Submitted:
29 September 2025
Posted:
30 September 2025
You are already at the latest version
Abstract
Keywords:
1. Introduction
2. Genetic Alterations in PDAC
3. Genetic Alterations in Precursor Lesions of PDAC
4. Transcriptomic PDAC Subtypes
5. Therapeutic implications of genetic alteration in PDAC
5.1. Targeted Therapies
5.1.1. Targeting KRAS Mutations in PDAC
5.1.2. Targeting Non-KRAS Drivier Mutations in PDAC
5.2. DNA Repair Pathway in PDAC
5.3. Tumor Suppressor Pathways
5.4. Other Oncogenic Pathways
5.5. Stromal Targets and Tumor Microenvironment
5.6. Therapeutic Implication of PDAC Molecular Subtype
6. Discussion
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Karamitopoulou, E. Tumour microenvironment of pancreatic cancer: Immune landscape is dictated by molecular and histopathological features. Br. J. Cancer 2019, 121, 5–14. [Google Scholar] [CrossRef]
- Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef]
- Hosoda, W.; Wood, L.D. Molecular Genetics of Pancreatic Neoplasms. Surg. Pathol. Clin. 2016, 9, 685–703. [Google Scholar] [CrossRef]
- Luo, J. KRAS mutation in pancreatic cancer. Semin Oncol. 2021, 48, 10–18. [Google Scholar] [CrossRef]
- Taherian M, Wang H, Wang H. Pancreatic Ductal Adenocarcinoma: Molecular Pathology and Predictive Biomarkers. Cells. 2022, 11. [Google Scholar]
- Waddell, N.; Pajic, M.; Patch, A.-M.; Chang, D.K.; Kassahn, K.S.; Bailey, P.; Johns, A.L.; Miller, D.; Nones, K.; Quek, K.; et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015, 518, 495–501. [Google Scholar] [CrossRef]
- Jones, S.; Zhang, X.; Parsons, D.W.; Lin, J.C.-H.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Kamiyama, H.; Jimeno, A.; et al. Core Signaling Pathways in Human Pancreatic Cancers Revealed by Global Genomic Analyses. Science 2008, 321, 1801–1806. [Google Scholar] [CrossRef]
- Redston, M.S.; Caldas, C.; Seymour, A.B.; Hruban, R.H.; Da Costa, L.; Yeo, C.J.; E Kern, S. p53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Res. 1994, 54, 3025–3033. [Google Scholar] [PubMed]
- Bieging, K.T.; Mello, S.S.; Attardi, L.D. Unravelling mechanisms of p53-mediated tumour suppression. Nat. Rev. Cancer 2014, 14, 359–370. [Google Scholar] [CrossRef] [PubMed]
- Voutsadakis, I.A. Mutations of p53 associated with pancreatic cancer and therapeutic implications. Ann. Hepato-Biliary-Pancreatic Surg. 2021, 25, 315–327. [Google Scholar] [CrossRef] [PubMed]
- Schutte, M.; Hruban, R.H.; Geradts, J.; Maynard, R.; Hilgers, W.; Rabindran, S.K.; A Moskaluk, C.; A Hahn, S.; Schwarte-Waldhoff, I.; Schmiegel, W.; et al. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 1997, 57, 3126–3130. [Google Scholar]
- Ueki, T.; Toyota, M.; Sohn, T.; Yeo, C.J.; Issa, J.P.; Hruban, R.H.; Goggins, M. Hypermethylation of multiple genes in pancreatic adenocarcinoma. Cancer Res 2000, 60, 1835–1839. [Google Scholar]
- Kim, W.Y.; Sharpless, N.E. The Regulation of INK4/ARF in Cancer and Aging. Cell 2006, 127, 265–275. [Google Scholar] [CrossRef]
- Bartsch, D.K.; Sina-Frey, M.; Lang, S.; Wild, A.; Gerdes, B.; Barth, P.; Kress, R.M.; Grützmann, R.; Colombo-Benkmann, M.; Ziegler, A.; et al. CDKN2A Germline Mutations in Familial Pancreatic Cancer. Ann. Surg. 2002, 236, 730–737. [Google Scholar] [CrossRef]
- A Hahn, S.; Hoque, A.T.; A Moskaluk, C.; Da Costa, L.T.; Schutte, M.; Rozenblum, E.; Seymour, A.B.; Weinstein, C.L.; Yeo, C.J.; Hruban, R.H.; et al. Homozygous deletion map at 18q21.1 in pancreatic cancer. Cancer Res. 1996, 56, 490–494. [Google Scholar]
- Siegel, P.M.; Massagué, J. Cytostatic and apoptotic actions of TGF-β in homeostasis and cancer. Nat. Rev. Cancer 2003, 3, 807–820. [Google Scholar] [CrossRef]
- Mello, S.S.; Flowers, B.M.; Mazur, P.K.; Lee, J.J.; Müller, F.; Denny, S.K.; Ferreira, S.; Hanson, K.; Kim, S.K.; Greenleaf, W.J.; et al. Multifaceted role for p53 in pancreatic cancer suppression. Proc. Natl. Acad. Sci. 2023, 120. [Google Scholar] [CrossRef] [PubMed]
- Hong SM, Park JY, Hruban RH, Goggins M. Molecular signatures of pancreatic cancer. Arch Pathol Lab Med. 2011;135(6):716-27.
