Published online Nov 15, 2025. doi: 10.4251/wjgo.v17.i11.111264
Revised: July 13, 2025
Accepted: September 24, 2025
Published online: November 15, 2025
Processing time: 140 Days and 10.4 Hours
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal ma
Core Tip: This review highlights the transformative impact of precision medicine in pancreatic ductal adenocarcinoma by focusing on clinically relevant genomic and molecular alterations such as Kirsten rat sarcoma viral oncogene homolog and breast cancer susceptibility gene mutations, rare actionable fusions, and co-mutational signatures. It emphasizes the importance of integrating routine molecular profiling into clinical workflows to guide targeted therapies and personalized treatment strategies.
- Citation: Li X, Jiao Y, Liu YH. Precision medicine advances in pancreatic cancer driven by genomic and molecular alterations. World J Gastrointest Oncol 2025; 17(11): 111264
- URL: https://www.wjgnet.com/1948-5204/full/v17/i11/111264.htm
- DOI: https://dx.doi.org/10.4251/wjgo.v17.i11.111264
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a dismal five-year survival rate of less than 10%. Despite modest advances in surgical and chemotherapeutic approaches, overall outcomes remain poor, especially in advanced stages[1]. In recent years, the advent of precision medicine - driven by high-throughput genomic sequencing and integrated molecular characterization - has led to a paradigm shift in oncology[2]. In PDAC, while challenges persist due to its complex tumor microenvironment and low immunogenicity, actionable genomic and molecular alterations are being increasingly recognized as important tools to guide targeted therapies and individualized treatment strategies (Table 1).
| Gene/alteration | Prevalence in PDAC | Associated targeted therapy/clinical implication |
| KRAS (G12D, G12V, G12R) | Approximately 80%-90% | Allele-specific inhibitors under development; RNAi strategies in preclinical models |
| KRAS G12C | < 2% | Sotorasib, adagrasib (limited efficacy in PDAC) |
| BRCA1/2 | Approximately 4%-7% | PARP inhibitors (e.g., olaparib); platinum-based chemotherapy |
| BRAF (V600) | < 1% | BRAF/MEK inhibitors (e.g., dabrafenib + trametinib; off-label use) |
| NTRK fusions | < 1% | TRK inhibitors (larotrectinib, entrectinib) |
| FGFR2 fusions | Rare | FGFR inhibitors (e.g., pemigatinib; in clinical trials) |
| RET fusions | Rare | RET inhibitors (e.g., selpercatinib; in clinical trials) |
| dMMR/MSI-H | < 2% | Immune checkpoint inhibitors (e.g., pembrolizumab) |
| High TMB | Rare | Potential responsiveness to ICIs |
| TP53/CDKN2A/SMAD4 (co-mutations) | > 50% | Prognostic markers; may influence resistance and response to combination therapies |
Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations are present in approximately 80%-90% of PDAC cases, as identified in multiple cohort studies including a large-scale analysis of 1000+ patients from The Cancer Genome Atlas[3,4]. These alterations, predominantly at codon 12, including G12D, G12V, and G12R, drive constitutive activation of the Ras-mitogen-activated protein kinase signaling pathway, promoting tumor growth, survival, and resistance to therapy. KRAS mutations are strongly associated with poor prognosis, and certain variants such as G12D and G12V correlate with worse overall survival compared to wild-type KRAS, while G12R may be linked to relatively better outcomes[5,6]. In addition to primary oncogenic activation, emerging data indicate that resistance to KRAS-targeted therapies can arise from secondary mutations in KRAS (e.g., Y96D), amplification of downstream effectors such as v-raf murine sarcoma viral oncogene homolog B1 (BRAF), or activation of bypass signaling through the phosphatidylinositide 3-kinases-protein kinase B-mammalian target of rapamycin pathway, thereby diminishing the efficacy of KRAS inhibition strategies.
Despite their prevalence, KRAS mutations have historically been considered “undruggable” due to the protein’s small, smooth surface and high affinity for guanosine triphosphate. However, recent breakthroughs have introduced novel strategies targeting KRAS, particularly the G12C variant. While KRAS G12C-specific inhibitors like sotorasib and adagrasib have shown promising efficacy in non-small cell lung cancer, their applicability in PDAC is limited due to the rarity of this variant in pancreatic tumors[4]. Meanwhile, innovative approaches such as exosome-mediated delivery of KRAS-targeted small interfering RNA and short hairpin RNA have demonstrated preclinical potential in suppressing KRAS-driven tumorigenesis[7].
