Preprint
Review

This version is not peer-reviewed.

Molecular Pathology of Pancreatic Ductal Adenocarcinoma

A peer-reviewed article of this preprint also exists.

Submitted:

29 September 2025

Posted:

30 September 2025

You are already at the latest version

Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer that frequently presents at an advanced stage with limited effective treatment options and a dismal prognosis. It is a highly heterogenous disease driven by various genetic and epigenetic alterations. Recent advances in sequencing modalities have significantly improved our understanding of the genetics of PDAC, which could lead to promising and novel therapeutic strategies. In this review, we summarize the most up-to-date literature on the molecular landscape of PDAC and its precursor lesions, and recent advances in targeted therapy.
Keywords: 
;  ;  ;  ;  ;  ;  ;  

1. Introduction

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy originating from the ductal epithelial cells of the exocrine pancreas. It ranks among the deadliest forms of cancer and is projected to become the second leading cause of cancer-related mortality by 2030 [1,2]. Due to the absence of specific clinical symptoms and the lack of reliable diagnostic biomarkers for early detection, the majority of patients are diagnosed at an advanced stage, often with locally advanced or metastatic disease that is not amenable to surgical resection. Compounding this challenge, PDAC is commonly resistant to most currently available therapeutic agents. In response, considerable research efforts have focused on elucidating the molecular mechanisms driving PDAC, with the goal of developing effective, targeted therapies. In this review, we highlight key genetic studies that have shaped our current understanding of PDAC pathogenesis.

