The Future Prospects of the KRAS Gene and Its Association with Cancer as Predictive Markers
Keywords:
genes, KRAS, immune evasion, cancer, mutationsAbstract
The importance of early cancer detection has grown significantly in recent years due to the widespread prevalence of this disease across various age groups. This article aims to highlight the crucial role of strategies and methods used in the early detection or prediction of cancer, given their vital impact on saving lives and determining appropriate treatment approaches. The KRAS gene has gained prominence in recent years for its role in predicting tumor development before it occurs, as it is one of the most frequently mutated genes in many human cancers. This gene is involved in transmitting cellular signals and regulating the cell cycle, primarily through expressing mutated protein products or altering cellular pathways. Among the most common and frequent KRAS mutations are (G12, G12C, Q61, and G12V), which are observed in cancers such as pancreatic, colorectal, and lung tumors, accounting for approximately 20% of all human cancers. KRAS gene mutations contribute to reinforcing the characteristics of mutated cells and preparing the tumor microenvironment to support their persistence. Several mechanisms by which these mutations evade immune surveillance and distract immune cells from the tumor itself have been identified and studied. In recent years, numerous potential treatment approaches have emerged. Some genetic and molecular therapies have already been approved, while others are in clinical trial stages The emergence of targeted therapies that address specific metabolic pathways of cancer or directly target compounds and molecules involved in disease progression has marked the rise of diagnostic or precision medicine. Various other approaches and strategies are still under investigation, aiming to achieve a comprehensive cure for cancer.
References
[1] L. Moss, What Genes Can't Do. Cambridge, MA, USA: MIT Press, 2003.
[2] M. B. Gerstein, C. Bruce, J. S. Rozowsky, D. Zheng, J. Du, J. O. Korbel, et al., “What is a gene, post-ENCODE? History and updated definition,” Genome Research, vol. 17, no. 6, pp. 669–681, 2007.
[3] M. Claussnitzer, J. H. Cho, R. Collins, N. J. Cox, E. T. Dermitzakis, M. E. Hurles, et al., “A brief history of human disease genetics,” Nature, vol. 577, no. 7789, pp. 179–189, 2020.
[4] N. Rahman, “Realizing the promise of cancer predisposition genes,” Nature, vol. 505, no. 7483, pp. 302–308, 2014.
[5] N. Ye and J. Zhou, “KRAS – An evolving cancer target,” Austin Journal of Cancer and Clinical Research, vol. 1, no. 1, 2014.
[6] S. Jančík, J. Drábek, D. Radzioch, and M. Hajdúch, “Clinical relevance of KRAS in human cancers,” BioMed Research International, vol. 2010, Art. no. 150960, 2010.
[7] M. H. Hofmann, D. Gerlach, S. Misale, M. Petronczki, and N. Kraut, “Expanding the reach of precision oncology by drugging all KRAS mutants,” Cancer Discovery, vol. 12, no. 4, pp. 924–937, 2022.
[8] L. Huang, Z. Guo, F. Wang, and L. Fu, “KRAS mutation: From undruggable to druggable in cancer,” Signal Transduction and Targeted Therapy, vol. 6, no. 1, Art. no. 386, 2021.
[9] S. Rahman, S. Garrel, M. Gerber, R. Maitra, and S. Goel, “Therapeutic targets of KRAS in colorectal cancer,” Cancers, vol. 13, no. 24, Art. no. 6233, 2021.
[10] A. Singhal, B. T. Li, and E. M. O’Reilly, “Targeting KRAS in cancer,” Nature Medicine, pp. 1–15, 2024.
[11] H. Liu, Z. Liang, S. Cheng, L. Huang, W. Li, C. Zhou, et al., “Mutant KRAS drives immune evasion by sensitizing cytotoxic T‐cells to activation‐induced cell death in colorectal cancer,” Advanced Science, vol. 10, no. 6, Art. no. 2203757, 2023.