Breast cancer susceptibility gene 1 (BRCA1) and BRCA2 mutations, although less frequent, represent one of the most actionable targets in PDAC. These alterations are detected in approximately 4%-7% of patients and are more common in those with a family history of breast, ovarian, or pancreatic cancer[8]. Loss-of-function mutations in BRCA genes com
This genomic vulnerability can be therapeutically exploited using platinum-based chemotherapy and poly(ADP-ribose) polymerase (PARP) inhibitors such as olaparib. Clinical studies have shown that BRCA-mutant pancreatic cancers demonstrate improved response rates and prolonged progression-free survival with these agents[9,10]. Moreover, the tumor-agnostic efficacy of PARP inhibitors across BRCA-mutated malignancies has reinforced the principle of biomarker-guided treatment and the importance of routine germline testing in patients with PDAC.
In addition to KRAS and BRCA mutations, a subset of PDAC tumors harbor other potentially actionable genomic alterations. BRAF (V600) mutations, although rare, may respond to combination therapies targeting the mitogen-activated protein kinase pathway[11]. Neurotrophic receptor tyrosine kinase gene fusions have also emerged as important pan-cancer biomarkers, with Food and Drug Administration-approved tropomyosin receptor kinase inhibitors such as larotrectinib and entrectinib demonstrating tumor-agnostic efficacy, including in rare PDAC cases[12]. Similarly, fusions involving fibroblast growth factor receptor 2 and RET, though infrequent, are targetable with selective kinase inhibitors currently being evaluated in clinical trials[4].
The rarity of these mutations necessitates broad molecular profiling to identify eligible patients. With the growing availability of multiplex sequencing and basket trials, even low-prevalence alterations are becoming actionable within individualized treatment frameworks. These findings support the routine implementation of next-generation sequencing in clinical practice, particularly for patients with advanced or treatment-refractory disease.
Pancreatic cancer is generally considered immunologically “cold” due to its low tumor mutational burden (TMB), dense stromal reaction, and immunosuppressive microenvironment. Nevertheless, a small subset of PDAC cases exhibit high TMB or mismatch repair deficiency, making them more likely to respond to immune checkpoint inhibitors such as anti-programmed death protein 1/programmed death ligand-1 agents[13]. While the prevalence of TMB-high or microsatellite instability-high (MSI-H) PDAC is low (< 2%), patients in this category may derive substantial clinical benefit from immune checkpoint inhibitors.
Efforts are ongoing to convert immunologically inert tumors into immune checkpoint inhibitor-responsive ones through combination approaches that include chemotherapy, radiation, or stromal modulation. Nonetheless, the use of immunotherapy in PDAC remains largely limited to biomarker-selected populations. Moreover, the dense desmoplastic stroma and extracellular matrix in PDAC act as physical and biochemical barriers, impeding T-cell infiltration and reducing drug penetration, thereby contributing to immunotherapy resistance even in TMB-high or microsatellite in
The co-occurrence of mutations in key tumor suppressor genes - such as tumor protein p53 (TP53), cyclin-dependent kinase inhibitor 2A, and SMAD4 - alongside KRAS mutations, is common in PDAC and carries significant prognostic implications. TP53 mutations, for example, are associated with enhanced genomic instability and tumor progression. The concurrent presence of KRAS and TP53 mutations correlates with more aggressive disease and worse overall survival[6,14]. In contrast, specific combinations of genetic alterations may identify subsets of patients who are more likely to benefit from particular therapies or trial enrollment. Understanding these co-mutational signatures not only enhances prognostic stratification but also facilitates the design of tailored combination regimens. Future studies may refine risk models based on integrated genomic profiling, guiding more personalized treatment decisions.