2. Genetic Alterations in PDAC

Both conventional studies and more recent next-generation sequencing (NGS) analyses have revealed that PDAC exhibits a highly heterogeneous molecular landscape. Among the most frequently altered genes are KRAS (Kirsten rat sarcoma viral oncogene), TP53 (tumor suppressor gene), SMAD4 (also known as Deleted in Pancreatic Cancer-4, DPC4), and CDKN2A (cyclin-dependent kinase inhibitor 2A) [3].
Activating mutations in KRAS, particularly at codons 12, 13, or 61, are found in over 90% of PDACs, with KRASG12D and KRASG12V accounting for 39.2% and 32.5% of all KRAS mutations, respectively [4]. KRAS encodes a small GTPase protein that functions as a molecular switch in signal transduction. When mutated, it constitutively activates several downstream signaling pathways—including the RAF–mitogen-activated protein kinase (MAPK), Ral guanine nucleotide dissociation stimulator (RalGDS), and phosphoinositide 3-kinase (PI3K) pathways—driving key oncogenic processes such as cancer cell proliferation, apoptosis resistance, migration, and metastasis [3]. KRAS mutations are also detected in early precursor lesions of PDAC, including low-grade pancreatic intraepithelial neoplasia (PanINs) and intraductal papillary mucinous neoplasms (IPMNs), underscoring their role in the initiation of neoplastic transformation [5]. Inactivating mutations in TP53 are present in approximately 50% to 80% of PDAC cases [6,7,8]. The TP53 gene encodes the tumor suppressor protein p53, a key regulator of cellular responses to genotoxic stress. p53 maintains genomic stability by inducing cell cycle arrest, DNA repair, or apoptosis in response to damage [9]. Loss of p53 function contributes to tumor progression and has also been implicated in promoting epithelial-to-mesenchymal transition (EMT) via upregulation of ZEB1, a zinc-finger transcription factor that suppresses epithelial gene expression and enhances cell motility, migration, and metastatic potential [10]. The tumor suppressor gene CDKN2A (also known as p16INK4a) is altered in approximately 95% of PDACs through mechanisms such as homozygous deletion, intragenic mutation, or promoter hypermethylation [11,12]. p16 inhibits CDK4/6-mediated phosphorylation of the retinoblastoma (RB) protein, thereby regulating cell cycle progression and preventing premature entry into the S phase. Inactivation of p16 disrupts this control, accelerating cellular proliferation [13]. Germline truncating mutations in CDKN2A, such as E119X and Q50X, have recently been identified in patients with familial “pancreatic cancer plus melanoma syndrome,” a rare cancer predisposition syndrome associated with increased risk of pancreatic cancer and malignant melanoma [14]. Inactivating mutations or deletions in SMAD4 (also known as DPC4), another key tumor suppressor gene, are observed in 30% to 60% of PDACs [3,6,15]. SMAD4 encodes a central mediator of the transforming growth factor-beta (TGF-β) signaling pathway, which normally functions to inhibit cell proliferation and maintain tissue homeostasis. Disruption of this pathway results in unchecked cellular growth and contributes to tumor progression [16]. Similar to KRAS, loss-of-function alterations in TP53 and SMAD4 are also observed in PDAC precursor lesions, though they typically emerge later during the progression to high-grade dysplasia [17].
In addition to the commonly mutated driver genes (KRAS, TP53, CDKN2A, and SMAD4), a variety of less frequent genetic alterations have been identified in PDAC. These include gene amplifications involving CMYC (chromosome 8q), MYB (chromosome 6q), AIB1/NCOA3 (chromosome 20q), EGFR (chromosome 7p), and GATA6 [18]. Germline mutations in genes associated with the Fanconi anemia (FA) DNA repair pathway—such as BRCA1, BRCA2, and PALB2—impair the cellular response to DNA damage, leading to the accumulation of genomic instability. These mutations are present in approximately 5–7% of patients with PDAC [19]. ATM germline mutations, which also disrupt DNA damage response, represent the second most common inherited alteration in PDAC and are found in about 6% of cases [20]. Alterations in chromatin-remodeling genes, particularly those encoding components of the SWI/SNF (switch/sucrose non-fermentable) complex, have also been implicated in PDAC [21]. For example, ARID1A (AT-rich interaction domain 1A) is a chromatin regulatory protein involved in DNA repair and cell cycle regulation. Loss-of-function mutations in ARID1A have been shown to promote pancreatic tumorigenesis through activation of multiple downstream oncogenic pathways [22]. Recent genomic studies have also identified mutations in genes involved in RNA splicing. Mutations in SF3B1, for instance, disrupt spliceosome recognition of pre-mRNA, contributing to abnormal splicing patterns [23]. These defects may play a role in tumor initiation and progression by promoting more aggressive disease phenotypes and facilitating metastasis [24].
Several histologic variants of PDAC are associated with specific genetic alterations. Colloid carcinoma, which often arises from intestinal-type intraductal papillary mucinous neoplasms (IPMNs), is frequently driven by GNAS mutations [25]. Adenosquamous carcinoma, a rare and aggressive variant of PDAC, commonly harbors somatic mutations in UPF1[26], a gene involved in nonsense-mediated mRNA decay (NMD)—a cellular mechanism that eliminates mRNAs with premature stop codons. Dysfunction of UPF1 may allow the accumulation of aberrant, toxic transcripts, potentially contributing to tumorigenesis [27]. However, the significance of UPF1 mutations in adenosquamous carcinoma remains controversial; for instance, a study by Polaski et al. found no significant impact of UPF1 mutations on tumor growth or squamous differentiation in human and murine models [28]. This subtype may also exhibit canonical mutations in KRAS, TP53, CDKN2A/p16, and SMAD4, as well as amplification of the MYC oncogene [29,30]. Medullary carcinoma, another rare PDAC subtype, may display microsatellite instability (MSI), similar to medullary colorectal cancers. It is typically associated with a lower frequency of KRAS mutations [31] and can occur in individuals with Lynch syndrome or germline mutations in mismatch repair (MMR) genes [32].