[12] É. O’Sullivan, A. Keogh, B. Henderson, S. P. Finn, S. G. Gray, and K. Gately, “Treatment strategies for KRAS-mutated non-small-cell lung cancer,” Cancers, vol. 15, no. 6, Art. no. 1635, 2023.
[13] F. McCormick, “Ras‐related proteins in signal transduction and growth control,” Molecular Reproduction and Development, vol. 42, no. 4, pp. 500–506, 1995.
[14] B. R. Voldborg, L. Damstrup, M. Spang-Thomsen, and H. S. Poulsen, “Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials,” Annals of Oncology, vol. 8, pp. 1197–1206, 1997.
[15] C. J. Tape, S. Ling, M. Dimitriadi, K. M. McMahon, J. D. Worboys, H. S. Leong, et al., “Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation,” Cell, vol. 165, no. 4, pp. 910–920, 2016.
[16] L. Huang, Z. Guo, F. Wang, and L. Fu, “KRAS mutation: From undruggable to druggable in cancer,” Signal Transduction and Targeted Therapy, vol. 6, no. 1, Art. no. 386, 2021.
[17] R. Chetty and D. Govender, “Gene of the month: KRAS,” Journal of Clinical Pathology, 2013.
[18] D. A. Fruman, et al., “The PI3K pathway in human disease,” Cell, vol. 170, pp. 605–635, 2017.
[19] J. A. Fresno Vara, et al., “PI3K/Akt signalling pathway and cancer,” Cancer Treatment Reviews, vol. 30, pp. 193–204, 2004.
[20] D. Vigil, J. Cherfils, K. L. Rossman, and C. J. Der, “Ras superfamily GEFs and GAP validated and tractable targets for cancer therapy?,” Nature Reviews Cancer, vol. 10, pp. 842–857, 2010.
[21] S. G. Rhee, “Regulation of phosphoinositide-specific phospholipase C,” Annual Review of Biochemistry, vol. 70, pp. 281–312, 2001.
[22] K. Wood, T. Hensing, R. Malik, and R. Salgia, “Prognostic and predictive value in KRAS in non-small-cell lung cancer: A review,” JAMA Oncology, vol. 2, pp. 805–812, 2016.
[23] H. Kodaz, O. Kostek, M. B. Hacioglu, B. Erdogan, C. E. Kodaz, I. Hacibekiroglu, et al., “Frequency of RAS mutations (KRAS, NRAS, HRAS) in human solid cancer,” Breast Cancer, vol. 7, no. 12, pp. 1–7, 2017.
[24] E. C. Stites, “The abundance of KRAS and RAS gene mutations in cancer,” in KRAS: Methods and Protocols, New York, NY, USA: Springer US, 2024, pp. 13–22.
[25] A. Singhal, B. T. Li, and E. M. O’Reilly, “Targeting KRAS in cancer,” Nature Medicine, pp. 1–15, 2024.
[26] K. Mondal, M. K. Posa, R. P. Shenoy, and S. Roychoudhury, “KRAS mutation subtypes and their association with other driver mutations in oncogenic pathways,” Cells, vol. 13, no. 14, 2024.
[27] T. Rendek, R. Saade, O. Pos, G. Kolnikova, M. Urbanova, J. Budis, et al., “Determination of the prevalence of microsatellite instability, BRAF and KRAS/NRAS mutation status in patients with colorectal cancer in Slovakia,” Cancers, vol. 16, no. 6, Art. no. 1128, 2024.
[28] S. Dearden, J. Stevens, Y. L. Wu, and D. Blowers, “Mutation incidence and coincidence in non-small-cell lung cancer: Meta-analyses by ethnicity and histology (mutMap),” Annals of Oncology, vol. 24, pp. 2371–2376, 2013.
[29] S. Jančík, J. Drábek, D. Radzioch, and M. Hajdúch, “Clinical relevance of KRAS in human cancers,” BioMed Research International, vol. 2010, Art. no. 150960, 2010.
[30] M. P. Zafra, et al., “An in vivo KRAS allelic series reveals distinct phenotypes of common oncogenic variants,” Cancer Discovery, vol. 10, pp. 1654–1671, 2020.
