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 -- 1528 2024-02-07 12:51:06 |
2 update references and layout Meta information modification 1528 2024-02-08 02:55:01 |

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.
Morgos, D.; Stefani, C.; Miricescu, D.; Greabu, M.; Stanciu, S.; Nica, S.; Stanescu-Spinu, I.; Balan, D.G.; Balcangiu-Stroescu, A.; Coculescu, E.; et al. PI3K/AKT/mTOR and MAPK Pathways in Gastric Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/54850 (accessed on 01 May 2024).
Morgos D, Stefani C, Miricescu D, Greabu M, Stanciu S, Nica S, et al. PI3K/AKT/mTOR and MAPK Pathways in Gastric Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/54850. Accessed May 01, 2024.
Morgos, Diana-Theodora, Constantin Stefani, Daniela Miricescu, Maria Greabu, Silviu Stanciu, Silvia Nica, Iulia-Ioana Stanescu-Spinu, Daniela Gabriela Balan, Andra-Elena Balcangiu-Stroescu, Elena-Claudia Coculescu, et al. "PI3K/AKT/mTOR and MAPK Pathways in Gastric Cancer" Encyclopedia, https://encyclopedia.pub/entry/54850 (accessed May 01, 2024).
Morgos, D., Stefani, C., Miricescu, D., Greabu, M., Stanciu, S., Nica, S., Stanescu-Spinu, I., Balan, D.G., Balcangiu-Stroescu, A., Coculescu, E., Georgescu, D., & Nica, R.I. (2024, February 07). PI3K/AKT/mTOR and MAPK Pathways in Gastric Cancer. In Encyclopedia. https://encyclopedia.pub/entry/54850
Morgos, Diana-Theodora, et al. "PI3K/AKT/mTOR and MAPK Pathways in Gastric Cancer." Encyclopedia. Web. 07 February, 2024.
PI3K/AKT/mTOR and MAPK Pathways in Gastric Cancer
Edit

Gastric cancer (GC) is the fourth leading cause of death worldwide, with more than 1 million cases diagnosed every year. Helicobacter pylori represents the main risk factor, being responsible for 78% of the cases. Increased amounts of salt, pickled food, red meat, alcohol, smoked food, and refined sugars negatively affect the stomach wall, contributing to GC development.

gastric cancer PI3K/AKT/mTOR MAPK gene mutations

1. Introduction

1.1. Gastric Cancer Incidence

Gastric cancer (GC) is a multifunctional disease that is the fourth leading cause of death worldwide [1][2][3][4][5], without any symptoms in early stages [6]. Even though the incidence of GC has decreased since the 1970s due to medical progress, it still represents a major health problem, particularly in Eastern Asian countries, where over 1 million cases were reported in 2020 [7]. GC is considered an aggressive malignancy characterized by uncontrolled cell proliferation and metastasis [8]. Globally, every year, more than 1 million people are diagnosed with GC [9][10].
This malignant pathology is more often found in men, being approximately twice as common compared with women, and usually occurs after 60 years of age [11]. The lower risk of women developing GC is due to estrogen, an effect that lasts until menopause [12]. Moreover, GC is often found in the tropics [13]. GC incidence and mortality vary, with increased rates being observed in Eastern and Western Asia, while in North America, Northern Europe, and most African regions, decreased incidence is reported [14]. The southern region of India has an increased incidence of gastric adenocarcinoma, while South Asia presents decreased incidence [15]. Usually, 90% of GC involves sporadic cases, and 10% present familial aggregation [16]. Australia also has a decreased rate of GC [17]. These variations may be explained by differences in Helicobacter pylori (H. pylori) genotypes cagA and vacA; in areas with elevated GC incidence, type cagA is predominant [18]. Worldwide, the GC overall 5-year survival rate is about 20%. In China, GC represents the second leading cause of death among women and the third leading cause in men [19]. Japan has reported a survival rate of more than 70% for GC diagnosed in stages I and II. This elevated survival rate may be due to the mass screening programs adopted [20], where endoscopy is the gold standard method [21]. In the last five decades, the US has reported a decreased incidence of GC, while a subtype of GC, non-cardia, is increasing among adults younger than 50 years of age [22].

