A comprehensive understanding of molecular targets involved in Colon Cancer pathogenesis is fundamental for elucidating disease mechanisms, identifying actionable vulnerabilities, and supporting the development of targeted therapies. The selected targets—filtered to include only those with direct, evidence-based roles in Colon Cancer—span key oncogenic processes such as aberrant cell proliferation, resistance to apoptosis, metabolic reprogramming, DNA replication and repair, drug resistance, and immune evasion. Collectively, these targets provide mechanistic insight into how Colon Cancer develops, progresses, and evades therapy. For example, receptor tyrosine kinases like EGFR drive proliferative signaling, while enzymes such as FASN and HMGCR support metabolic adaptations. Drug resistance is mediated by ABCB1 and DPYD, influencing chemotherapeutic efficacy. Immune checkpoint proteins like PDCD1 and LAG3 facilitate immune escape, underpinning the rationale for immunotherapies. Understanding these targets enables rational drug design, biomarker development, and patient stratification, thereby advancing precision medicine in Colon Cancer.
This category encompasses targets that drive aberrant cell proliferation, survival, and tumor progression in Colon Cancer through dysregulated growth factor signaling. It includes receptor tyrosine kinases and their downstream effectors, which are frequently mutated or overexpressed in colon tumors, thereby activating pathways such as MAPK, PI3K-AKT, and STAT. These targets are central to disease onset and progression, making them key nodes for therapeutic intervention.
EGFR (epidermal growth factor receptor, gene symbol EGFR) is a transmembrane receptor tyrosine kinase characterized by an extracellular ligand-binding domain, a single transmembrane helix, and an intracellular tyrosine kinase domain. EGFR is regulated by ligand binding (e.g., EGF, TGF-α), which induces dimerization and autophosphorylation. In Colon Cancer, EGFR is frequently overexpressed or dysregulated, leading to constitutive activation of downstream signaling cascades including RAS-RAF-MEK-ERK, PI3K-AKT, and JAK-STAT pathways. This promotes uncontrolled proliferation, survival, angiogenesis, and metastasis. EGFR interacts with other growth factor receptors and is implicated in resistance to anti-EGFR therapies through mutations in downstream effectors (e.g., KRAS, BRAF). EGFR is a validated therapeutic target; monoclonal antibodies (cetuximab, panitumumab) and tyrosine kinase inhibitors are approved for metastatic colorectal cancer, with efficacy dependent on wild-type RAS/BRAF status. EGFR expression and mutational status serve as predictive biomarkers. [Entrez: 1956, KEGG: 1956, UniProt: P00533]
FGFR3 (fibroblast growth factor receptor 3, gene symbol FGFR3) is a receptor tyrosine kinase with three immunoglobulin-like extracellular domains, a single transmembrane segment, and a cytoplasmic tyrosine kinase domain. Ligand-induced dimerization triggers autophosphorylation and activation of downstream signaling pathways such as MAPK, PI3K-AKT, and PLCγ. FGFR3 mutations and overexpression have been documented in a subset of colorectal cancers, contributing to enhanced proliferation, survival, and chemoresistance. FGFR3 may interact with other oncogenic pathways, amplifying malignant phenotypes. FGFR inhibitors are under clinical investigation for tumors harboring FGFR alterations. [Entrez: 2261, KEGG: 2261, UniProt: P22607]
RET (ret proto-oncogene, gene symbol RET) encodes a receptor tyrosine kinase with a cadherin-like extracellular domain, a single transmembrane segment, and an intracellular kinase domain. RET activation occurs via ligand binding (GDNF family ligands) and co-receptors (GFRα), leading to downstream activation of RAS/MAPK and PI3K/AKT pathways. While RET is less commonly altered in Colon Cancer compared to other cancers, activating fusions and mutations have been detected in a subset of colorectal tumors and are associated with aggressive disease and resistance to standard therapies. RET inhibitors (e.g., selpercatinib, pralsetinib) are being evaluated in clinical trials. [Entrez: 5979, KEGG: 5979, UniProt: P07949]
FLT3 (fms related receptor tyrosine kinase 3, gene symbol FLT3) is a class III receptor tyrosine kinase with five immunoglobulin-like extracellular domains, a transmembrane region, and a split intracellular kinase domain. Ligand binding induces dimerization and autophosphorylation, activating downstream PI3K-AKT, RAS-MAPK, and STAT5 pathways. FLT3 is infrequently mutated in Colon Cancer but is expressed in some tumors, where it may contribute to cell survival and proliferation. Its pathogenic role is less prominent than in hematologic malignancies but remains a potential target in select colorectal cancer subsets. [Entrez: 2322, KEGG: 2322, UniProt: P36888]
This category includes targets that mediate metabolic adaptations in Colon Cancer cells, supporting rapid growth, survival under hypoxic conditions, and resistance to metabolic stress. These enzymes are upregulated or hyperactive in tumors, facilitating increased lipid and cholesterol biosynthesis, which are essential for membrane biogenesis and signaling.