[31] N. T. Ihle, et al., “Effect of KRAS oncogene substitutions on protein behavior: Implications for signaling and clinical outcome,” Journal of the National Cancer Institute, vol. 104, pp. 228–239, 2012.
[32] C. Perrone, R. Angioli, D. Luvero, A. Giannini, V. Di Donato, I. Cuccu, et al., “Targeting BRAF pathway in low-grade serous ovarian cancer,” Journal of Gynecologic Oncology, vol. 35, no. 4, 2024.
[33] M. Scheffler, et al., “K-ras mutation subtypes in NSCLC and associated co-occurring mutations in other oncogenic pathways,” Journal of Thoracic Oncology, vol. 14, pp. 606–616, 2019.
[34] S. Zheng, et al., “Targeted next-generation sequencing for cancer-associated gene mutation and copy number detection in 206 patients with non-small-cell lung cancer,” Bioengineered, vol. 12, pp. 791–802, 2021.
[35] D. Reita, L. Pabst, E. Pencreach, E. Guérin, L. Dano, V. Rimelen, et al., “Direct targeting KRAS mutation in non-small cell lung cancer: Focus on resistance,” Cancers, vol. 14, no. 5, Art. no. 1321, 2022.
[36] B. Ricciuti, G. C. Leonardi, G. Metro, F. Grignani, L. Paglialunga, G. Bellezza, et al., “Targeting the KRAS variant for treatment of non-small cell lung cancer: Potential therapeutic applications,” Expert Review of Respiratory Medicine, vol. 10, no. 1, pp. 53–68, 2016.
[37] Y. Pylayeva-Gupta, E. Grabocka, and D. Bar-Sagi, “RAS oncogenes: Weaving a tumorigenic web,” Nature Reviews Cancer, vol. 11, pp. 761–774, 2011.
[38] P. Dias Carvalho, et al., “KRAS oncogenic signaling extends beyond cancer cells to orchestrate the microenvironment,” Cancer Research, vol. 78, pp. 7–14, 2018.
[39] K. Taniguchi and M. Karin, “IL-6 and related cytokines as the critical lynchpin between inflammation and cancer,” Seminars in Immunology, vol. 26, pp. 54–74, 2014.
[40] Y. Zhang, et al., “Interleukin-6 is required for pancreatic cancer progression by promoting MAPK signaling activation and oxidative stress resistance,” Cancer Research, vol. 73, pp. 6359–6374, 2013.
[41] Y. Zhou, M. Qian, J. Li, L. Ruan, Y. Wang, C. Cai, et al., “The role of tumor-associated macrophages in lung cancer: From mechanism to small molecule therapy,” Biomedicine & Pharmacotherapy, vol. 170, Art. no. 116014, 2024.
[42] M. Alanazi, T. Weng, L. J. McLeod, J. A. Smith, B. Kumar, and B. J. Jenkins, “Cytosolic DNA sensor AIM2 promotes KRAS‐driven lung cancer independent of inflammasomes,” Cancer Science, 2024.
[43] F. Pereira, A. Ferreira, C. A. Reis, M. J. Sousa, M. J. Oliveira, and A. Preto, “KRAS as a modulator of the inflammatory tumor microenvironment: Therapeutic implications,” Cells, vol. 11, no. 3, Art. no. 398, 2022.
[44] H. Ji, A. M. Houghton, T. J. Mariani, S. Perera, C. B. Kim, R. Padera, et al., “K-ras activation generates an inflammatory response in lung tumors,” Oncogene, vol. 25, no. 14, pp. 2105–2112, 2006.
[45] J. Ling, et al., “KrasG12D-induced IKK2/β/NF-κB activation by IL-1α and p62 feedforward loops is required for development of pancreatic ductal adenocarcinoma,” Cancer Cell, vol. 21, pp. 105–120, 2012.