Although many genomic alterations found in PDAC are also present in other cancers, their therapeutic relevance is context-dependent. For instance, KRAS G12C inhibitors have demonstrated efficacy in non-small cell lung cancer but limited activity in PDAC, possibly due to different downstream signaling dynamics and tumor microenvironments. Similarly, BRCA mutations are a well-established therapeutic target in breast and ovarian cancers, but their impact in PDAC has only recently been recognized. These differences underscore the importance of pancreas-specific translational studies, rather than extrapolating directly from other tumor types. Nevertheless, the concept of tumor-agnostic treatment based on shared genomic drivers continues to gain traction, particularly in the context of Food and Drug Administration-approved indications for agents like PARP inhibitors and tropomyosin receptor kinase inhibitors.
As precision oncology evolves in PDAC, routine germline and somatic testing is critical for identifying actionable targets and guiding therapy. Beyond genomic alterations, epigenomics, proteomics, and metabolomics offer new insights into drug response and resistance, enabling personalized treatment strategies. Despite progress, real-world implementation of molecular profiling is hindered by cost, reimbursement issues, and limited infrastructure, particularly outside academic centers. Overcoming these barriers is essential for equitable access to precision medicine. Liquid biopsy techniques like circulating tumor DNA and exosomal RNA are promising for monitoring tumor evolution, minimal residual disease, and early relapse. Methylation assays are also being explored for early detection. Proteolysis-targeting chimeras (PROTACs), particularly for KRAS G12C degradation, represent a new frontier. Recent preclinical studies have demonstrated the feasibility of PROTAC-based KRAS degradation using von Hippel-Lindau-recruiting PROTACs like YF135[15], with efforts underway to develop BRCA-targeting PROTACs for homologous recombination-deficient tumors. Ultimately, improving outcomes in pancreatic cancer requires integrating molecular data with clinical features and addressing resistance mechanisms.
Incorporating genomic and molecular profiling into the management of PDAC has opened new avenues for personalized therapy, offering hope in a disease historically characterized by limited treatment responsiveness and poor survival. KRAS and BRCA mutations remain the most clinically relevant alterations, guiding the development of targeted the
| 1. | Stoop TF, Javed AA, Oba A, Koerkamp BG, Seufferlein T, Wilmink JW, Besselink MG. Pancreatic cancer. Lancet. 2025;405:1182-1202. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 35] [Article Influence: 35.0] [Reference Citation Analysis (0)] |
| 2. | George B. Precision Medicine and Pancreatic Cancer. Surg Oncol Clin N Am. 2021;30:693-708. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 3. | Lee B, Tian E, Croagh D, Harris M, Thomson B, Michael M, Loveday B, Mclachlan S, Knowles B, Shapiro JD, Wong R, Fox A, Jennens R, Yong T, Hodgson R, Houli N, Ananda S, Lipton LR, Mendis SR, Gibbs P. Mutated KRASas a promising target in pancreas cancer: PURPLE registry data to inform real-world incidence and prognostic significance and to aid trial recruitment. J Clin Oncol. 2024;42:11150-11150. [DOI] [Full Text] |
| 4. | Lee JK, Sivakumar S, Schrock AB, Madison R, Fabrizio D, Gjoerup O, Ross JS, Frampton GM, Napalkov P, Montesion M, Schutzman JL, Ye X, Hegde PS, Nagasaka M, Oxnard GR, Sokol ES, Ou SI, Shi Z. Comprehensive pan-cancer genomic landscape of KRAS altered cancers and real-world outcomes in solid tumors. NPJ Precis Oncol. 2022;6:91. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 90] [Reference Citation Analysis (0)] |