- Sivapalan, L.; Kocher, H.; Ross-Adams, H.; Chelala, C. The molecular landscape of pancreatic ductal adenocarcinoma. Pancreatology 2022, 22, 925–936. [Google Scholar] [CrossRef] [PubMed]
- Kitamura, H.; Morizane, C.; Tanabe, N.; Go, I.; Maruki, Y.; Ohba, A.; Nagashio, Y.; Kondo, S.; Hijioka, S.; Ueno, H.; et al. Clinical features of germline BRCA1/2 or ATM pathogenic variant positive pancreatic cancer in Japan. Pancreatology 2023, 23, 964–969. [Google Scholar] [CrossRef]
- Bazzichetto, C.; Luchini, C.; Conciatori, F.; Vaccaro, V.; Di Cello, I.; Mattiolo, P.; Falcone, I.; Ferretti, G.; Scarpa, A.; Cognetti, F.; et al. Morphologic and Molecular Landscape of Pancreatic Cancer Variants as the Basis of New Therapeutic Strategies for Precision Oncology. Int. J. Mol. Sci. 2020, 21, 8841. [Google Scholar] [CrossRef] [PubMed]
- Wilson, B.G.; Roberts, C.W.M. SWI/SNF nucleosome remodellers and cancer. Nat. Rev. Cancer 2011, 11, 481–492. [Google Scholar] [CrossRef]
- Alors-Perez, E.; Blázquez-Encinas, R.; Alcalá, S.; Viyuela-García, C.; Pedraza-Arevalo, S.; Herrero-Aguayo, V.; Jiménez-Vacas, J.M.; Mafficini, A.; Sánchez-Frías, M.E.; Cano, M.T.; et al. Dysregulated splicing factor SF3B1 unveils a dual therapeutic vulnerability to target pancreatic cancer cells and cancer stem cells with an anti-splicing drug. J. Exp. Clin. Cancer Res. 2021, 40, 1–21. [Google Scholar] [CrossRef] [PubMed]
- Alors-Pérez, E.; Pedraza-Arevalo, S.; Blázquez-Encinas, R.; Moreno-Montilla, M.T.; García-Vioque, V.; Berbel, I.; Luque, R.M.; Sainz, B.; Ibáñez-Costa, A.; Castaño, J.P. Splicing alterations in pancreatic ductal adenocarcinoma: A new molecular landscape with translational potential. J. Exp. Clin. Cancer Res. 2023, 42, 1–16. [Google Scholar] [CrossRef]
- Tan, M.C.; Basturk, O.; Brannon, R.A.; Bhanot, U.; Scott, S.N.; Bouvier, N.; LaFemina, J.; Jarnagin, W.R.; Berger, M.F.; Klimstra, D.; et al. GNAS and KRAS Mutations Define Separate Progression Pathways in Intraductal Papillary Mucinous Neoplasm-Associated Carcinoma. J. Am. Coll. Surg. 2015, 220, 845–854e1. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Karam, R.; Zhou, Y.; Su, F.; Ji, Y.; Li, G.; Xu, G.; Lu, L.; Wang, C.; Song, M.; et al. The UPF1 RNA surveillance gene is commonly mutated in pancreatic adenosquamous carcinoma. Nat. Med. 2014, 20, 596–598. [Google Scholar] [CrossRef]
- Chen, B.-L.; Wang, H.-M.; Lin, X.-S.; Zeng, Y.-M. UPF1: A potential biomarker in human cancers. Front. Biosci. 2021, 26, 76–84. [Google Scholar] [CrossRef]
- Polaski, J.T.; Udy, D.B.; Escobar-Hoyos, L.F.; Askan, G.; Leach, S.D.; Ventura, A.; Kannan, R.; Bradley, R.K. The origins and consequences of UPF1 variants in pancreatic adenosquamous carcinoma. eLife 2021, 10. [Google Scholar] [CrossRef]
- Brody JR, Costantino CL, Potoczek M, Cozzitorto J, McCue P, Yeo CJ; et al. Adenosquamous carcinoma of the pancreas harbors KRAS2, DPC4 and TP53 molecular alterations similar to pancreatic ductal adenocarcinoma. Mod Pathol. 2009;22(5):651-9.