1.2. Gastric Cancer Classification

The Cancer Genome Atlas (TCGA) classifies GC into four subtypes [23]. Anatomic gastric adenocarcinoma is classified into two subtypes: cardia, which includes the upper stomach adjoining the esophagus, and non-cardia, which involves the mid and distal stomach [24]. Histologically, gastric adenocarcinomas represent 90% of GC and have been subdivided into two main types: the intestinal type, or well-differentiated, and the diffuse type, or undifferentiated [25]. The cardia subtype represents one-quarter of cases, while non-cardia represents three-quarters of the incidence worldwide [26]. According to the Laurean classification, the majority of GC cases are represented by adenocarcinoma (90%), and 10% are lymphoma or gastrointestinal stromal tumors [27]. The intestinal type accounts for 50% of cases, the diffuse type 33%, and 17% of cases are mixed or unclassified [27].
The World Health Organization (WHO) classifies GC into four subtypes: papillary, tubular, mucinous, and poorly cohesive [28]. Moreover, Epstein–Barr virus (EBV)-associated GC is another GC subtype, with definitive clinical and molecular characteristics [29], together with human epithelial growth receptor 2 (HER2), which is overexpressed in 6–30% of GC cases [30]. Infection with EBV contributes to 9% of all GC [31]. HER2 is also overexpressed in other malignant pathologies, including breast cancer [32]. The TCGA analyzed 295 primary gastric adenocarcinomas and further proposed a new classification of GC into four subtypes, including EBV-positive, microsatellite instability (MSI), genomic stability (GS), and chromosomal instability (CIN). The Asian Cancer Research Group (ACRG) also provides another classification of the four mentioned GC subtypes, as follows: (i) MSI, (ii) epithelial-to-mesenchymal transition (EMT)/GC, (iii) microsatellite stable with intact TP53 activity (MSS/TP53+), and (iv) microsatellite stable GC with loss of TP53 activity (MSS/TP53−) [33][34]. Moreover, GC intestinal histology is often found in Caucasians.
The cardia subtype is less detected in Africa and Latin America [35]. With the lack of significant symptoms in the early stages, most GC cases are diagnosed in middle or late stages, leading to a decreased survival rate [36][37]. In the early stages, a small number of patients manifest nausea, vomiting, or symptoms similar to ulcers. In advanced GC stages, patients usually report pain and weight loss as the most common symptoms [38]. Unfortunately, the 5-year survival rate is about 20–30% for advanced GC cases [39].

2. PI3K/AKT/mTOR and MAPK Pathways in Gastric Cancer

PI3K/AKT/mTOR signaling pathway dysregulation is found in various pathologies, including cancer progression such as in GC. This pathway is involved in various cancer-vital processes, such as apoptosis, autophagy, cell growth, survival, and proliferation [40][41][42][43][44]. This signaling pathway is one of the main regulators of the cell cycle, growth, proliferation, metastasis, apoptosis, and autophagy [45]. In GC, PI3K/AKT/mTOR inhibits apoptosis and induces the chemo-resistance phenotype, metastasis, angiogenesis, and EMT [46].
Besides the catalytic subunit of Class IA PI3K, other compounds such as the Class IA p85α-regulatory subunit, AKT, mTOR, and eukaryotic translation initiation factor 4E (eIF4E) possess oncogenic potential [47]. The downstream effectors of the PI3K/AKT/mTOR signaling pathway are usually dysregulated in the majority of solid tumors [48]. Moreover, PI3K/AKT/mTOR also controls cancer metabolism and genomic instability [49], having immunomodulatory potential as well [50].