| 5. | Norton C, Shaw MS, Rubnitz Z, Smith J, Soares HP, Nevala-Plagemann CD, Garrido-Laguna I, Florou V. KRAS Mutation Status and Treatment Outcomes in Patients With Metastatic Pancreatic Adenocarcinoma. JAMA Netw Open. 2025;8:e2453588. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 6. | Yousef A, Yousef M, Chowdhury S, Abdilleh K, Knafl M, Edelkamp P, Alfaro-munoz K, Chacko R, Peterson J, Smaglo B, Wolff R, Pant S, Lee M, Willis J, Overman M, Doss S, Matrisian L, Hurd M, Snyder R, Katz M, Wang H, Maitra A, Shen JP, Zhao D. Abstract C098: Impact of KRAS mutations and co-mutations on clinical outcomes in pancreatic ductal adenocarcinoma. Cancer Res. 2024;84:C098. [DOI] [Full Text] |
| 7. | Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, Lee JJ, Kalluri R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546:498-503. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1167] [Cited by in RCA: 1956] [Article Influence: 244.5] [Reference Citation Analysis (1)] |
| 8. | Golan T, Kanji ZS, Epelbaum R, Devaud N, Dagan E, Holter S, Aderka D, Paluch-Shimon S, Kaufman B, Gershoni-Baruch R, Hedley D, Moore MJ, Friedman E, Gallinger S. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. Br J Cancer. 2014;111:1132-1138. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 259] [Cited by in RCA: 344] [Article Influence: 31.3] [Reference Citation Analysis (0)] |
| 9. | Kaufman B, Shapira-Frommer R, Schmutzler RK, Audeh MW, Friedlander M, Balmaña J, Mitchell G, Fried G, Stemmer SM, Hubert A, Rosengarten O, Steiner M, Loman N, Bowen K, Fielding A, Domchek SM. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol. 2015;33:244-250. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1153] [Cited by in RCA: 1337] [Article Influence: 121.5] [Reference Citation Analysis (0)] |
| 10. | Showalter SL, Charles S, Belin J, Cozzitorto J, Einstein P, Richards NG, Sauter PK, Kennedy EP, Witkiewicz A, Brody JR, Yeo CJ. Identifying pancreatic cancer patients for targeted treatment: the challenges and limitations of the current selection process and vision for the future. Expert Opin Drug Deliv. 2010;7:273-284. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 12] [Cited by in RCA: 14] [Article Influence: 0.9] [Reference Citation Analysis (0)] |
| 11. | Aguirre AJ, Nowak JA, Camarda ND, Moffitt RA, Ghazani AA, Hazar-Rethinam M, Raghavan S, Kim J, Brais LK, Ragon D, Welch MW, Reilly E, McCabe D, Marini L, Anderka K, Helvie K, Oliver N, Babic A, Da Silva A, Nadres B, Van Seventer EE, Shahzade HA, St Pierre JP, Burke KP, Clancy T, Cleary JM, Doyle LA, Jajoo K, McCleary NJ, Meyerhardt JA, Murphy JE, Ng K, Patel AK, Perez K, Rosenthal MH, Rubinson DA, Ryou M, Shapiro GI, Sicinska E, Silverman SG, Nagy RJ, Lanman RB, Knoerzer D, Welsch DJ, Yurgelun MB, Fuchs CS, Garraway LA, Getz G, Hornick JL, Johnson BE, Kulke MH, Mayer RJ, Miller JW, Shyn PB, Tuveson DA, Wagle N, Yeh JJ, Hahn WC, Corcoran RB, Carter SL, Wolpin BM. Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine. Cancer Discov. 2018;8:1096-1111. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 173] [Cited by in RCA: 271] [Article Influence: 38.7] [Reference Citation Analysis (0)] |
| 12. | Reddy NK, Subbiah V. Redefining pancreatic cancer management with tumor-agnostic precision medicine. Carcinogenesis. 2024;45:836-844. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 13. | Quintanilha JCF, Storandt MH, Graf RP, Li G, Keller R, Lin DI, Ross JS, Huang RSP, Schrock AB, Oxnard GR, Chakrabarti S, Mahipal A. Tumor Mutational Burden in Real-World Patients With Pancreatic Cancer: Genomic Alterations and Predictive Value for Immune Checkpoint Inhibitor Effectiveness. JCO Precis Oncol. 2023;7:e2300092. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 16] [Cited by in RCA: 24] [Article Influence: 12.0] [Reference Citation Analysis (0)] |
| 14. | Ghosh P, George R, Singh N, Agrawal S. Abstract 2545: KRAS co-mutational landscape dictates treatment outcomes in pancreatic cancer (PC) patients. Cancer Res. 2024;84:2545. [DOI] [Full Text] |
| 15. | Yang F, Wen Y, Wang C, Zhou Y, Zhou Y, Zhang ZM, Liu T, Lu X. Efficient targeted oncogenic KRAS(G12C) degradation via first reversible-covalent PROTAC. Eur J Med Chem. 2022;230:114088. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 8] [Cited by in RCA: 64] [Article Influence: 21.3] [Reference Citation Analysis (0)] |