[46] S. Casacuberta-Serra, Í. González-Larreategui, D. Capitán-Leo, and L. Soucek, “MYC and KRAS cooperation: From historical challenges to therapeutic opportunities in cancer,” Signal Transduction and Targeted Therapy, vol. 9, no. 1, Art. no. 205, 2024.
[47] J. Budczies, E. Romanovsky, M. Kirchner, O. Neumann, M. Blasi, J. Schnorbach, et al., “KRAS and TP53 co-mutation predicts benefit of immune checkpoint blockade in lung adenocarcinoma,” British Journal of Cancer, pp. 1–10, 2024.
[48] S. A. Hamarsheh, O. Groß, T. Brummer, and R. Zeiser, “Immune modulatory effects of oncogenic KRAS in cancer,” Nature Communications, vol. 11, Art. no. 5439, 2020.
[49] A. Watterson and M. A. Coelho, “Cancer immune evasion through KRAS and PD-L1 and potential therapeutic interventions,” Cell Communication and Signaling, vol. 21, no. 1, Art. no. 45, 2023.
[50] Y. Tokumaru, M. Oshi, E. Katsuta, L. Yan, V. Satyananda, N. Matsuhashi, and K. Takabe, “KRAS signaling enriched triple negative breast cancer is associated with favorable tumor immune microenvironment and better survival,” American Journal of Cancer Research, vol. 10, no. 3, pp. 897–907, 2020.
[51] J. Dubrot, S. K. Lane-Reticker, E. A. Kessler, A. J. Muscato, A. Mehta, and R. T. Manguso, “In vivo CRISPR screens reveal the landscape of immune evasion pathways across cancer,” Nature Immunology, vol. 23, no. 10, pp. 1495–1506, 2022.
[52] Y. Y. Lim, A. M. A. Zaidi, M. Haque, and A. Miskon, “Relationship between tumorigenesis, metastasis, immune evasion, and chemoresistance in osteosarcoma therapy,” Journal of Applied Pharmaceutical Science, vol. 14, no. 1, pp. 64–79, 2024.
[53] B. Seliger, “Strategies of tumor immune evasion,” BioDrugs, vol. 19, pp. 347–354, 2005.
[54] L. Gourmet, A. Sottoriva, S. Walker-Samuel, M. Secrier, and L. Zapata, “Immune evasion impacts the landscape of driver genes during cancer evolution,” Genome Biology, vol. 25, no. 1, Art. no. 168, 2024.
[55] F. van Maldegem and J. Downward, “Mutant KRAS at the heart of tumor immune evasion,” Immunity, vol. 52, pp. 14–16, 2020.
[56] A. Anichini, V. E. Perotti, F. Sgambelluri, and R. Mortarini, “Immune escape mechanisms in non small cell lung cancer,” Cancers, vol. 12, no. 12, Art. no. 3605, 2020.
[57] N. Chen, et al., “KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma,” Cancer Immunology, Immunotherapy, vol. 66, pp. 1175–1187, 2017.
[58] C. Testorelli, S. Bussini, R. De Filippi, O. Marelli, L. Orlando, J. W. Greiner, et al., “Dacarbazine-induced immunogenicity of a murine leukemia is attenuated in cells transfected with mutated K-ras gene,” Journal of Experimental & Clinical Cancer Research, vol. 16, no. 1, pp. 15–22, 1997.
[59] Z. Y. Dong, W. Z. Zhong, X. C. Zhang, J. Su, Z. Xie, S. Y. Liu, et al., “Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma,” Clinical Cancer Research, vol. 23, no. 12, pp. 3012–3024, 2017.
[60] N. Mundhara and P. Sadhukhan, “Cracking the codes behind cancer cells’ immune evasion,” International Journal of Molecular Sciences, vol. 25, no. 16, Art. no. 8899, 2024.
[61] T. Zaein, “The Role of Immune Cells in the Tumor Microenvironment,” Yashfeen Journal of Health Innovation and Practice, vol. 2, no. 01, pp. 158–172, 2023.