In a sufficient supply of nutrients and energy, activated AKT will phosphorylate mTOR, further promoting tumor cell growth [51]. A very important component of this pathway, mTOR, is upregulated in malignancies, suppressing autophagy [52]. AKT was overexpressed in various neoplastic pathologies, such as pancreatic and ovarian cancer, as well as in gastric carcinoma, where the first isoform of AKT was found to be overexpressed [53]. GC-PI3K/AKT activation is induced by aberrant epigenetic regulations, necessary for GC development [54]. It is considered that PIK3CA is the second most common mutated gene in GC, responsible for 4–25% of cases, and also found in 85% of EBV GC cases [55]. Iranpour and his research group analyzed 100 patients with GC who underwent surgical resection and detected mutations in exon 20 of the PI3KCA gene in 11% of GC patients [56].
In gastric malignant tumors, p-AKT had elevated levels in 74% to 78%, being associated with GC angiogenesis and lymph nodes, leading to tumor invasion [57][58]. Overexpression of p-AKT is significantly correlated with HER2 overexpression and not with PIK3CA mutations. Moreover, the loss of PTEN is associated with GC initiation and development [57].
GC-EMT implies that the gastric cells lose their identity and acquire a mesenchymal phenotype with upregulation of N-cadherin, MMPs, and vimentin and downregulation of E-cadherin. PI3K/AKT/mTOR plays a pivotal role in EMT, leading to apoptosis resistance, invasion, and metastasis [59]. p-AKT induces dysregulation of the cell cycle, apoptosis suppression, and finally activation of angiogenesis. p-AKT will induce the phosphorylation of other proteins, including GSK-3β, BAD, caspase-9, and FoxOs [58].
Infection of gastric-epithelial cells with H. pylori will activate PI3K/AKT/mTOR and MAPK cascades, inducing the transformation of epithelial cells into neoplastic cells through changes including apoptosis, proliferation, and differentiation. The increased release of proinflammatory cytokines implies a higher production of free radicals, inducing DNA methylation [60].
RTK/MAPK pathway alteration, together with TP53 mutations, was detected in GC subtypes. A cell motility gene, CDH1, is usually mutated in GC subtypes and induces the activation of the RTK/MAPK cascade [61]. The PIK3CA gene mutation rate was increased in GC, compared with AKT1, AKT2, and AKT3-mutations genes. Moreover, AKT2 was significantly increased in EBV-positive GC as compared to EBV-negative GC. AKT2 mutation was correlated with a poor survival rate in EBV-positive GC [62]. HER2 is frequently overexpressed in GC, leading to PI3K/AKT/mTOR and MAPK signaling pathway activation via RTK receptors and to increased cell proliferation, invasion, and metastasis [63].
Raf-1 rs3729931, HRAS rs45604736, MAPK1 rs2283792, and MAPK1 rs9610417 genes are correlated with GC development [64]. Ras and Raf family member mutations induce the activation of RAF/MEK/ERK in GC [65]. GC-HER2 overexpression induces PI3K/AKT/mTOR and MAPK activation, promoting tumor-cell survival, proliferation, adhesion, and migration [66]. Lian et al. reported that IL-8 secretion via the ROS/NF-κB and ROS/MAPK (Erk1/2, p38)/AP-1 axis stimulates endothelial cell proliferation and angiogenesis in the GC tumor microenvironment [67]. Although ROS have a negative impact, they can also have positive roles, because they can be produced even by anti-cancer drugs to induce autophagy and apoptosis of malignant cells [68]. The PI3K/AKT/mTOR and Ras/Raf/MEK/ERK signaling pathways can be activated by common targets, such as Ras, and present some compensatory signaling properties; when one pathway is inhibited, the other is activated. Therefore, if mTOR is inhibited, PI3K will activate MAPK via Ras. These two cascades are also coactivated in various tumors, such as found in melanoma, prostate, and colorectal cancer [69]. Casein kinase II (CSNK2A1) is a serine/threonine kinase that is able to phosphorylate multiple substrates involved in cell cycle regulation, DNA repair and replication, transcription, apoptosis, and carcinogenesis. This kinase may be implicated in the processes of cancer invasion and migration via EMT transition and NF-kB signaling pathways. Regarding GC pathogenesis, CSNK2A1 is overexpressed, inducing the activation of AKT and mTOR, being involved in migration, invasion, and proliferation of GC cells [70].