- Xiong, Q.; Zhang, Z.; Xu, Y.; Zhu, Q. Pancreatic Adenosquamous Carcinoma: A Rare Pathological Subtype of Pancreatic Cancer. J. Clin. Med. 2022, 11, 7401. [Google Scholar] [CrossRef]
- Wilentz RE, Goggins M, Redston M, Marcus VA, Adsay NV, Sohn TA; et al. Genetic, immunohistochemical, and clinical features of medullary carcinoma of the pancreas: A newly described and characterized entity. Am J Pathol. 2000;156(5):1641-51.
- Banville, N.; Geraghty, R.; Fox, E.; Leahy, D.; Green, A.; Keegan, D.; Geoghegan, J.; Odonoghue, D.; Hyland, J.; Sheahan, K. Medullary carcinoma of the pancreas in a man with hereditary nonpolyposis colorectal cancer due to a mutation of the MSH2 mismatch repair gene. Hum. Pathol. 2006, 37, 1498–1502. [Google Scholar] [CrossRef]
- Feldmann, G.; Beaty, R.; Hruban, R.H.; Maitra, A. Molecular genetics of pancreatic intraepithelial neoplasia. J. Hepato-Biliary-Pancreatic Surg. 2007, 14, 224–232. [Google Scholar] [CrossRef] [PubMed]
- Paini, M.; Crippa, S.; Partelli, S.; Scopelliti, F.; Tamburrino, D.; Baldoni, A.; Falconi, M. Molecular pathology of intraductal papillary mucinous neoplasms of the pancreas. World J. Gastroenterol. 2014, 20, 10008–10023. [Google Scholar] [CrossRef]
- Loregger, A.; Grandl, M.; Mejías-Luque, R.; Allgäuer, M.; Degenhart, K.; Haselmann, V.; Oikonomou, C.; Hatzis, P.; Janssen, K.-P.; Nitsche, U.; et al. The E3 ligase RNF43 inhibits Wnt signaling downstream of mutated β-catenin by sequestering TCF4 to the nuclear membrane. Sci. Signal. 2015, 8, ra90–ra90. [Google Scholar] [CrossRef]
- Zhou, X.; Sun, Z.; Zhang, M.; Qu, X.; Yang, S.; Wang, L.; Jing, Y.; Li, L.; Deng, W.; Liu, F.; et al. Deficient Rnf43 potentiates hyperactive Kras-mediated pancreatic preneoplasia initiation and malignant transformation. Anim. Model. Exp. Med. 2022, 5, 61–71. [Google Scholar] [CrossRef]
- Kuboki, Y.; Shimizu, K.; Hatori, T.; Yamamoto, M.; Shibata, N.; Shiratori, K.; Furukawa, T. Molecular Biomarkers for Progression of Intraductal Papillary Mucinous Neoplasm of the Pancreas. Pancreas 2015, 44, 227–235. [Google Scholar] [CrossRef]
- Basturk, O.; Tan, M.; Bhanot, U.; Allen, P.; Adsay, V.; Scott, S.N.; Shah, R.; Berger, M.F.; Askan, G.; Dikoglu, E.; et al. The oncocytic subtype is genetically distinct from other pancreatic intraductal papillary mucinous neoplasm subtypes. Mod. Pathol. 2016, 29, 1058–1069. [Google Scholar] [CrossRef]
- Vyas, M.; Hechtman, J.F.; Zhang, Y.; Benayed, R.; Yavas, A.; Askan, G.; Shia, J.; Klimstra, D.S.; Basturk, O. DNAJB1-PRKACA fusions occur in oncocytic pancreatic and biliary neoplasms and are not specific for fibrolamellar hepatocellular carcinoma. Mod. Pathol. 2019, 33, 648–656. [Google Scholar] [CrossRef]