[62] J. Cullis, S. Das, and D. Bar-Sagi, “Kras and tumor immunity: friend or foe?,” Cold Spring Harbor Perspectives in Medicine, vol. 8, no. 9, Art. no. a031849, 2018.
[63] J. Boumelha, M. Molina-Arcas, and J. Downward, “Facts and hopes on RAS inhibitors and cancer immunotherapy,” Clinical Cancer Research, vol. 29, no. 24, pp. 5012–5020, 2023.
[64] B. Lv, Y. Wang, D. Ma, W. Cheng, J. Liu, T. Yong, et al., “Immunotherapy: reshape the tumor immune microenvironment,” Frontiers in Immunology, vol. 13, Art. no. 844142, 2022.
[65] Y. Xing, G. Ruan, H. Ni, H. Qin, S. Chen, X. Gu, et al., “Tumor immune microenvironment and its related miRNAs in tumor progression,” Frontiers in Immunology, vol. 12, Art. no. 624725, 2021.
[66] N. Mundhara and P. Sadhukhan, “Cracking the Codes behind Cancer Cells’ Immune Evasion,” International Journal of Molecular Sciences, vol. 25, no. 16, Art. no. 8899, 2024.
[67] P. K. Tripathi, K. R. Mittal, N. Jain, N. Sharma, and C. K. Jain, “KRAS pathways: a potential gateway for cancer therapeutics and diagnostics,” Recent Patents on Anti-Cancer Drug Discovery, vol. 19, no. 3, pp. 268–279, 2024.
[68] N. Lal, B. S. White, G. Goussous, O. Pickles, M. J. Mason, A. D. Beggs, and G. W. Middleton, “KRAS mutation and consensus molecular subtypes 2 and 3 are independently associated with reduced immune infiltration and reactivity in colorectal cancer,” Clinical Cancer Research, vol. 24, no. 1, pp. 224–233, 2018.
[69] G. P. Linette, A. S. Bear, and B. M. Carreno, “Facts and hopes in immunotherapy strategies targeting antigens derived from KRAS mutations,” Clinical Cancer Research, vol. 30, no. 10, pp. 2017–2024, 2024.
[70] Y. Chen, Q. P. Liu, H. Xie, and J. Ding, “From bench to bedside: current development and emerging trend of KRAS-targeted therapy,” Acta Pharmacologica Sinica, vol. 45, no. 4, pp. 686–703, 2024.
[71] M. B. Ryan and R. B. Corcoran, “Therapeutic strategies to target RAS-mutant cancers,” Nature Reviews Clinical Oncology, vol. 15, no. 11, pp. 709–720, 2018.
[72] M. Molina-Arcas and J. Downward, “Exploiting the therapeutic implications of KRAS inhibition on tumor immunity,” Cancer Cell, vol. 42, no. 3, pp. 338–357, 2024.
[73] W. H. Gmeiner, “Recent Advances in Therapeutic Strategies to Improve Colorectal Cancer Treatment,” Cancers, vol. 16, no. 5, Art. no. 1029, 2024.
[74] P. B. Chapman, D. B. Solit, and N. Rosen, “Combination of RAF and MEK inhibition for the treatment of BRAF-mutated melanoma: feedback is not encouraged,” Cancer Cell, vol. 26, no. 5, pp. 603–604, 2014.
[75] I. M. Sanchez, T. J. Purwin, I. Chervoneva, D. A. Erkes, M. Q. Nguyen, M. A. Davies, et al., “In vivo ERK1/2 reporter predictively models response and resistance to combined BRAF and MEK inhibitors in melanoma,” Molecular Cancer Therapeutics, vol. 18, no. 9, pp. 1637–1645, 2019.
[76] X. Ke and L. Shen, “Molecular targeted therapy of cancer: The progress and future prospect,” Frontiers in Laboratory Medicine, vol. 1, no. 2, pp. 69–75, 2017.
[77] J. Liu, “Cancer Targeted Molecular Therapy,” in Anesthesia for Oncological Surgery, Cham, Switzerland: Springer, 2024, pp. 27–34.