References

  1. Machlowska, J.; Baj, J.; Sitarz, M.; Maciejewski, R.; Sitarz, R. Gastric cancer: Epidemiology, risk factors, classification, genomic characteristics and treatment strategies. Int. J. Mol. Sci. 2020, 21, 4012.
  2. Guggenheim, D.E.; Shah, M.A. Gastric cancer epidemiology and risk factors. J. Surg. Oncol. 2013, 107, 230–236.
  3. Bai, D.; Xiang, W.; Chen, X.; Hu, J. Risk factors of postoperative pulmonary infection of gastric cancer and perioperative intervention measures. Zhonghua Wei Chang. Wai Ke Za Zhi = Chin. J. Gastrointest. Surg. 2021, 24, 185–190.
  4. Yoon, H.; Kim, N. Diagnosis and management of high risk group for gastric cancer. Gut Liver 2015, 9, 5.
  5. Zhang, K.; Hu, X.; Su, J.; Li, D.; Thakur, A.; Gujar, V.; Cui, H. Gastrointestinal Cancer Therapeutics via Triggering Unfolded Protein Response and Endoplasmic Reticulum Stress by 2-Arylbenzofuran. Int. J. Mol. Sci. 2024, 25, 999.
  6. Yang, K.; Lu, L.; Liu, H.; Wang, X.; Gao, Y.; Yang, L.; Li, Y.; Su, M.; Jin, M.; Khan, S. A comprehensive update on early gastric cancer: Defining terms, etiology, and alarming risk factors. Expert Rev. Gastroenterol. Hepatol. 2021, 15, 255–273.
  7. Shah, D.; Bentrem, D. Environmental and genetic risk factors for gastric cancer. J. Surg. Oncol. 2022, 125, 1096–1103.
  8. Hu, Y.; Zhang, Y.; Ding, M.; Xu, R. Long noncoding RNA TMPO-AS1/miR-126-5p/BRCC3 axis accelerates gastric cancer progression and angiogenesis via activating PI3K/Akt/mTOR pathway. J. Gastroenterol. Hepatol. 2021, 36, 1877–1888.
  9. Thrift, A.P.; El-Serag, H.B. Burden of gastric cancer. Clin. Gastroenterol. Hepatol. 2020, 18, 534–542.
  10. Esposito, T.; Pisanti, S.; Mauro, L.; Mencherini, T.; Martinelli, R.; Aquino, R.P. Activity of Colocasia esculenta (Taro) Corms against Gastric Adenocarcinoma Cells: Chemical Study and Molecular Characterization. Int. J. Mol. Sci. 2023, 25, 252.
  11. Petryszyn, P.; Chapelle, N.; Matysiak-Budnik, T. Gastric cancer: Where are we heading? Dig. Dis. 2020, 38, 280–285.
  12. Camargo, M.C.; Song, M.; Xu, X.; Zhao, I.; Sampson, J.N.; Etemadi, A.; Brenner, H.; Lee, H.W.; Trabert, B.; Holleczek, B. Urinary estrogen metabolites and gastric cancer risk among postmenopausal women. Cancer Rep. 2022, 5, e1574.
  13. Imounga, L.M.; Plenet, J.; Belliardo, S.; Chine, E.C.; Louvel, D.; Cenciu, B.; Couppié, P.; Alsibai, K.D.; Nacher, M. Gastric cancer incidence and mortality in French Guiana: South American or French? J. Gastrointest. Cancer 2022, 53, 204–210.
  14. Gullo, I.; Grillo, F.; Mastracci, L.; Vanoli, A.; Carneiro, F.; Saragoni, L.; Limarzi, F.; Ferro, J.; Parente, P.; Fassan, M. Precancerous lesions of the stomach, gastric cancer and hereditary gastric cancer syndromes. Pathologica 2020, 112, 166.
  15. Daniyal, M.; Ahmad, S.; Ahmad, M.; Asif, H.M.; Akram, M.; Rehman, S.U.; Sultana, S. Risk factors and epidemiology of gastric cancer in Pakistan. Asian Pac. J. Cancer Prev. 2015, 16, 4821–4824.