- Springer, S.; Wang, Y.; Dal Molin, M.; Masica, D.L.; Jiao, Y.; Kinde, I.; Blackford, A.; Raman, S.P.; Wolfgang, C.L.; Tomita, T.; et al. A Combination of Molecular Markers and Clinical Features Improve the Classification of Pancreatic Cysts. Gastroenterology 2015, 149, 1501–1510. [Google Scholar] [CrossRef] [PubMed]
- Sakihama, K.; Koga, Y.; Yamamoto, T.; Shimada, Y.; Yamada, Y.; Kawata, J.; Shindo, K.; Nakamura, M.; Oda, Y. RNF43 as a predictor of malignant transformation of pancreatic mucinous cystic neoplasm. Virchows Arch. 2022, 480, 1189–1199. [Google Scholar] [CrossRef] [PubMed]
- Hosoda, W.; Sasaki, E.; Murakami, Y.; Yamao, K.; Shimizu, Y.; Yatabe, Y. GNAS mutation is a frequent event in pancreatic intraductal papillary mucinous neoplasms and associated adenocarcinomas. Virchows Arch. 2015, 466, 665–674. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Matthaei, H.; Maitra, A.; Dal Molin, M.; Wood, L.D.; Eshleman, J.R.; Goggins, M.; Canto, M.I.; Schulick, R.D.; Edil, B.H.; et al. Recurrent GNAS Mutations Define an Unexpected Pathway for Pancreatic Cyst Development. Sci. Transl. Med. 2011, 3, 92ra66. [Google Scholar] [CrossRef]
- Fischer, C.G.; Wood, L.D. From somatic mutation to early detection: Insights from molecular characterization of pancreatic cancer precursor lesions. J. Pathol. 2018, 246, 395–404. [Google Scholar] [CrossRef]
- Iacobuzio-Donahue CA, Wilentz RE, Argani P, Yeo CJ, Cameron JL, Kern SE, Hruban RH. Dpc4 protein in mucinous cystic neoplasms of the pancreas: Frequent loss of expression in invasive carcinomas suggests a role in genetic progression. Am J Surg Pathol. 2000;24(11):1544-8.
- Mafficini, A.; Simbolo, M.; Shibata, T.; Hong, S.-M.; Pea, A.; Brosens, L.A.; Cheng, L.; Antonello, D.; Sciammarella, C.; Cantù, C.; et al. Integrative characterization of intraductal tubulopapillary neoplasm (ITPN) of the pancreas and associated invasive adenocarcinoma. Mod. Pathol. 2022, 35, 1929–1943. [Google Scholar] [CrossRef] [PubMed]
- Basturk, O.; Berger, M.F.; Yamaguchi, H.; Adsay, V.; Askan, G.; Bhanot, U.K.; Zehir, A.; Carneiro, F.; Hong, S.-M.; Zamboni, G.; et al. Pancreatic intraductal tubulopapillary neoplasm is genetically distinct from intraductal papillary mucinous neoplasm and ductal adenocarcinoma. Mod. Pathol. 2017, 30, 1760–1772. [Google Scholar] [CrossRef]
- Salati, M.; Caputo, F.; Baldessari, C.; Carotenuto, P.; Messina, M.; Caramaschi, S.; Dominici, M.; Bonetti, L.R. The Evolving Role of FGFR2 Inhibitors in Intrahepatic Cholangiocarcinoma: From Molecular Biology to Clinical Targeting. Cancer Manag. Res. 2021, ume 13, 7747–7757. [Google Scholar] [CrossRef]
- Collisson, E.A.; Sadanandam, A.; Olson, P.; Gibb, W.J.; Truitt, M.; Gu, S.; Cooc, J.; Weinkle, J.; Kim, G.E.; Jakkula, L.; et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat. Med. 2011, 17, 500–503. [Google Scholar] [CrossRef]
