
  48. Kamran, S.; Seyedrezazadeh, E.; Shanehbandi, D.; Asadi, M.; Zafari, V.; Shekari, N.; Namvar, L.; Zarredar, H. Combination Therapy with KRAS and P38alpha siRNA Suppresses Colorectal Cancer Growth and Development in SW480 Cell Line. J. Gastrointest. Cancer. 2021. Epub ahead of print.
  49. Zarredar, H.; Pashapour, S.; Farajnia, S.; Ansarin, K.; Baradaran, B.; Ahmadzadeh, V.; Safari, F. Targeting the KRAS, p38alpha, and NF-kappaB in lung adenocarcinoma cancer cells: The effect of combining RNA interferences with a chemical inhibitor. J. Cell Biochem. 2019, 120, 10670–10677.
  50. Kitai, H.; Ebi, H.; Tomida, S.; Floros, K.V.; Kotani, H.; Adachi, Y.; Oizumi, S.; Nishimura, M.; Faber, A.C.; Yano, S. Epithelial-to-Mesenchymal Transition Defines Feedback Activation of Receptor Tyrosine Kinase Signaling Induced by MEK Inhibition in KRAS-Mutant Lung Cancer. Cancer Discov. 2016, 6, 754–769.
  51. Misale, S.; Fatherree, J.P.; Cortez, E.; Li, C.; Bilton, S.; Timonina, D.; Myers, D.T.; Lee, D.; Gomez-Caraballo, M.; Greenberg, M.; et al. KRAS G12C NSCLC Models Are Sensitive to Direct Targeting of KRAS in Combination with PI3K Inhibition. Clin. Cancer Res. 2019, 25, 796–807.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 344
Revisions: 2 times (View History)
Update Date: 29 Nov 2021
1000/1000