  16. Choi, Y.J.; Kim, N. Gastric cancer and family history. Korean J. Intern. Med. 2016, 31, 1042.
  17. Arnold, M.; Park, J.Y.; Camargo, M.C.; Lunet, N.; Forman, D.; Soerjomataram, I. Is gastric cancer becoming a rare disease? A global assessment of predicted incidence trends to 2035. Gut 2020, 69, 823–829.
  18. Yamaoka, Y.; Kato, M.; Asaka, M. Geographic differences in gastric cancer incidence can be explained by differences between Helicobacter pylori strains. Intern. Med. 2008, 47, 1077–1083.
  19. Ke, J.; Ma, P.; Chen, J.; Qin, J.; Qian, H. LGR6 promotes the progression of gastric cancer through PI3K/AKT/mTOR pathway. OncoTargets Ther. 2018, 11, 3025–3033.
  20. Karimi, P.; Islami, F.; Anandasabapathy, S.; Freedman, N.D.; Kamangar, F. Gastric cancer: Descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol. Biomark. Prev. 2014, 23, 700–713.
  21. Akbarpour, E.; Sadjadi, A.; Derakhshan, M.; Roshandel, G.; Alimohammadian, M. Gastric cancer in Iran: An overview of risk factors and preventive measures. Arch. Iran. Med. 2021, 24, 556–567.
  22. Thrift, A.P.; Nguyen, T.H. Gastric cancer epidemiology. Gastrointest. Endosc. Clin. 2021, 31, 425–439.
  23. van Velzen, M.J.; Braemer, M.; Nieuwenhuijzen, G.A.; van Sandick, J.W.; Siersema, P.D.; Ruurda, J.P.; Verheij, M.; Spaander, M.C.; Beerepoot, L.V.; Mohammad, N.H. Incidence, Stage, Treatment, and Survival of Noncardia Gastric Cancer. JAMA Netw. Open 2023, 6, e2330018.
  24. Xia, J.Y.; Aadam, A.A. Advances in screening and detection of gastric cancer. J. Surg. Oncol. 2022, 125, 1104–1109.
  25. Crew, K.D.; Neugut, A.I. Epidemiology of gastric cancer. World J. Gastroenterol. WJG 2006, 12, 354.
  26. Huang, R.J.; Hwang, J.H. Improving the early diagnosis of gastric cancer. Gastrointest. Endosc. Clin. 2021, 31, 503–517.
  27. Riquelme, I.; Saavedra, K.; Espinoza, J.A.; Weber, H.; García, P.; Nervi, B.; Garrido, M.; Corvalán, A.H.; Roa, J.C.; Bizama, C. Molecular classification of gastric cancer: Towards a pathway-driven targeted therapy. Oncotarget 2015, 6, 24750.
  28. Chia, N.-Y.; Tan, P. Molecular classification of gastric cancer. Ann. Oncol. 2016, 27, 763–769.
  29. Moon, S.H.; Park, N.-S.; Noh, M.H.; Kim, Y.S.; Cheong, S.H.; Hur, D.Y. Olaparib-induced Apoptosis Through EBNA1-ATR-p38 MAPK Signaling Pathway in Epstein-Barr Virus-positive Gastric Cancer Cells. Anticancer Res. 2022, 42, 555–563.
  30. Mezynski, M.J.; Farrelly, A.M.; Cremona, M.; Carr, A.; Morgan, C.; Workman, J.; Armstrong, P.; McAuley, J.; Madden, S.; Fay, J. Targeting the PI3K and MAPK pathways to improve response to HER2-targeted therapies in HER2-positive gastric cancer. J. Transl. Med. 2021, 19, 184.
  31. Osumi, H.; Kawachi, H.; Yoshio, T.; Fujisaki, J. Clinical impact of Epstein-Barr virus status on the incidence of lymph node metastasis in early gastric cancer. Dig. Endosc. 2020, 32, 316–322.