3. Genetic Alterations in Precursor Lesions of PDAC

PDAC often arises from a spectrum of noninvasive precursor lesions, including pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasm (IPMN), intraductal oncocytic papillary neoplasm (IOPN), mucinous cystic neoplasm (MCN), and intraductal tubulopapillary neoplasm (ITPN).
Feldmann et al. proposed a chronological classification of molecular alterations in PanINs into early, intermediate, and late events. KRAS mutations, telomere shortening, and p21 upregulation were observed across all PanIN grades, indicating their role as early events in tumorigenesis. In contrast, alterations in TP53, SMAD4, and BRCA2 were predominantly associated with high-grade PanINs, suggesting they represent later events in the progression toward invasive PDAC [33].
Recent advances in molecular and sequencing technologies have greatly expanded our understanding of the molecular biology of IPMNs. Somatic KRAS mutations are among the earliest genetic alterations in IPMN pathogenesis, occurring in approximately 60–80% of cases [25]. These mutations are most commonly associated with the pancreatobiliary subtype and are least frequent in the intestinal subtype [34]. IPMNs also frequently harbor somatic mutations in GNAS, an oncogene located on chromosome 20q that encodes the stimulatory alpha subunit of the G-protein (Gαs). Mutations at codon 201 of GNAS are identified in about 60% of IPMNs and are more prevalent in higher-grade lesions [34]. In contrast to KRAS, GNAS mutations are most commonly observed in the intestinal subtype of IPMN. Another gene frequently altered in IPMNs is RNF43, which encodes an E3 ubiquitin ligase that negatively regulates the Wnt signaling pathway by promoting the ubiquitination and degradation of Frizzled family Wnt receptors. Somatic mutations in RNF43 are found in approximately 24% of IPMNs [35,36]. Later-stage genetic alterations, such as inactivating mutations in TP53 and SMAD4, are typically associated with high-grade dysplasia and the transition to invasive carcinoma [37].
IOPN is a recently characterized, distinct subtype of pancreatic intraductal neoplasm. Unlike IPMNs, IOPNs typically lack the common mutations found in IPMNs but instead exhibit recurrent somatic alterations in genes such as ARHGAP26, ASXL1, EPHA8, and ERBB4 [38]. Notably, a subset of IOPNs has been found to harbor DNAJB1-PRKACA gene fusions, a genetic alteration originally identified in fibrolamellar hepatocellular carcinoma [39].
Activating mutations in codon 12 of KRAS are detected in approximately 50% of MCNs [40]. Mutations in RNF43 are also frequently observed in MCNs, particularly in high-grade and invasive lesions, occurring in 56% of such cases compared to 33% in low-grade lesions [41]. Unlike IPMNs, GNAS mutations have not been reported in MCNs [42,43]. Additionally, alterations in CDKN2A, TP53, and SMAD4 are commonly associated with MCNs exhibiting high-grade dysplasia [44,45].
ITPN is molecularly distinct from other pancreatic precursor lesions, with KRAS and TP53 mutations being notably rare [46]. In a study by Basturk et al. analyzing 22 ITPN cases through targeted next-generation or whole-exome sequencing, approximately 25% of tumors harbored CDKN2A mutations. Mutations in chromatin remodeling genes—including MLL1, MLL2, MLL3, BAP1, PBRM1, EED, and ATRX—were identified in about one-third of cases. Additionally, 27% of ITPNs carried mutations in the PI3K pathway, such as PIK3CA [47]. Notably, around 20% of ITPNs harbor FGFR2 gene fusions with various fusion partners, representing a promising therapeutic target. FGFR2 inhibitors like pemigatinib and infigratinib have already been FDA-approved for treating intrahepatic cholangiocarcinoma, highlighting potential treatment avenues for ITPN [48].