FASN (fatty acid synthase, gene symbol FASN) is a large, multifunctional enzyme complex with domains for acyl carrier protein, ketoacyl synthase, and thioesterase activities, catalyzing the de novo synthesis of long-chain fatty acids from acetyl-CoA and malonyl-CoA. FASN is transcriptionally upregulated in Colon Cancer via oncogenic signaling (e.g., PI3K-AKT, SREBP1c) and is regulated by nutrient and hormonal cues. Overexpression of FASN promotes tumor growth, survival, and metastasis by supplying lipids for membrane synthesis and signaling molecules. FASN activity is associated with chemoresistance and poor prognosis. FASN inhibitors (e.g., TVB-2640) are in clinical trials for solid tumors, including colorectal cancer. [Entrez: 2194, KEGG: 2194, UniProt: P49327]
HMGCR (3-hydroxy-3-methylglutaryl-CoA reductase, gene symbol HMGCR) is the rate-limiting enzyme in the mevalonate pathway, catalyzing the conversion of HMG-CoA to mevalonate, a precursor for cholesterol and isoprenoids. HMGCR contains a membrane-anchored N-terminal domain and a cytosolic catalytic domain. It is regulated by sterol levels, phosphorylation, and degradation. In Colon Cancer, increased HMGCR expression supports membrane synthesis, cell proliferation, and oncogenic signaling (e.g., via prenylation of RAS). Statins, which inhibit HMGCR, demonstrate anti-proliferative effects in preclinical models and have been associated with reduced colorectal cancer risk and improved outcomes in some epidemiological studies. [Entrez: 3156, KEGG: 3156, UniProt: P04035]
This category covers targets involved in DNA topology, repair, and drug metabolism, which are central to maintaining genomic integrity and mediating resistance to chemotherapy in Colon Cancer. These proteins impact disease progression by enabling tumor cell survival under genotoxic stress and by modulating drug efficacy.
TOP1 (DNA topoisomerase I, gene symbol TOP1) is a nuclear enzyme with a core domain that relaxes supercoiled DNA by introducing transient single-strand breaks, facilitating DNA replication and transcription. TOP1 is regulated via post-translational modifications and interaction with other DNA repair proteins. In Colon Cancer, TOP1 is frequently overexpressed, contributing to increased proliferation and survival. It is the molecular target of irinotecan, a topoisomerase inhibitor used in colorectal cancer treatment. Altered TOP1 expression or mutations can influence drug sensitivity and resistance. [Entrez: 7150, KEGG: 7150, UniProt: P11387]
DPYD (dihydropyrimidine dehydrogenase, gene symbol DPYD) is a cytosolic flavoprotein with multiple domains for electron transfer, catalyzing the reduction of uracil and thymine. DPYD is the rate-limiting enzyme in 5-fluorouracil (5-FU) catabolism. Genetic polymorphisms or reduced expression of DPYD result in impaired 5-FU metabolism, leading to increased toxicity or, conversely, resistance. DPYD activity is a critical determinant of 5-FU efficacy and safety in colorectal cancer chemotherapy. DPYD genotyping and phenotyping are recommended for patient stratification. [Entrez: 1806, KEGG: 1806, UniProt: Q12882]
This category includes targets that mediate multidrug resistance in Colon Cancer by actively exporting chemotherapeutic agents out of tumor cells, thereby reducing drug efficacy and contributing to treatment failure.