  32. Jafarinezhad, S.; Assaran Darban, R.; Javid, H.; Hashemy, S.I. The SP/NK1R system promotes the proliferation of breast cancer cells through NF-κB-mediated inflammatory responses. Cell Biochem. Biophys. 2023, 81, 787–794.
  33. Puliga, E.; Corso, S.; Pietrantonio, F.; Giordano, S. Microsatellite instability in Gastric Cancer: Between lights and shadows. Cancer Treat. Rev. 2021, 95, 102175.
  34. Kim, Y.; Cho, M.-Y.; Kim, J.; Kim, S.N.; Oh, S.C.; Lee, K.-A. Profiling cancer-associated genetic alterations and molecular classification of cancer in Korean gastric cancer patients. Oncotarget 2017, 8, 69888.
  35. Ang, T.L.; Fock, K.M. Clinical epidemiology of gastric cancer. Singap. Med. J. 2014, 55, 621.
  36. Qiu, P.; Li, X.; Gong, M.; Wen, P.; Wen, J.; Xu, L.; Wang, G. SPI1 Mediates N-Myristoyltransferase 1 to Advance Gastric Cancer Progression via PI3K/AKT/mTOR Pathway. Can. J. Gastroenterol. Hepatol. 2023, 2023, 2021515.
  37. He, P.; He, Y.; Ma, J.; Liu, Y.; Liu, C.; Baoping, Y.; Dong, W. Thymoquinone induces apoptosis and protective autophagy in gastric cancer cells by inhibiting the PI3K/Akt/mTOR pathway. Phytother. Res. 2023, 37, 3467–3480.
  38. Zhou, D.; He, Y.; Li, H.; Huang, W. KLK6 mediates stemness and metabolism of gastric carcinoma cells via the PI3K/AKT/mTOR signaling pathway. Oncol. Lett. 2021, 22, 824.
  39. Li, C.; Liang, G.; Yang, S.; Sui, J.; Wu, W.; Xu, S.; Ye, Y.; Shen, B.; Zhang, X.; Zhang, Y. LncRNA-LOC101928316 contributes to gastric cancer progression through regulating PI3K-Akt-mTOR signaling pathway. Cancer Med. 2019, 8, 4428–4440.
  40. Fattahi, S.; Amjadi-Moheb, F.; Tabaripour, R.; Ashrafi, G.H.; Akhavan-Niaki, H. PI3K/AKT/mTOR signaling in gastric cancer: Epigenetics and beyond. Life Sci. 2020, 262, 118513.
  41. Zhao, W.; Zhao, X.; Xu, M.; Cheng, Z.; Zhang, Z. Knockdown of LINC01279 Suppresses Gastric Cancer Proliferation and Migration by Inhibiting PI3K/Akt/mTOR Signaling Pathway. J. Oncol. 2022, 2022, 6228982.
  42. Gu, X.-J.; Li, Y.-J.; Wang, F.; Ye, T. MiR-30e-3p inhibits gastric cancer development by negatively regulating THO complex 2 and PI3K/AKT/mTOR signaling. World J. Gastrointest. Oncol. 2022, 14, 2170.
  43. Li, H.; Zhao, S.; Shen, L.; Wang, P.; Liu, S.; Ma, Y.; Liang, Z.; Wang, G.; Lv, J.; Qiu, W. E2F2 inhibition induces autophagy via the PI3K/Akt/mTOR pathway in gastric cancer. Aging (Albany NY) 2021, 13, 13626.
  44. Wang, N.; Dong, Q.; Zhou, X.-N. LMO4 promotes the invasion and proliferation of gastric cancer by activating PI3K-Akt-mTOR signaling. Am. J. Transl. Res. 2019, 11, 6534.
  45. Wang, R.; Ha, K.-Y.; Dhandapani, S.; Kim, Y.-J. Biologically synthesized black ginger-selenium nanoparticle induces apoptosis and autophagy of AGS gastric cancer cells by suppressing the PI3K/Akt/mTOR signaling pathway. J. Nanobiotechnol. 2022, 20, 441.