4. Transcriptomic PDAC Subtypes

PDAC has recently been the focus of intensive transcriptomic analysis, leading to important insights into its molecular subtypes. A landmark study by Collisson et al. was the first to classify PDAC based on transcriptomic data derived from patient-derived cell lines. Their analysis identified three distinct subtypes: classical, quasi-mesenchymal (QM), and exocrine-like—each with unique gene expression profiles, clinical outcomes, and therapeutic sensitivities. The classical subtype exhibited high expression of epithelial and adhesion-associated genes, was linked to the best overall survival, and showed predicted sensitivity to erlotinib, an EGFR inhibitor [49]. In contrast, the QM subtype was characterized by elevated expression of mesenchymal genes, associated with the poorest prognosis, but showed greater sensitivity to gemcitabine, a standard chemotherapeutic agent. The exocrine-like subtype displayed the highest expression of genes involved in digestive enzyme production, although its clinical relevance has been debated [49].
By computationally removing the transcriptional signals contributed by stromal and immune cells from bulk RNA-seq data, Moffitt et al. identified two tumor-specific subtypes of PDAC: classical and basal-like. The classical subtype closely resembled the classical group previously described by Collisson et al., while the basal-like subtype shared molecular features with basal-like breast carcinoma and was associated with a poorer prognosis [50]. In addition to tumor subtypes, Moffitt et al. also defined two distinct stromal subtypes: normal stroma and activated stroma. The normal stroma was enriched for pancreatic stellate cell markers, such as desmin, smooth muscle actin, and vimentin. In contrast, the activated stroma showed high expression of macrophage-associated genes (ITGAM, CCL13, CCL18) and other genes linked to tumor progression, including SPARC, WNT2, WNT5A, MMP9, and MMP11 [50]. Importantly, the combination of tumor and stromal subtypes had prognostic significance. Patients with the classical tumor subtype and activated stroma had significantly worse survival compared to those with classical tumors and normal stroma. However, in tumors of the basal-like subtype, stromal classification did not significantly impact survival, suggesting that tumor-intrinsic features play a dominant role in determining prognosis in these cases [50].
A comprehensive genomic analysis of 456 PDACs by Bailey et al. identified four distinct molecular subtypes: squamous, pancreatic progenitor, aberrantly differentiated endocrine exocrine (ADEX), and immunogenic [51]. The squamous subtype (31%) was associated with the poorest survival and characterized by genetic alterations in the regulation of inflammation, hypoxia response, and TGF-β signaling. It showed upregulation of TP63ΔN and frequent TP53 mutations. The pancreatic progenitor subtype (19%) featured altered expression of transcription factors involved in early pancreatic development, including PDX1, MNX1, and FOXA2/3. It also exhibited enrichment in metabolic pathways such as fatty acid oxidation and drug metabolism [51]. The ADEX subtype (21%), considered a subclass of the progenitor group, showed upregulation of gene networks involved in KRAS activation, as well as genes related to both exocrine differentiation (NR5A2, MIST1, RBPJL) and endocrine differentiation (NEUROD1, INS, NKX2-2, and MODY-related genes). The immunogenic subtype (29%) was distinguished by significant immune cell infiltration, particularly CD4+ and CD8+ T cells, along with upregulation of immune-related pathways. These included B cell signaling, Toll-like receptor signaling, antigen presentation, and immune checkpoint pathways such as PD-L1 and CTLA-4, suggesting an immunosuppressive tumor microenvironment [51]. These molecular subtypes also correlated with distinct histopathological features. The squamous subtype was often associated with adenosquamous carcinoma; the progenitor and immunogenic subtypes were linked to colloid carcinomas and carcinomas arising from intraductal papillary mucinous neoplasms (IPMNs); and the ADEX subtype was aligned with rare acinar cell carcinomas [51].
Puleo et al. validated the basal-like and classical tumor subtypes previously identified by Moffitt et al., and further expanded upon this classification by incorporating gene expression patterns from the tumor microenvironment. This allowed them to define five distinct PDAC subtypes: pure basal-like, stroma-activated, desmoplastic, pure classical, and immune classical [52]. These subtypes showed strong correspondence with most of the molecular categories proposed by Bailey et al., with one notable exception: Bailey’s ADEX subtype. Puleo et al. suggested that ADEX was likely an artifact resulting from contamination with transcripts from normal pancreatic acinar cells, rather than representing a true tumor-specific subtype.
An integrated analysis of mRNA, miRNA, lncRNA, and DNA methylation profiles from 150 PDAC samples by the Cancer Genome Atlas (TCGA) Research Network identified only two robust molecular subtypes: SNF-1 and SNF-2, based on Similarity Network Fusion (SNF) clustering. [53]. The SNF-1 subtype corresponded to the basal-like subtype described by Moffitt et al., the squamous subtype in Bailey et al.’s classification, and the quasi-mesenchymal (QM) subtype in Collisson et al.’s study. This group was characterized by the expression of basal markers and was associated with poor prognosis. In contrast, the SNF-2 subtype aligned with the classical subtypes identified by both Moffitt et al. and Collisson et al., as well as the pancreatic progenitor subtype described by Bailey et al. SNF-2 tumors showed expression of differentiated ductal markers and were associated with more favorable clinical outcomes. Other subtypes proposed in earlier classifications, such as ADEX and immunogenic, were found to have low neoplastic cellularity in the TCGA analysis. This suggests that their molecular signatures may have been heavily influenced by stromal or normal pancreatic tissue contamination, rather than representing true tumor-intrinsic profiles [53].
Recently, Chan-Seng-Yue et al. conducted comprehensive whole-genome and transcriptome analyses on purified tumor cells from 314 primary and metastatic PDAC patients, identifying five distinct molecular subtypes: basal-like A and B (corresponding to the previously defined basal-like subtype), hybrid, and classical A and B (related to the classical subtype). Patients with basal-like A tumors typically presented with advanced disease and exhibited the poorest response to gemcitabine-based chemotherapy and FOLFIRINOX. In contrast, those with basal-like B and hybrid tumors were more often diagnosed with resectable disease. These subtypes are linked to specific genomic alterations: classical A and B tumors showed frequent GATA6 amplification and complete loss of SMAD4, while basal-like A and B tumors were characterized by complete loss of CDKN2A and a higher incidence of TP53 mutations. The hybrid subtype, marked by the presence of multiple expression signatures, did not align consistently with previously established classification systems. Importantly, single-cell analysis revealed that basal-like and classical subtype cells can coexist within the same tumor, underscoring the significant intratumoral molecular heterogeneity in PDAC [54].
Consensus has largely formed around two primary PDAC transcriptional subtypes—classical and basal-like—while leaving room for further subclassification. Notably, different subtypes can coexist within individual tumors. For example, using single-nucleus RNA sequencing and whole digital spatial transcriptome profiling of 43 primary PDAC samples (18 untreated and 25 treated), Hwang et al. identified a treatment-enriched subtype [55]. They found that the neural-like progenitor (NRP) malignant cell program was enriched in residual tumors following chemoradiation therapy. NRP cells were linked to treatment resistance and poor survival, driven by the regulation of genes involved in inhibiting cell death and chemotherapy resistance (such as ABCB1, BCL2, PDGFD, and SPP1), tumor–nerve interactions (including SEMA3E, RELN, and SEMA5A), and increased metastatic potential (NFIB) [55]. For a comprehensive overview of PDAC molecular subtypes, see Table 1.

