Understanding the molecular targets implicated in Bladder Cancer pathogenesis is fundamental to elucidating the mechanisms driving tumor initiation, progression, and therapeutic resistance. The listed targets—when critically filtered for direct relevance—collectively reveal key oncogenic signaling pathways, immune evasion mechanisms, cell proliferation regulators, and druggable vulnerabilities. For instance, receptor tyrosine kinases such as EGFR (epidermal growth factor receptor) and ERBB2 (HER2) are frequently overexpressed or mutated in urothelial carcinoma, fueling aberrant cell growth and survival. MYC proto-oncogene acts as a central transcriptional driver of proliferation and metabolic reprogramming. Immune checkpoint molecules like CD274 (PD-L1) are pivotal in facilitating tumor immune escape, forming the rationale for immune checkpoint blockade therapies. These targets not only deepen our understanding of disease etiology but also serve as biomarkers and direct drug targets, enabling precision medicine approaches. Their mechanistic roles provide a roadmap for developing targeted therapies, optimizing immunotherapeutic strategies, and identifying predictive biomarkers, thereby supporting ongoing drug research and clinical translation in Bladder Cancer.
This category includes receptor tyrosine kinases that are directly implicated in the pathogenesis of Bladder Cancer. EGFR (epidermal growth factor receptor) and ERBB2 (erb-b2 receptor tyrosine kinase 2, HER2) are frequently overexpressed and/or mutated in urothelial carcinoma, promoting oncogenic signaling through the MAPK and PI3K/AKT pathways. These receptors drive uncontrolled proliferation, survival, and invasion, and their activation correlates with aggressive disease and poor prognosis. Their clinical relevance is underscored by the efficacy of targeted therapies (e.g., anti-EGFR monoclonal antibodies, HER2 inhibitors) in select patient subsets.
Epidermal Growth Factor Receptor (EGFR) is a transmembrane glycoprotein with an extracellular ligand-binding domain, a single transmembrane helix, and an intracellular tyrosine kinase domain. Upon ligand binding, EGFR dimerizes and autophosphorylates, activating downstream MAPK, PI3K/AKT, and JAK/STAT pathways. In Bladder Cancer, EGFR overexpression and gene amplification are observed in up to 74% of high-grade tumors (Knowles MA et al., Nat Rev Cancer 2015), correlating with aggressive phenotype and resistance to chemotherapy. EGFR interacts with other RTKs (e.g., ERBB2) and facilitates epithelial-mesenchymal transition (EMT) and angiogenesis. Therapeutically, EGFR is targeted by monoclonal antibodies (e.g., cetuximab) and TKIs (e.g., erlotinib), though clinical benefit in Bladder Cancer remains modest, possibly due to molecular heterogeneity and compensatory signaling. EGFR is also under investigation as a predictive biomarker for targeted therapy selection. (Entrez: 1956, KEGG: 1956, UniProt: P00533)
Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2, also known as HER2) is a member of the EGFR family, characterized by an extracellular domain, a transmembrane region, and an intracellular tyrosine kinase domain. Unlike EGFR, ERBB2 has no known ligand but forms active heterodimers with other EGFR family members, amplifying downstream signaling. ERBB2 amplification/overexpression occurs in 9–12% of muscle-invasive Bladder Cancers (Cancer Genome Atlas Research Network, Cell 2017), driving oncogenesis via MAPK and PI3K/AKT pathways. ERBB2 is associated with poor prognosis and chemoresistance. Targeted therapies (trastuzumab, lapatinib, antibody-drug conjugates) are being evaluated in clinical trials, with early evidence of efficacy in HER2-positive urothelial carcinoma. ERBB2 status is also a potential biomarker for patient stratification. (Entrez: 2064, KEGG: 2064, UniProt: P04626)
This category encompasses targets involved in immune checkpoint signaling that enable Bladder Cancer cells to evade immune surveillance. The primary target in this category is CD274 (PD-L1), which is upregulated in a significant subset of urothelial carcinomas and serves as the molecular basis for immune checkpoint blockade therapies. CD274 expression correlates with tumor immune microenvironment modulation, T-cell exhaustion, and resistance to cytotoxic immune responses.
