Targets for Pancreatic Cancer
Drug R&D Solutions

Targets for Pancreatic Cancer

Inquiry

A comprehensive understanding of molecular targets in pancreatic cancer is pivotal for elucidating disease pathogenesis, identifying actionable therapeutic strategies, and advancing drug development. The targets listed here represent key molecular nodes implicated in the progression and maintenance of pancreatic cancer, including enzymes involved in nucleotide metabolism and DNA repair, growth factor receptors mediating oncogenic signaling, and immune checkpoints that facilitate tumor immune evasion. Collectively, these targets highlight the multifaceted biology of pancreatic cancer, encompassing dysregulated cell proliferation, resistance to apoptosis, genomic instability, and immune escape. By dissecting the functional roles and mechanistic pathways of these proteins, researchers can pinpoint vulnerabilities in the tumor, inform rational drug design, and guide the development of targeted therapies and biomarkers. This strategic mapping of disease-relevant targets not only enhances our understanding of pancreatic cancer biology but also supports the translation of molecular insights into clinical interventions.

Oncogenic Signaling And Receptor Tyrosine Kinases

This category includes targets that drive pancreatic cancer progression through aberrant activation of receptor tyrosine kinases and downstream oncogenic signaling pathways. The primary target in this category is Epidermal Growth Factor Receptor (EGFR), which is frequently overexpressed or hyperactivated in pancreatic ductal adenocarcinoma (PDAC). EGFR signaling promotes uncontrolled cell proliferation, survival, angiogenesis, and metastasis. Therapeutic targeting of EGFR has shown clinical benefit in combination with chemotherapy. Only EGFR is included here, as other listed receptor tyrosine kinases (e.g., KIT) lack strong evidence for direct involvement in pancreatic cancer pathogenesis.

Epidermal Growth Factor Receptor (EGFR)

Epidermal Growth Factor Receptor (EGFR) is a transmembrane receptor tyrosine kinase encoded by the EGFR gene (Entrez: 1956, KEGG: 1956, UniProt: P00533). Structurally, EGFR contains an extracellular ligand-binding domain, a single transmembrane helix, and an intracellular tyrosine kinase domain with multiple autophosphorylation sites. EGFR is regulated by ligand binding (e.g., EGF, TGF-α), receptor dimerization, and downstream adaptor proteins. In pancreatic cancer, EGFR is frequently overexpressed or activated via ligand-dependent or -independent mechanisms, leading to constitutive activation of the RAS-RAF-MEK-ERK and PI3K-AKT pathways. This drives tumor cell proliferation, survival, and resistance to apoptosis. EGFR cross-talks with other pathways, including KRAS (mutated in >90% of PDAC), amplifying oncogenic signals. Clinical evidence supports the pathogenic role of EGFR: high EGFR expression correlates with poor prognosis and therapeutic resistance. The EGFR inhibitor erlotinib, in combination with gemcitabine, has shown modest survival benefit in advanced PDAC (M. Moore et al., NEJM 2007). EGFR is also being explored as a biomarker for patient stratification and as a target for novel antibody-drug conjugates.

Nucleotide Metabolism And Dna Synthesis

This category encompasses enzymes involved in nucleotide biosynthesis, salvage, and metabolism, which are essential for the rapid proliferation of pancreatic cancer cells. Key targets include Dihydrofolate Reductase (DHFR), Thymidylate Synthetase (TYMS), DNA Topoisomerase I (TOP1), and Uridine Monophosphate Synthetase (UMPS). These enzymes are directly targeted by cytotoxic chemotherapies, and their expression and activity contribute to drug sensitivity and resistance. Their dysregulation supports enhanced DNA synthesis, repair, and tumor growth.

Dihydrofolate Reductase (DHFR)

Dihydrofolate Reductase (DHFR) is a key folate cycle enzyme (Entrez: 1719, KEGG: 1719, UniProt: P00374) that catalyzes the reduction of dihydrofolate to tetrahydrofolate, supporting thymidylate and purine synthesis. Structurally, DHFR is a monomeric enzyme with a central NADPH-binding domain and a dihydrofolate-binding site. DHFR is regulated at the transcriptional and translational levels, with upregulation observed in rapidly proliferating cancer cells. In pancreatic cancer, increased DHFR activity supports nucleotide biosynthesis required for DNA replication and repair, contributing to tumor growth and chemoresistance. DHFR is the target of antifolate drugs (e.g., methotrexate), though these are not standard in PDAC due to limited efficacy. DHFR expression is a potential biomarker for antifolate sensitivity.

Thymidylate Synthetase (TYMS)

Thymidylate Synthetase (TYMS) (Entrez: 7298, KEGG: 7298, UniProt: P04818) catalyzes the methylation of deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP), a critical step in DNA synthesis. TYMS is a homodimeric enzyme with a central folate-binding domain. Its expression is cell cycle-regulated and elevated in proliferating tumor cells. In pancreatic cancer, high TYMS levels are associated with increased DNA synthesis, tumor aggressiveness, and resistance to fluoropyrimidine-based chemotherapies (e.g., 5-FU, capecitabine). TYMS inhibition leads to DNA damage and apoptosis. TYMS expression serves as a predictive biomarker for 5-FU response (N. Nakao et al., Cancer Chemother Pharmacol 2007).