- Moffitt, R.A.; Marayati, R.; Flate, E.L.; Volmar, K.E.; Loeza, S.G.H.; Hoadley, K.A.; Rashid, N.U.; Williams, L.A.; Eaton, S.C.; Chung, A.H.; et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet. 2015, 47, 1168–1178. [Google Scholar] [CrossRef]
- Bailey, P.; Chang, D.K.; Nones, K.; Johns, A.L.; Patch, A.-M.; Gingras, M.-C.; Miller, D.K.; Christ, A.N.; Bruxner, T.J.C.; Quinn, M.C.; et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016, 531, 47–52. [Google Scholar] [CrossRef] [PubMed]
- Puleo, F.; Nicolle, R.; Blum, Y.; Cros, J.; Marisa, L.; Demetter, P.; Quertinmont, E.; Svrcek, M.; Elarouci, N.; Iovanna, J.L.; et al. Stratification of Pancreatic Ductal Adenocarcinomas Based on Tumor and Microenvironment Features. Gastroenterology 2018, 155, 1999–2013.e1993. [Google Scholar] [CrossRef]
- Cancer Genome Atlas Research Network. Electronic address aadhe, Cancer Genome Atlas Research N. Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma. Cancer Cell. 2017;32(2):185-203 e13.
- Chan-Seng-Yue, M.; Kim, J.C.; Wilson, G.W.; Ng, K.; Figueroa, E.F.; O’kAne, G.M.; Connor, A.A.; Denroche, R.E.; Grant, R.C.; McLeod, J.; et al. Author Correction: Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution. Nat. Genet. 2020, 52, 463–463. [Google Scholar] [CrossRef]
- Hwang, W.L.; Jagadeesh, K.A.; Guo, J.A.; Hoffman, H.I.; Yadollahpour, P.; Reeves, J.W.; Mohan, R.; Drokhlyansky, E.; Van Wittenberghe, N.; Ashenberg, O.; et al. Single-nucleus and spatial transcriptome profiling of pancreatic cancer identifies multicellular dynamics associated with neoadjuvant treatment. Nat. Genet. 2022, 54, 1178–1191. [Google Scholar] [CrossRef] [PubMed]
- Huffman, B.M.; Ellis, H.; Jordan, A.C.; Freed-Pastor, W.A.; Perez, K.; Rubinson, D.A.; Sethi, N.; Singh, H.; Surana, R.; Wolpin, B.M.; et al. Emerging Role of Targeted Therapy in Metastatic Pancreatic Adenocarcinoma. Cancers 2022, 14, 6223. [Google Scholar] [CrossRef]
- Kuboki, Y.; Fakih, M.; Strickler, J.; Yaeger, R.; Masuishi, T.; Kim, E.J.; Bestvina, C.M.; Kopetz, S.; Falchook, G.S.; Langer, C.; et al. Sotorasib with panitumumab in chemotherapy-refractory KRASG12C-mutated colorectal cancer: A phase 1b trial. Nat. Med. 2024, 30, 265–270. [Google Scholar] [CrossRef] [PubMed]
- Thummalapalli R, Bernstein E, Herzberg B, Li BT, Iqbal A, Preeshagul I; et al. Clinical and Genomic Features of Response and Toxicity to Sotorasib in a Real-World Cohort of Patients With Advanced KRAS G12C-Mutant Non-Small Cell Lung Cancer. JCO Precis Oncol. 2023;7:e2300030.
- 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. New Engl. J. Med. 2021, 384, 2371–2381. [Google Scholar] [CrossRef] [PubMed]
- Strickler JH, Satake H, George TJ, Yaeger R, Hollebecque A, Garrido-Laguna I; et al. Sotorasib in KRAS p.G12C-Mutated Advanced Pancreatic Cancer. N Engl J Med. 2023;388(1):33-43.