CD274 Molecule (CD274, PD-L1) is a type I transmembrane protein with an IgV-like extracellular domain that binds PD-1 on T cells, inhibiting T-cell activation and proliferation. In Bladder Cancer, CD274 is frequently upregulated via genomic amplification, inflammatory signaling (e.g., IFN-γ), and oncogenic pathways (e.g., EGFR/MYC). High PD-L1 expression is associated with poor prognosis, increased tumor-infiltrating lymphocytes, and response to checkpoint inhibitors. Mechanistically, CD274 suppresses adaptive immunity, enabling tumor immune escape. Anti-PD-L1/PD-1 therapies (e.g., atezolizumab, pembrolizumab) are FDA-approved for advanced urothelial carcinoma, with CD274 expression serving as a companion diagnostic in some settings. (Entrez: 29126, KEGG: 29126, UniProt: Q9NZQ7)
This category includes transcription factors that are directly involved in driving Bladder Cancer growth and progression. MYC proto-oncogene is a master regulator of cell proliferation, metabolism, and apoptosis, and is frequently amplified or overexpressed in urothelial carcinoma. MYC activation is associated with aggressive disease, poor differentiation, and therapeutic resistance.
MYC Proto-Oncogene, bHLH Transcription Factor (MYC) encodes a nuclear phosphoprotein with a basic helix-loop-helix leucine zipper (bHLH-LZ) domain, enabling dimerization with MAX and binding to E-box DNA sequences. In Bladder Cancer, MYC is frequently amplified (up to 15% of cases; TCGA, 2017) and overexpressed, driving transcription of genes involved in cell cycle progression, metabolism, and ribosome biogenesis. MYC also interacts with chromatin modifiers and orchestrates global transcriptional amplification. MYC activation is linked to aggressive tumor behavior, chemoresistance, and poor clinical outcome. Although direct MYC inhibition remains challenging, indirect approaches (e.g., BET inhibitors, CDK inhibitors) are under investigation. MYC overexpression is also being evaluated as a negative prognostic biomarker. (Entrez: 4609, KEGG: 4609, UniProt: P01106)
Name | Short Name | Entrez Gene | KEGG | UniProtKB |
---|---|---|---|---|
androgen receptor | AR | 367 | 367 | P10275 |
C-C motif chemokine receptor 5 | CCR5 | 1234 | 1234 | P51681 |
CD19 molecule | CD19 | 930 | 930 | P15391 |
CD274 molecule | CD274 | 29126 | 29126 | Q9NZQ7 |
dihydrofolate reductase | DHFR | 1719 | 1719 | P00374 |
DNA topoisomerase I | TOP1 | 7150 | 7150 | P11387 |
epidermal growth factor receptor | EGFR | 1956 | 1956 | P00533 |
erb-b2 receptor tyrosine kinase 2 | ERBB2 | 2064 | 2064 | P04626 |
fms related receptor tyrosine kinase 3 | FLT3 | 2322 | 2322 | P36888 |
KIT proto-oncogene, receptor tyrosine kinase | KIT | 3815 | 3815 | P10721; A0A8I5KS03 |
MYC proto-oncogene, bHLH transcription factor | MYC | 4609 | 4609 | P01106 |
receptor interacting serine/threonine kinase 2 | RIPK2 | 8767 | 8767 | O43353 |
retinoic acid receptor beta | RARB | 5915 | 5915 | P10826 |
retinoic acid receptor gamma | RARG | 5916 | 5916 | P13631 |
thioredoxin | TXN | 7295 | 7295 | P10599 |
Make Order
Experimental Scheme
Implementation
Conclusion