ABCB1 (ATP binding cassette subfamily B member 1, gene symbol ABCB1, also known as MDR1 or P-glycoprotein) is a transmembrane transporter with two nucleotide-binding domains and two transmembrane domains, forming a channel for substrate efflux. ABCB1 is regulated at the transcriptional and post-translational level by oncogenic signaling and drug exposure. Overexpression of ABCB1 in Colon Cancer leads to active efflux of chemotherapeutic agents (e.g., irinotecan, doxorubicin), resulting in multidrug resistance. Inhibitors of ABCB1 have been explored to overcome resistance, but clinical success is limited due to toxicity and compensatory mechanisms. ABCB1 expression is a negative prognostic biomarker. [Entrez: 5243, KEGG: 5243, UniProt: P08183]
This category encompasses targets that modulate the tumor microenvironment, including stromal remodeling and immune cell interactions, thereby facilitating tumor invasion, metastasis, and immune evasion in Colon Cancer.
FAP (fibroblast activation protein alpha, gene symbol FAP) is a type II transmembrane serine protease with a short cytoplasmic tail, a transmembrane domain, and a large extracellular catalytic domain. FAP is upregulated in cancer-associated fibroblasts within the colon tumor stroma, where it remodels the extracellular matrix and promotes tumor cell invasion, angiogenesis, and immune suppression. FAP also interacts with growth factor signaling pathways. FAP inhibitors and FAP-targeted therapies (e.g., antibody-drug conjugates, CAR-T cells) are under investigation for their potential to disrupt tumor-stroma interactions and enhance anti-tumor immunity in colorectal cancer. [Entrez: 2191, KEGG: 2191, UniProt: Q12884]
This category includes immune checkpoint molecules that are upregulated in Colon Cancer to suppress anti-tumor immune responses, enabling tumor cells to evade immune surveillance. These targets are central to the development and success of immunotherapies.
PDCD1 (programmed cell death 1, gene symbol PDCD1, also known as PD-1) is an immunoinhibitory receptor with an extracellular IgV domain, a transmembrane region, and an intracellular tail containing ITIM and ITSM motifs. PDCD1 is expressed on activated T cells and, upon binding to its ligands (PD-L1, PD-L2), transduces inhibitory signals that dampen T cell activation and proliferation. In colorectal cancer, particularly in microsatellite instability-high (MSI-H) tumors, PD-1/PD-L1 expression is upregulated, leading to immune escape. PD-1 blockade (e.g., pembrolizumab, nivolumab) has demonstrated efficacy in MSI-H/dMMR colorectal cancer. PDCD1 is a predictive biomarker for immunotherapy response. [Entrez: 5133, KEGG: 5133, UniProt: Q15116]
LAG3 (lymphocyte activating 3, gene symbol LAG3) is a type I transmembrane protein with four extracellular Ig-like domains, a transmembrane region, and a cytoplasmic tail containing a unique KIEELE motif. LAG3 is upregulated on exhausted T cells in the tumor microenvironment, where it binds MHC class II and transduces inhibitory signals, further suppressing anti-tumor immunity. LAG3 cooperates with PD-1 to mediate T cell dysfunction in colorectal tumors. LAG3 inhibitors are in early-phase clinical trials for solid tumors, including colorectal cancer, often in combination with PD-1 blockade. [Entrez: 3902, KEGG: 3902, UniProt: P18627]
| Name | Short Name | Entrez Gene | KEGG | UniProtKB |
|---|---|---|---|---|
| 3-hydroxy-3-methylglutaryl-CoA reductase | HMGCR | 3156 | 3156 | P04035 |
| ATP binding cassette subfamily B member 1 | ABCB1 | 5243 | 5243 | P08183 |
| dihydropyrimidine dehydrogenase | DPYD | 1806 | 1806 | Q12882 |
| DNA topoisomerase I | TOP1 | 7150 | 7150 | P11387 |
| epidermal growth factor receptor | EGFR | 1956 | 1956 | P00533 |
| fatty acid synthase | FASN | 2194 | 2194 | P49327 |
| fibroblast activation protein alpha | FAP | 2191 | 2191 | Q12884 |
| fibroblast growth factor receptor 3 | FGFR3 | 2261 | 2261 | P22607; F8W9L4 |
| fms related receptor tyrosine kinase 3 | FLT3 | 2322 | 2322 | P36888 |
| lymphocyte activating 3 | LAG3 | 3902 | 3902 | P18627 |
| programmed cell death 1 | PDCD1 | 5133 | 5133 | Q15116 |
| programmed cell death 1 | Pdcd1 | 18566 | 18566 | Q02242 |
| ret proto-oncogene | RET | 5979 | 5979 | P07949 |
| sirtuin 1 | SIRT1 | 23411 | 23411 | Q96EB6 |
Make Order
Experimental Scheme
Implementation
Conclusion