- Wang-Gillam, A.; Hubner, R.A.; Siveke, J.T.; Von Hoff, D.D.; Belanger, B.; de Jong, F.A.; Mirakhur, B.; Chen, L.-T. NAPOLI-1 phase 3 study of liposomal irinotecan in metastatic pancreatic cancer: Final overall survival analysis and characteristics of long-term survivors. Eur. J. Cancer 2019, 108, 78–87. [Google Scholar] [CrossRef] [PubMed]
- Hallin, J.; Bowcut, V.; Calinisan, A.; Briere, D.M.; Hargis, L.; Engstrom, L.D.; Laguer, J.; Medwid, J.; Vanderpool, D.; Lifset, E.; et al. Anti-tumor efficacy of a potent and selective non-covalent KRASG12D inhibitor. Nat. Med. 2022, 28, 2171–2182. [Google Scholar] [CrossRef]
- Hosein, A.N.; Dougan, S.K.; Aguirre, A.J.; Maitra, A. Translational advances in pancreatic ductal adenocarcinoma therapy. Nat. Cancer 2022, 3, 272–286. [Google Scholar] [CrossRef]
- Kawaguchi, K.; Igarashi, K.; Murakami, T.; Kiyuna, T.; Lwin, T.M.; Hwang, H.K.; Delong, J.C.; Clary, B.M.; Bouvet, M.; Unno, M.; et al. MEK inhibitors cobimetinib and trametinib, regressed a gemcitabine-resistant pancreatic-cancer patient-derived orthotopic xenograft (PDOX). Oncotarget 2017, 8, 47490–47496. [Google Scholar] [CrossRef]
- Kaplan, D.R.; Hempstead, B.L.; Martin-Zanca, D.; Chao, M.V.; Parada, L.F.; Kaplan, D.R. The trk Proto-Oncogene Product: A Signal Transducing Receptor for Nerve Growth Factor. Science 1991, 252, 554–558. [Google Scholar] [CrossRef]
- Kaplan, D.R.; Martin-Zanca, D.; Parada, L.F. Tyrosine phosphorylation and tyrosine kinase activity of the trk proto-oncogene product induced by NGF. Nature 1991, 350, 158–160. [Google Scholar] [CrossRef]
- Christenson, E.S.; Jaffee, E.; Azad, N.S. Current and emerging therapies for patients with advanced pancreatic ductal adenocarcinoma: A bright future. Lancet Oncol. 2020, 21, e135–e145. [Google Scholar] [CrossRef]
- Okamura, R.; Boichard, A.; Kato, S.; Sicklick, J.K.; Bazhenova, L.; Kurzrock, R. Analysis of NTRK Alterations in Pan-Cancer Adult and Pediatric Malignancies: Implications for NTRK-Targeted Therapeutics. JCO Precis. Oncol. 2018, 2, 1–20. [Google Scholar] [CrossRef]
- Xiang, S.; Zheng, Y.; Wang, M.; Liu, X.; Zhang, X.; Chen, D.; Meng, G.; Xu, H.; Wang, X. Comprehensive identification of NRG1 fusions in 25,203 patients with solid tumors. npj Precis. Oncol. 2025, 9, 262. [Google Scholar] [CrossRef] [PubMed]
- Heining C, Horak P, Uhrig S, Codo PL, Klink B, Hutter B; et al. NRG1 Fusions in KRAS Wild-Type Pancreatic Cancer. Cancer Discov. 2018;8(9):1087-95.
- https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-zenocutuzumab-zbco-non-small-cell-lung-cancer-and-pancreatic.
- Parimi, V.; Tolba, K.; Danziger, N.; Kuang, Z.; Sun, D.; Lin, D.I.; Hiemenz, M.C.; Schrock, A.B.; Ross, J.S.; Oxnard, G.R.; et al. Genomic landscape of 891 RET fusions detected across diverse solid tumor types. npj Precis. Oncol. 2023, 7, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Bhamidipati, D.; Yedururi, S.; Huse, J.; Chinapuvvula, S.V.; Wu, J.; Subbiah, V. Exceptional Responses to Selpercatinib in RET Fusion–Driven Metastatic Pancreatic Cancer. JCO Precis. Oncol. 2023, 7, e2300252–e2300252. [Google Scholar] [CrossRef]
- https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-selpercatinib-locally-advanced-or-metastatic-ret-fusion-positive-solid-tumors.