  46. Khorasani, A.B.S.; Pourbagheri-Sigaroodi, A.; Pirsalehi, A.; Safaroghli-Azar, A.; Zali, M.R.; Bashash, D. The PI3K/Akt/mTOR signaling pathway in gastric cancer; from oncogenic variations to the possibilities for pharmacologic interventions. Eur. J. Pharmacol. 2021, 898, 173983.
  47. Aoki, M.; Fujishita, T. Oncogenic roles of the PI3K/AKT/mTOR axis. Viruses Genes Cancer 2017, 407, 153–189.
  48. Kang, B.W.; Chau, I. Molecular target: Pan-AKT in gastric cancer. ESMO Open 2020, 5, e000728.
  49. Wang, Z.; Wang, X.; Xu, Y.; Li, J.; Zhang, X.; Peng, Z.; Hu, Y.; Zhao, X.; Dong, K.; Zhang, B. Mutations of PI3K-AKT-mTOR pathway as predictors for immune cell infiltration and immunotherapy efficacy in dMMR/MSI-H gastric adenocarcinoma. BMC Med. 2022, 20, 133.
  50. Koh, V.; Chakrabarti, J.; Torvund, M.; Steele, N.; Hawkins, J.A.; Ito, Y.; Wang, J.; Helmrath, M.A.; Merchant, J.L.; Ahmed, S.A. Hedgehog transcriptional effector GLI mediates mTOR-Induced PD-L1 expression in gastric cancer organoids. Cancer Lett. 2021, 518, 59–71.
  51. Han, S.-H.; Lee, J.-H.; Woo, J.-S.; Jung, G.-H.; Jung, S.-H.; Han, E.-J.; Kim, B.; Cho, S.D.; Nam, J.S.; Che, J.H. Myricetin induces apoptosis and autophagy in human gastric cancer cells through inhibition of the PI3K/Akt/mTOR pathway. Heliyon 2022, 8, e09309.
  52. Rong, L.; Li, Z.; Leng, X.; Li, H.; Ma, Y.; Chen, Y.; Song, F. Salidroside induces apoptosis and protective autophagy in human gastric cancer AGS cells through the PI3K/Akt/mTOR pathway. Biomed. Pharmacother. 2020, 122, 109726.
  53. Xia, P.; Xu, X.-Y. PI3K/Akt/mTOR signaling pathway in cancer stem cells: From basic research to clinical application. Am. J. Cancer Res. 2015, 5, 1602.
  54. Yu, L.; Wei, J.; Liu, P. Attacking the PI3K/Akt/mTOR signaling pathway for targeted therapeutic treatment in human cancer. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2022; pp. 69–94.
  55. Stanland, L.J.; Ang, H.X.; Hoj, J.P.; Chu, Y.; Tan, P.; Wood, K.C.; Luftig, M.A. CBF-Beta Mitigates PI3K-Alpha–Specific Inhibitor Killing through PIM1 in PIK3CA-Mutant Gastric Cancer. Mol. Cancer Res. 2023, 21, 1148–1162.
  56. Iranpour, M.; Nourian, M.; Saffari, S.; Samizadeh, E.; Mirghafori, M.; Iravani, S.; Ghafouri-Fard, S. PIK3CA mutation analysis in iranian patients with gastric cancer. Iran. Biomed. J. 2019, 23, 87.
  57. Matsuoka, T.; Yashiro, M. The role of PI3K/Akt/mTOR signaling in gastric carcinoma. Cancers 2014, 6, 1441–1463.
  58. Kipkeeva, F.; Muzaffarova, T.; Korotaeva, A.; Nikulin, M.; Grishina, K.; Mansorunov, D.; Apanovich, P.; Karpukhin, A. MicroRNA in gastric cancer development: Mechanisms and biomarkers. Diagnostics 2020, 10, 891.
  59. Wang, C.; Yang, Z.; Xu, E.; Shen, X.; Wang, X.; Li, Z.; Yu, H.; Chen, K.; Hu, Q.; Xia, X. Apolipoprotein C-II induces EMT to promote gastric cancer peritoneal metastasis via PI3K/AKT/mTOR pathway. Clin. Transl. Med. 2021, 11, e522.