5. Therapeutic implications of genetic alteration in PDAC

5.1. Targeted Therapies

5.1.1. Targeting KRAS Mutations in PDAC

Gemcitabine/nab-paclitaxel and modified FOLFIRINOX are the National Comprehensive Cancer Network (NCCN)–recommended treatments for patients with locally advanced or metastatic PDAC, but their effectiveness remains limited. Despite numerous efforts, targeting the most common molecular alterations in PDAC has proven challenging [56].
KRAS mutations occur in over 90% of PDAC cases, yet the KRASG12C mutation—which accounts for only about 1% of PDAC patients—has been the first KRAS mutant to be effectively targeted by a specific small-molecule inhibitor. Sotorasib irreversibly binds to a cryptic pocket in KRASG12C, locking the protein in an inactive state and thereby inhibiting tumor growth [57]. Clinical trials have shown significant radiographic responses to sotorasib in KRASG12C -mutated lung adenocarcinoma [58], leading to its approval by the U.S. Food and Drug Administration (FDA) as the first RAS inhibitor [59]. In a phase 1–2 clinical trial involving 38 patients with previously treated KRASG12C -mutated advanced pancreatic cancer, sotorasib demonstrated promising results: eight patients achieved an objective response, with a median progression-free survival of 4.0 months and a median overall survival of 6.9 months [60]. Another KRASG12C inhibitor, adagrasib, has also shown efficacy in preclinical models and produced radiographic responses in KRASG12C -mutated lung and colon adenocarcinomas. Preliminary data from the KRYSTAL-1 trial reported clinical benefits in KRASG12C -mutated metastatic PDAC, with partial responses observed in 5 out of 10 patients [61]. Meanwhile, a third small-molecule inhibitor, MRTX1133, which specifically targets KRASG12D —the most common KRAS mutation in PDAC—is currently in preclinical development. This compound has demonstrated potent inhibition of activated KRASG12D in vitro [62]. Given the high prevalence of KRASG12D mutations in PDAC, these emerging inhibitors hold substantial promise for improving treatment outcomes.