- Acher, A.W.; Hallet, J. Mismatch Repair, Matched with Hope? Revisiting Survival in MSI-H Pancreatic Ductal Adenocarcinoma. Ann. Surg. Oncol. 2025, 1–2. [Google Scholar] [CrossRef]
- https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-chemotherapy-biliary-tract-cancer.
- Rosen, M.N.; A Goodwin, R.; Vickers, M.M. BRCA mutated pancreatic cancer: A change is coming. World J. Gastroenterol. 2021, 27, 1943–1958. [Google Scholar] [CrossRef]
- Sahin, I.H.; Lowery, M.A.; Stadler, Z.K.; Salo-Mullen, E.; Iacobuzio-Donahue, C.A.; Kelsen, D.P.; O’reilly, E.M. Genomic instability in pancreatic adenocarcinoma: A new step towards precision medicine and novel therapeutic approaches. Expert Rev. Gastroenterol. Hepatol. 2016, 10, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Golan, T.; Hammel, P.; Reni, M.; Van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.-O.; Hochhauser, D.; Arnold, D.; Oh, D.-Y.; et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N. Engl. J. Med. 2019, 381, 317–327. [Google Scholar] [CrossRef]
- Gout, J.; Perkhofer, L.; Morawe, M.; Arnold, F.; Ihle, M.; Biber, S.; Lange, S.; Roger, E.; Kraus, J.M.; Stifter, K.; et al. Synergistic targeting and resistance to PARP inhibition in DNA damage repair-deficient pancreatic cancer. Gut 2020, 70, 743–760. [Google Scholar] [CrossRef]
- Lindemann, A.; Patel, A.A.; Silver, N.L.; Tang, L.; Liu, Z.; Wang, L.; Tanaka, N.; Rao, X.; Takahashi, H.; Maduka, N.K.; et al. COTI-2, A Novel Thiosemicarbazone Derivative, Exhibits Antitumor Activity in HNSCC through p53-dependent and -independent Mechanisms. Clin. Cancer Res. 2019, 25, 5650–5662. [Google Scholar] [CrossRef]
- Vassilev, L.T.; Vu, B.T.; Graves, B.; Carvajal, D.; Podlaski, F.; Filipovic, Z.; Kong, N.; Kammlott, U.; Lukacs, C.; Klein, C.; et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 2004, 303, 844–848. [Google Scholar] [CrossRef] [PubMed]
- Gaspar, N.J.; Li, L.; Kapoun, A.M.; Medicherla, S.; Reddy, M.; Li, G.; O’yOung, G.; Quon, D.; Henson, M.; Damm, D.L.; et al. Inhibition of Transforming Growth Factor β Signaling Reduces Pancreatic Adenocarcinoma Growth and Invasiveness. Mol. Pharmacol. 2007, 72, 152–161. [Google Scholar] [CrossRef]
- Fang, Y.-T.; Yang, W.-W.; Niu, Y.-R.; Sun, Y.-K. Recent advances in targeted therapy for pancreatic adenocarcinoma. World J. Gastrointest. Oncol. 2023, 15, 571–595. [Google Scholar] [CrossRef] [PubMed]
- Zhang Y, Morris JPt, Yan W, Schofield HK, Gurney A, Simeone DM; et al. Canonical wnt signaling is required for pancreatic carcinogenesis. Cancer Res. 2013;73(15):4909-22.