  60. Yousefi, B.; Mohammadlou, M.; Abdollahi, M.; Salek Farrokhi, A.; Karbalaei, M.; Keikha, M.; Kokhaei, P.; Valizadeh, S.; Rezaiemanesh, A.; Arabkari, V. Epigenetic changes in gastric cancer induction by Helicobacter pylori. J. Cell. Physiol. 2019, 234, 21770–21784.
  61. Seidlitz, T.; Schmäche, T.; Lee, J.H.; Qin, N.; Kochall, S.; Fohgrub, J.; Pauck, D.; Rothe, A.; Koo, B.K.; Weitz, J. Sensitivity towards HDAC inhibition is associated with RTK/MAPK pathway activation in gastric cancer. EMBO Mol. Med. 2022, 14, e15705.
  62. Lin, Y.; Wu, Z.; Guo, W.; Li, J. Gene mutations in gastric cancer: A review of recent next-generation sequencing studies. Tumor Biol. 2015, 36, 7385–7394.
  63. Shi, W.; Zhang, G.; Ma, Z.; Li, L.; Liu, M.; Qin, L.; Yu, Z.; Zhao, L.; Liu, Y.; Zhang, X. Hyperactivation of HER2-SHCBP1-PLK1 axis promotes tumor cell mitosis and impairs trastuzumab sensitivity to gastric cancer. Nat. Commun. 2021, 12, 2812.
  64. Gonzalez-Hormazabal, P.; Musleh, M.; Bustamante, M.; Stambuk, J.; Pisano, R.; Valladares, H.; Lanzarini, E.; Chiong, H.; Rojas, J.; Suazo, J. Polymorphisms in RAS/RAF/MEK/ERK pathway are associated with gastric cancer. Genes 2018, 10, 20.
  65. Ahn, S.; Brant, R.; Sharpe, A.; Dry, J.R.; Hodgson, D.R.; Kilgour, E.; Kim, K.; Kim, S.T.; Park, S.H.; Kang, W.K. Correlation between MEK signature and Ras gene alteration in advanced gastric cancer. Oncotarget 2017, 8, 107492.
  66. Ning, G.; Zhu, Q.; Kang, W.; Lee, H.; Maher, L.; Suh, Y.-S.; Michaud, M.; Silva, M.; Kwon, J.Y.; Zhang, C. A novel treatment strategy for lapatinib resistance in a subset of HER2-amplified gastric cancer. BMC Cancer 2021, 21, 923.
  67. Lian, S.; Li, S.; Zhu, J.; Xia, Y.; Do Jung, Y. Nicotine stimulates IL-8 expression via ROS/NF-κB and ROS/MAPK/AP-1 axis in human gastric cancer cells. Toxicology 2022, 466, 153062.
  68. Zhu, Q.; Guo, Y.; Chen, S.; Fu, D.; Li, Y.; Li, Z.; Ni, C. Irinotecan induces autophagy-dependent apoptosis and positively regulates ROS-related JNK-and P38-MAPK pathways in gastric cancer cells. OncoTargets Ther. 2020, 13, 2807–2817.
  69. Shimizu, T.; Tolcher, A.; Papadopoulos, K.; Beeram, M.; Rasco, D.; Smith, L.; Gunn, S.; Smetzer, L.; Mays, T.; Kaiser, B. The clinical effect of the dual-targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in first-in-human phase I study: The START Center experience. J. Clin. Oncol. 2011, 29, 2502.
  70. Jiang, C.; Ma, Z.; Zhang, G.; Yang, X.; Du, Q.; Wang, W. CSNK2A1 promotes gastric cancer invasion through the PI3K-Akt-mTOR signaling pathway. Cancer Manag. Res. 2019, 11, 10135–10143.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , , ,
View Times: 108
Entry Collection: Gastrointestinal Disease
Revisions: 2 times (View History)
Update Date: 08 Feb 2024
1000/1000