5.1.2. Targeting Non-KRAS Drivier Mutations in PDAC

Approximately 10% of PDACs do not harbor KRAS mutations [63], but instead feature a range of potentially druggable non-KRAS driver alterations. Among these, activating BRAF mutations occur in about 2% of PDACs, most commonly due to in-frame deletions or the V600E point mutation (BRAFV600E), which result in constitutive activation of the MAPK pathway [63]. A recent study demonstrated that MEK inhibitors such as cobimetinib and trametinib were effective in regressing gemcitabine-resistant PDAC in patient-derived orthotopic xenograft (PDOX) models [64]. Based on these findings, a phase 2 multicenter clinical trial is underway to evaluate the combination of BRAF and MEK inhibitors (encorafenib and binimetinib, respectively) in patients with advanced BRAFV600E-mutated PDAC [64].
Neurotrophic receptor tyrosine kinase (NTRK) gene fusions, which drive mitogenic signaling in the central nervous system [65,66], are present in approximately 0.3% of PDACs [67]. These fusions arise from chromosomal rearrangements that result in the expression of chimeric tropomyosin receptor kinases. In a multicenter study of advanced cancers with NTRK fusions, including PDAC, 75% of patients responded to treatment with the TRK inhibitor larotrectinib, and 71% of those responses were sustained after one year [68]. These results led to the regulatory approval of larotrectinib for the treatment of advanced solid tumors with NTRK fusions.
Activating neuregulin 1 (NRG1) fusions, identified in approximately 0.13–0.5% of pancreatic ductal adenocarcinomas (PDACs) [67,69], represent a promising therapeutic target. NRG1 acts as a ligand for ERBB3 and ERBB4 receptors, and its fusion-driven overexpression promotes tumorigenesis through hyperactivation of ERBB signaling. This pathway is particularly relevant in NRG1 fusion–positive, KRAS wild-type PDAC [70]. Reflecting this, the FDA recently approved zenocutuzumab-zbco for patients with advanced or metastatic PDAC harboring an NRG1 gene fusion [71].
In a similar context, RET (rearranged during transfection) gene fusions, though rare, are clinically significant, occurring in approximately 0.6% of PDAC cases [72,73]. These fusions generate constitutively active RET receptor tyrosine kinases that drive cell proliferation and survival. Tumors with RET fusions often lack other canonical driver mutations, rendering them particularly sensitive to targeted therapy. Accordingly, the FDA has approved selpercatinib, a selective RET inhibitor, for adults with advanced or metastatic solid tumors harboring RET fusions, including PDAC [74].

5.2. DNA Repair Pathway in PDAC

Microsatellite instability (MSI) or deficient mismatch repair (dMMR) is observed in a small subset of PDACs (approximately 1–2%) [75] and is associated with a high tumor mutational burden. Tumors with MSI/dMMR are more likely to respond to immune checkpoint blockade. Reflecting this, the FDA has approved pembrolizumab, a PD-1 inhibitor, for the treatment of patients with advanced or metastatic MSI-high or dMMR solid tumors, including pancreatic cancer, providing a targeted immunotherapy option for this molecularly defined subgroup [76].
Similarly, defects in DNA repair due to germline BRCA1 or BRCA2 mutations, which are present in a subset of PDACs [77], create another actionable vulnerability. These tumors may be particularly sensitive to therapies that exploit deficiencies in DNA repair, such as platinum-based chemotherapies, which induce double-strand DNA breaks, or poly (ADP-ribose) polymerase (PARP) inhibitors, which block the repair of these lesions [1,78]. The POLO trial demonstrated that the PARP inhibitor olaparib provides meaningful clinical benefit for patients with metastatic PDAC harboring germline BRCA1 or BRCA2 mutations who had previously responded to platinum-based chemotherapy [79]. In this randomized, placebo-controlled study, patients treated with olaparib experienced a median progression-free survival of 7.4 months, compared to 3.8 months in the placebo group. These findings led to FDA approval of olaparib as maintenance therapy for platinum-sensitive metastatic PDAC in patients with germline BRCA mutations [79].
In addition to BRCA1/2, other key regulators of DNA damage response, such as ATM and ATR, may serve as therapeutic targets. These genes encode members of the phosphatidylinositol 3-kinase-like kinase (PIKK) family, which play essential roles in DNA repair signaling. Tumors with ATM mutations have shown sensitivity to combined ATR and PARP inhibition, whereas PARP inhibitor monotherapy alone has demonstrated limited and short-lived activity in this context [80]. As a result, several early-phase clinical trials are currently underway to evaluate ATR inhibitors, both alone and in combination with cytotoxic chemotherapy or PARP inhibitors. For a summary of common genetic alterations in PDAC and corresponding targeted therapies, see Table 2.