- Gurney, A.; Axelrod, F.; Bond, C.J.; Cain, J.; Chartier, C.; Donigan, L.; Fischer, M.; Chaudhari, A.; Ji, M.; Kapoun, A.M.; et al. Wnt pathway inhibition via the targeting of Frizzled receptors results in decreased growth and tumorigenicity of human tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 11717–11722. [Google Scholar] [CrossRef]
- Kung, H.; Yu, J. Targeted therapy for pancreatic ductal adenocarcinoma: Mechanisms and clinical study. Medcomm 2023, 4, e216. [Google Scholar] [CrossRef]
- Venkatasubbarao, K.; Peterson, L.; Zhao, S.; Hill, P.; Cao, L.; Zhou, Q.; Nawrocki, S.T.; Freeman, J.W. Inhibiting signal transducer and activator of transcription-3 increases response to gemcitabine and delays progression of pancreatic cancer. Mol. Cancer 2013, 12, 104–104. [Google Scholar] [CrossRef]
- De Jesus-Acosta, A.; Laheru, D.; Maitra, A.; Arcaroli, J.; Rudek, M.A.; Dasari, A.; Blatchford, P.J.; Quackenbush, K.; Messersmith, W. A phase II study of the gamma secretase inhibitor RO4929097 in patients with previously treated metastatic pancreatic adenocarcinoma. Investig. New Drugs 2014, 32, 739–745. [Google Scholar] [CrossRef]
- Hu, Z.I.; Bendell, J.C.; Bullock, A.; LoConte, N.K.; Hatoum, H.; Ritch, P.; Hool, H.; Leach, J.W.; Sanchez, J.; Sohal, D.P.S.; et al. A randomized phase II trial of nab-paclitaxel and gemcitabine with tarextumab or placebo in patients with untreated metastatic pancreatic cancer. Cancer Med. 2019, 8, 5148–5157. [Google Scholar] [CrossRef]
- Leroux, C.; Konstantinidou, G. Targeted Therapies for Pancreatic Cancer: Overview of Current Treatments and New Opportunities for Personalized Oncology. Cancers 2021, 13, 799. [Google Scholar] [CrossRef]
- Provenzano, P.P.; Cuevas, C.; Chang, A.E.; Goel, V.K.; Von Hoff, D.D.; Hingorani, S.R. Enzymatic Targeting of the Stroma Ablates Physical Barriers to Treatment of Pancreatic Ductal Adenocarcinoma. Cancer Cell 2012, 21, 418–429. [Google Scholar] [CrossRef] [PubMed]
- Rémond, M.; Pellat, A.; Brezault, C.; Dhooge, M.; Coriat, R. Are targeted therapies or immunotherapies effective in metastatic pancreatic adenocarcinoma? ESMO Open 2022, 7, 100638. [Google Scholar] [CrossRef] [PubMed]
- Aung KL, Fischer SE, Denroche RE, Jang GH, Dodd A, Creighton S; et al. Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial. Clin Cancer Res. 2018, 24, 1344–1354. [Google Scholar] [CrossRef] [PubMed]
| Reference, year | Subtypes | Relevant clinical infomation |
|---|---|---|
| Collisson et al., 2011 | Classical Quasi-mesenchymal Exocrine-like |
Best survival, more sensitive to erlotinib Poorest prognosis, more sensitive to gemcitabine High expression of genes related to digestive enzymes |
| Moffitt et al., 2015 | Classical Basal-like |
Resemble the classical group from Collisson et al. Poor prognosis |
| Bailey et al., 2016 | Squamous Pancreatic progenitor Aberrantly differentiated endocrine exocrine (ADEX) Immunogenic |
Frequent TP53 mutations, worse survival Association with transcriptional factors and metabolic pathways Upregulation of genes involved in KRAS activation and endocrine and exocrine differentiation Upregulated immune network |
| Cancer Genome Atlas Research Network, 2017 | SNF-1 SNF-2 |
Poor prognosis Better prognosis |
| Puleo et al., 2018 | Pure basal-like Stroma-activated Desmoplastic Pure classical Immune classical based |
|
| Chan-Seng-Yue, 2020 | Basal-like A Basal-Like B Hybrid Classical A and B |
Advanced disease, worst response to gemcitabine and FOLFIRINOX Resectable disease Resectable disease GATA6 amplification, complete SMAD4 loss |
| High frequency alterations | FDA approved targeted therapy (indication) |
|---|---|
|
CDKN2A/p16 KRAS TP53 SMAD4/DPC4 |
Sotorasib – Adagrasib (G12C variant) |
| Low frequency alterations | Targeted therapy |
|
AKT2 BRAF BRCA1/2 NRG1 NTRK RET STK11/LKB1 Microsatellite Instability |
Encorafenib (V600E variant) Olaparib (germline mutation) Zenocutuzumab (gene fusion) Larotrectinib (gene fusion) Selpercatinib (gene fusion) Pembrolizumab (MSI-H or dMMR) |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