[78] C. L. Hann and J. R. Brahmer, “Who should receive epidermal growth factor receptor inhibitors for non-small cell lung cancer and when?,” Current Treatment Options in Oncology, vol. 8, no. 1, pp. 28–37, 2007.
[79] F. McCormick, “KRAS as a therapeutic target,” Clinical Cancer Research, vol. 21, no. 8, pp. 1797–1801, 2015.
[80] I. M. Meraz, M. Majidi, R. Song, F. Meng, L. Gao, Q. Wang, et al., “Mechanism of NPRL2 gene therapy induced antitumor immunity in KRAS/STK11mt aPD1 resistant metastatic NSCLC,” Cancer Research, vol. 84, no. 6_Supplement, p. 1420, 2024.
[81] C. Yang, Q. Fang, T. Zhou, Y. Xie, J. Yu, and J. Hao, “Enhancing PDAC therapy through in vivo correction of KRAS G12D using enIscB-directed base editor combined with ICB,” Cancer Research, vol. 84, no. 6_Supplement, p. 7246, 2024.
[82] X. Gong, J. Du, R. W. Peng, C. Chen, and Z. Yang, “CRISPRing KRAS: A Winding Road with a Bright Future in Basic and Translational Cancer Research,” Cancers, vol. 16, no. 2, Art. no. 460, 2024.
[83] R. Nussinov, T. Weichhart, Z. Dlamini, D. L. Gibbons, I. Van Seuningen, J. Konen, and H. Q. Ju, “Directions to overcome therapy resistance in cancer,” Trends in Pharmacological Sciences, 2024.
[84] R. Nussinov, C. J. Tsai, and H. Jang, “Anticancer drug resistance: An update and perspective,” Drug Resistance Updates, vol. 59, Art. no. 100796, 2021.
[85] Y. Shang, S. Fu, Q. Hao, H. Ying, J. Wang, and T. Shen, “Multiple medicinal chemistry strategies of targeting KRAS: State-of-the art and future directions,” Bioorganic Chemistry, Art. no. 107092, 2024.
[86] S. Kato, T. McFall, K. Takahashi, K. Bamel, S. Ikeda, R. N. Eskander, et al., “KRAS‐Mutated, Estrogen Receptor‐Positive Low‐Grade Serous Ovarian Cancer: Unraveling an Exceptional Response Mystery,” The Oncologist, vol. 26, no. 4, pp. e530–e536, 2021.
[87] A. Zamora, “Current and future treatments to target KRAS mutation in pancreatic ductal adenocarcinoma,” unpublished.
[88] S. Pant, Z. A. Wainberg, C. D. Weekes, M. Furqan, P. M. Kasi, C. E. Devoe, and E. M. O’Reilly, “Lymph-node-targeted, mKRAS-specific amphiphile vaccine in pancreatic and colorectal cancer: the phase 1 AMPLIFY-201 trial,” Nature Medicine, vol. 30, no. 2, pp. 531–542, 2024.
[89] H. Shen and C. Li, “Global research trends in immunotherapy for non-small cell lung cancer patients with KRAS mutations: a bibliometric analysis,” Frontiers in Oncology, 2024.
[90] I. Ranđelović, K. Nyíri, G. Koppány, M. Baranyi, J. Tóvári, A. Kigyós, and V. Grolmusz, “Gluing GAP to RAS Mutants: A New Approach to an Old Problem in Cancer Drug Development,” International Journal of Molecular Sciences, vol. 25, no. 5, Art. no. 2572, 2024.
[91] H. Khan, U. Shahab, A. Alshammari, A. R. Alyahyawi, R. Akasha, T. Alharazi, and Moinuddin, “Nano‐therapeutics: The upcoming nanomedicine to treat cancer,” IUBMB Life, 2024.
Downloads
Published
How to Cite
Issue
Section
License
Copyright (c) 2025 Ali Mohammed Hussein, Alaa Ramthan Hussein

This work is licensed under a Creative Commons Attribution 4.0 International License.