5.3. Tumor Suppressor Pathways

TP53 is the most commonly inactivated tumor suppressor gene in PDAC. Emerging therapeutic strategies are exploring ways to restore or modulate the TP53 pathway. One such approach involves zinc chelators like COTI-2, which may stabilize mutant p53, promote proper protein folding, and prevent the aggregation of dysfunctional p53 [81]. Another strategy targets Mouse double minute 2 homolog (MDM2), a negative regulator of p53 that promotes its degradation through direct binding and ubiquitin-mediated mechanisms [82]. Inhibiting MDM2 may help restore p53 activity in tumors with wild-type TP53. Another pathway of interest in PDAC is the transforming growth factor-beta (TGF-β)/SMAD4 signaling cascade. This pathway plays a dual role in tumorigenesis, but in advanced stages, it promotes tumor invasion and metastasis. Inhibitors of TGF-β, such as trabedersen and galunisertib, have been shown to reduce tumor progression and metastatic potential in preclinical animal models [83]. CDKN2A, another frequently altered tumor suppressor gene in PDAC, encodes the proteins p16 and p19, which regulate cell cycle progression at the G1/S checkpoint through CDK4/6 inhibition [84]. Loss of CDKN2A function leads to unchecked cell proliferation. CDK4/6 inhibitors not only restore cell cycle control but have also been shown to impair DNA repair pathways and enhance the sensitivity of PDAC cells to PARP inhibitors [77]. These agents have demonstrated efficacy in preclinical PDAC models, and several related clinical trials are currently ongoing [84].

5.4. Other Oncogenic Pathways

The Wnt signaling pathway plays a key role in regulating cell differentiation, proliferation, and epithelial-to-mesenchymal transition [84]. Zhang et al. demonstrated that ligand-mediated activation of the Wnt/β-catenin pathway is essential for both the initiation and progression of PDAC [85]. In preclinical models, the monoclonal antibody vantictumab, which blocks Wnt signaling, was shown to reduce cancer stem cell frequency and enhance the efficacy of chemotherapy [86]. The JAK/STAT signaling pathway is also critically involved in tumor progression across several cancer types. In PDAC, elevated expression of JAK and activation of the IL-6R/JAK/STAT axis have been associated with poorer prognosis in patients with resectable disease [87]. Inhibition of STAT3 has been found to enhance chemosensitivity and delay tumor progression [88]. Although JAK inhibitors such as itacitinib and momelotinib demonstrated acceptable safety profiles and some clinical activity in trials, they failed to produce a significant survival benefit when compared to chemotherapy alone [84]. Another pathway implicated in PDAC progression is Notch signaling, which is frequently upregulated and contributes to tumorigenesis. Through crosstalk with the Hedgehog, KRAS, and NF-κB pathways, Notch signaling promotes tumor cell proliferation and differentiation by modulating gene transcription [84]. As the pathway is activated by γ-secretase, inhibitors targeting this enzyme were developed as potential therapies. However, clinical trials were discontinued due to intolerable toxicity. Additional agents targeting Notch2 and Notch3 receptors have also been explored, but results to date have been suboptimal [89,90].

5.5. Stromal Targets and Tumor Microenvironment

PDAC is characterized by a dense fibrous stroma, resulting from the proliferation of fibrous tissue and alterations in the extracellular matrix (ECM). The stromal elements in PDAC play a critical role in regulating tumor growth, vascularization, immune response, drug delivery, and metastasis [91]. The ECM consists of a complex network of structural proteins, adaptor molecules, proteoglycans, and enzymes that contribute to the rigidity and resistance of the tumor microenvironment. One key ECM component is hyaluronic acid (HA), which increases interstitial pressure within the tumor, leading to vascular collapse and reduced perfusion [92]. This limits the accessibility of chemotherapeutic agents to cancer cells. Therapeutic approaches targeting this barrier, such as recombinant human hyaluronidase (PEGPH20), have been explored, but clinical trials have yielded disappointing results [93]. Matrix metalloproteinases (MMPs) are another group of stromal modifiers that degrade ECM components and the basement membrane, thereby facilitating tumor invasion, angiogenesis, and metastasis. Marimastat, an MMP inhibitor, demonstrated safety and biological activity in early-phase studies but failed to provide a survival benefit when combined with gemcitabine in clinical trials [84].

5.6. Therapeutic Implication of PDAC Molecular Subtype

Molecular subtyping of advanced PDAC provides valuable insights into the determinants of chemotherapy response. The recent COMPASS trial evaluated this by performing tumor biopsies followed by RNA sequencing in 195 patients. Tumors were classified into two major subtypes: classical (80%) and basal-like (20%) [94]. Patients with the classical subtype showed significantly better responses to first-line 5-fluorouracil–based chemotherapy regimens, while those with the basal-like subtype exhibited reduced chemo-responsiveness [94]. Ongoing validation studies aim to further refine the clinical utility of PDAC subtyping in guiding personalized treatment strategies.