DNA Topoisomerase I (TOP1)

DNA Topoisomerase I (TOP1) (Entrez: 7150, KEGG: 7150, UniProt: P11387) is a nuclear enzyme that relieves torsional stress during DNA replication and transcription by inducing transient single-strand breaks. The protein consists of an N-terminal domain, a core DNA-binding and cleavage domain, and a C-terminal catalytic domain. TOP1 is regulated via post-translational modifications and protein-protein interactions. In pancreatic cancer, increased TOP1 activity supports rapid DNA replication and cell division. TOP1 is the target of camptothecin derivatives (e.g., irinotecan), which trap the TOP1-DNA cleavage complex, causing lethal DNA breaks. Clinical trials have evaluated irinotecan-based regimens (e.g., FOLFIRINOX) in advanced PDAC, demonstrating improved survival (Conroy et al., NEJM 2011). TOP1 expression may predict sensitivity to these agents.

Uridine Monophosphate Synthetase (UMPS)

Uridine Monophosphate Synthetase (UMPS) (Entrez: 7372, KEGG: 7372, UniProt: P11172) is a bifunctional enzyme catalyzing the final steps of de novo pyrimidine biosynthesis (orotate to UMP). UMPS has two domains: orotate phosphoribosyltransferase and orotidine-5'-phosphate decarboxylase. Its expression is upregulated in proliferating tumor cells to meet increased nucleotide demand. In pancreatic cancer, enhanced UMPS activity supports DNA/RNA synthesis and cell proliferation. UMPS is also required for the activation of 5-fluorouracil (5-FU) to its cytotoxic metabolites, making it a determinant of 5-FU efficacy (M. Peters et al., Cancer Res 2002).

Dna Damage Response And Repair

This category includes enzymes central to DNA repair and the cellular response to genotoxic stress, which are crucial for pancreatic cancer cell survival in the face of DNA-damaging therapies. Poly(ADP-ribose) Polymerase 1 (PARP1) is the principal target in this group, playing a pivotal role in base excision repair and facilitating the repair of single-strand breaks. Inhibition of PARP1 induces synthetic lethality in tumors with homologous recombination deficiency (e.g., BRCA1/2-mutant PDAC).

Poly(ADP-ribose) Polymerase 1 (PARP1)

Poly(ADP-ribose) Polymerase 1 (PARP1) (Entrez: 142, KEGG: 142, UniProt: P09874) is a nuclear enzyme with multiple functional domains: DNA-binding zinc fingers, an automodification domain, and a catalytic PARP domain. PARP1 detects DNA single-strand breaks and catalyzes poly(ADP-ribosyl)ation of target proteins, recruiting DNA repair machinery. PARP1 activity is regulated by DNA damage, post-translational modifications, and protein-protein interactions. In pancreatic cancer, especially in tumors with BRCA1/2 or PALB2 mutations, PARP1-mediated repair is critical for cell survival. PARP inhibitors (e.g., olaparib) exploit synthetic lethality in homologous recombination-deficient tumors, causing accumulation of DNA damage and cell death. The POLO trial (Golan et al., NEJM 2019) demonstrated significant benefit of olaparib as maintenance therapy in germline BRCA-mutated metastatic PDAC, establishing PARP1 as a validated therapeutic target.

Immune Evasion And Immune Checkpoints

This category comprises immune checkpoint molecules that regulate T cell activation and contribute to immune evasion in pancreatic cancer. Programmed Cell Death 1 (PDCD1, PD-1) is the main target included here. PD-1 is expressed on T cells and, upon engagement by its ligands (PD-L1/PD-L2), suppresses anti-tumor immune responses, facilitating tumor progression. Although immune checkpoint inhibitors have limited efficacy in unselected PDAC, they are effective in rare cases with high microsatellite instability (MSI-H) or mismatch repair deficiency (dMMR).

Programmed Cell Death 1 (PDCD1)

Programmed Cell Death 1 (PDCD1, PD-1) (Entrez: 5133, KEGG: 5133, UniProt: Q15116) is a type I transmembrane protein comprising an immunoglobulin V-like extracellular domain, a transmembrane region, and an intracellular tail with ITIM and ITSM motifs. PD-1 expression is induced on activated T cells. Binding of PD-1 to its ligands PD-L1/PD-L2, which are upregulated on pancreatic tumor cells and stroma, delivers inhibitory signals that attenuate T cell proliferation and cytokine production. This immune checkpoint pathway is a major mechanism of immune evasion in PDAC. Clinical trials of anti-PD-1 antibodies (e.g., pembrolizumab) have shown efficacy in MSI-H/dMMR pancreatic tumors (Le et al., Science 2017), and PD-1 expression is being explored as a biomarker for immunotherapy response.

Name Short Name Entrez Gene KEGG UniProtKB
dihydrofolate reductase DHFR 1719 1719 P00374
dihydropyrimidine dehydrogenase DPYD 1806 1806 Q12882
DNA topoisomerase I TOP1 7150 7150 P11387
epidermal growth factor receptor EGFR 1956 1956 P00533
growth hormone 1 GH1 2688 2688 P01241
KIT proto-oncogene, receptor tyrosine kinase KIT 3815 3815 P10721; A0A8I5KS03
poly(ADP-ribose) polymerase 1 PARP1 142 142 P09874
poly(ADP-ribose) polymerase 2 PARP2 10038 10038 Q9UGN5
poly(ADP-ribose) polymerase family member 3 PARP3 10039 10039 Q9Y6F1
programmed cell death 1 PDCD1 5133 5133 Q15116
receptor interacting serine/threonine kinase 2 RIPK2 8767 8767 O43353
somatostatin receptor 2 SSTR2 6752 6752 P30874
somatostatin receptor 2 Sstr2 20606 20606 P30875
somatostatin receptor 3 SSTR3 6753 6753 P32745
thioredoxin TXN 7295 7295 P10599
thymidylate synthetase TYMS 7298 7298 P04818
uridine monophosphate synthetase UMPS 7372 7372 P11172
HOW WE WORK

Make Order

Make Order

Experimental Scheme

Experimental Scheme

Implementation

Implementation

Conclusion

Conclusion
Make an Inquiry