6. Discussion

Over the past several decades, genomic research has significantly advanced our understanding of the complex molecular landscape of PDAC. While many of these insights have yet to be fully translated into clinical practice, the identification of key genetic alterations and molecular subtypes holds substantial promise. These discoveries not only offer opportunities to optimize existing therapies but also pave the way for the development of novel, targeted treatment strategies. With continued progress, there is hope that PDAC—historically one of the most lethal malignancies—can become a more manageable and treatable disease.

7. Conclusions

PDAC remains a highly lethal cancer with a complex and heterogeneous molecular landscape. Advances in next-generation sequencing have identified frequent mutations in critical genes such as KRAS, TP53, SMAD4/DPC4, and CDKN2A, along with numerous less common genetic alterations. Clear genotype-phenotype correlations have been established across various PDAC histologic subtypes and precursor lesions. When integrated with transcriptomic analyses, these molecular insights have paved the way for the development of targeted therapies. Particularly encouraging are recent clinical trials of KRAS-specific inhibitors and the FDA’s approval of PARP inhibitors, both representing significant strides forward in PDAC treatment.

Author Contributions

Both authors have read and agreed to the published version of the manuscript. The authors thank Dr. Mark L. Cohen for his critical review editing.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

As this is a review article discussing the available literature, no new data were created.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 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]
  2. 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]
  3. Hosoda, W.; Wood, L.D. Molecular Genetics of Pancreatic Neoplasms. Surg. Pathol. Clin. 2016, 9, 685–703. [Google Scholar] [CrossRef]
  4. Luo, J. KRAS mutation in pancreatic cancer. Semin Oncol. 2021, 48, 10–18. [Google Scholar] [CrossRef]
  5. Taherian M, Wang H, Wang H. Pancreatic Ductal Adenocarcinoma: Molecular Pathology and Predictive Biomarkers. Cells. 2022, 11. [Google Scholar]
  6. 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]
  7. 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]
  8. 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]
  9. 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]
  10. 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]
  11. 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]
  12. 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]
  13. Kim, W.Y.; Sharpless, N.E. The Regulation of INK4/ARF in Cancer and Aging. Cell 2006, 127, 265–275. [Google Scholar] [CrossRef]
  14. 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]
  15. 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]
  16. 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]
  17. 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]
  18. Hong SM, Park JY, Hruban RH, Goggins M. Molecular signatures of pancreatic cancer. Arch Pathol Lab Med. 2011;135(6):716-27.
  19. 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]
  20. 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]
  21. 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]
  22. Wilson, B.G.; Roberts, C.W.M. SWI/SNF nucleosome remodellers and cancer. Nat. Rev. Cancer 2011, 11, 481–492. [Google Scholar] [CrossRef]
  23. 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]
  24. 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]
  25. 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]
  26. 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]
  27. 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]
  28. 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]
  29. 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.
  30. 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]
  31. 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.
  32. 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]
  33. 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]
  34. 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]
  35. 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]
  36. 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]
  37. 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]
  38. 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]
  39. 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]
  40. 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]
  41. 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]
  42. 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]
  43. 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]
  44. 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]
  45. 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.
  46. 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]
  47. 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]
  48. 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]
  49. 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]
  50. 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]
  51. 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]
  52. 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]
  53. 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.
  54. 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]
  55. 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]
  56. 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]
  57. 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]
  58. 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.
  59. 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]
  60. 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.
  61. 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]
  62. 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]
  63. 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]
  64. 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]
  65. 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]
  66. 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]
  67. 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]
  68. 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]
  69. 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]
  70. 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.
  71. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-zenocutuzumab-zbco-non-small-cell-lung-cancer-and-pancreatic.
  72. 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]
  73. 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]
  74. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-selpercatinib-locally-advanced-or-metastatic-ret-fusion-positive-solid-tumors.
  75. 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]
  76. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-chemotherapy-biliary-tract-cancer.
  77. 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]
  78. 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]
  79. 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]
  80. 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]
  81. 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]
  82. 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]
  83. 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]
  84. 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]
  85. 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.
  86. 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]
  87. Kung, H.; Yu, J. Targeted therapy for pancreatic ductal adenocarcinoma: Mechanisms and clinical study. Medcomm 2023, 4, e216. [Google Scholar] [CrossRef]
  88. 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]
  89. 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]
  90. 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]
  91. 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]
  92. 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]
  93. 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]
  94. 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]
Table 1. Molecular subtypes of PDAC.
Table 1. Molecular subtypes of PDAC.
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
Table 2. Common genetic alterations in PDAC.
Table 2. Common genetic alterations in PDAC.
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated