Copyright
©The Author(s) 2026.
World J Gastroenterol. Jan 7, 2026; 32(1): 112496
Published online Jan 7, 2026. doi: 10.3748/wjg.v32.i1.112496
Published online Jan 7, 2026. doi: 10.3748/wjg.v32.i1.112496
| Mechanism | How it works | Effect on genes with example |
| DNA methylation[6] | Addition of methyl groups | MLH1 silenced: Leads to microsatellite instability increased mutation rate |
| Histone modification[7] | Acetylation, methylation, phosphorylation of histone proteins | CDH1 (E-cadherin) silenced by H3 acetylation in promoter regions of cytokine genes (e.g., TNF-α) leads to increased transcription |
| Chromatin remodeling[8] | Changing the physical structure of chromatin | Loss of ARID1A failure of chromatin remodeling improper gene silencing or activation. Progression of HCC, CRC |
| Non-coding RNAs (e.g., miRNA, lncRNA)[9] | Bind to mRNA or DNA to regulate expression | CRC (miR-21, miR-135b, lncRNA HOTAIR); gastric cancer (miR-148a, miR-21, lncRNA MALAT1, circPVT1); inflammatory bowel disease (miR-155, miR-21, lncRNA IFNG-AS1); HCC (miR-122, miR-221/222, lncRNA HULC); celiac disease (miR-449a) |
Table 2 Different genetic pathway disorders, its mechanism of action in gastrointestinal tract
| Genetic pathway disorder | Mechanism | Examples |
| Monogenic disorders | Mutations in a single gene that often follow Mendelian inheritance patterns | Hereditary hemochromatosis (HFE gene), Wilson disease (ATP7B gene), alpha-1 antitrypsin deficiency |
| Polygenic and multifactorial disorders | Involve multiple genes and environmental interactions | Inflammatory bowel disease (over 200 loci have been identified), celiac disease (HLA-DQ2 and HLA-DQ8) |
| Cancer predisposition syndromes | Inherited mutations in tumor suppressor genes or DNA repair genes increase GI cancer risk | Lynch syndrome (HNPCC) (MLH1, MSH2), familial adenomatous polyposis (APC) gene |
| Mosaicism | Two or more genetically distinct cell populations within the same individual, derived from a single zygote | Mosaic APC gene mutations may cause attenuated forms of FAP. Very early changes in IBD |
| Genes | Function | Associated disorders |
| Genes involved in DNA repair and genomic stability[10] | ||
| MLH1, MSH2, MSH6, PMS2, EPCAM | Mismatch repair (MMR) system | Lynch syndrome colorectal, gastric, pancreatic cancer |
| MUTYH | Base excision repair | MUTYH-associated polyposis |
| BRCA1/BRCA2, ATM, PALB2 | Double-strand break repair | Familial pancreatic and gastric cancers |
| TP53 | Tumor suppressor, DNA damage response | CRC, gastric, pancreatic, hepatocellular carcinoma |
| Genes involved in cell adhesion and structural integrity[11] | ||
| CDH1 | E-cadherin (cell-cell adhesion) | Hereditary diffuse gastric cancer |
| CTNNA1 | Catenin alpha-1 (adherens junctions) | HDGC |
| SMAD4, BMPR1A | TGF-β pathway mediators | Juvenile polyposis syndrome, pancreatic cancer |
| Genes regulating inflammation and immune response[12] | ||
| NOD2 | Innate immunity, bacterial sensing | Crohn’s disease |
| IL23R, IL10, IL12B | Cytokine signaling | IBD susceptibility |
| IRGM, ATG16 L1 | Autophagy genes | Crohn’s disease |
| HLA-DQA1/HLA-DQB1 | Antigen presentation | Celiac disease |
| TLR4, TLR9 | Pattern recognition receptors | Functional dyspepsia, IBD |
| Genes involved in bile acid transport and cholestasis[13] | ||
| ABCB11 | Bile salt export pump | PFIC2, BRIC |
| ABCC2 (MRP2) | Bile excretion | Dubin-Johnson syndrome |
| ATP8B1 | Phospholipid transporter | PFIC1 |
| TJP2 | Tight junction protein | PFIC4 |
| Genes in neuronal/gut motility and enteric nervous system[14] | ||
| RET, EDNRB, GDNF | ENS development | Hirschsprung’s disease |
| SCN5A | Sodium channel in ICCs/ENS | IBS with constipation |
| NEUROG3 | Enteroendocrine differentiation | Congenital malabsorptive diarrhoea |
| Genes affecting nutrient absorption and metabolism[15] | ||
| LCT | Lactase enzyme | Lactose intolerance |
| SAR1B | Chylomicron transport | Chylomicron retention disease |
| SLC26A3 | Cl-/HCO3- exchange | Congenital chloride diarrhea |
| SLC5A1 (SGLT1) | Glucose transport | Glucose-galactose malabsorption |
| Genes in oncogenic signaling and growth factors[15] | ||
| KRAS, NRAS | MAPK signaling | CRC, pancreatic cancer |
| BRAF | Downstream of KRAS | CRC (BRAF V600E in MSI tumors) |
| PIK3CA | PI3K/AKT pathway | CRC, gastric cancer |
| EGFR, HER2 (ERBB2) | Receptor tyrosine kinases | Gastric, colorectal cancers |
| FGFR2, IDH1/IDH2 | Growth factor pathways | Cholangiocarcinoma |
| Genes related to epigenetic and transcriptional regulation[16] | ||
| ARID1A | Chromatin remodeling | Biliary cancer, CRC, gastric |
| MLH3, MSH3 | Mismatch repair (minor MMR genes) | Polyposis syndromes |
| TET2, DNMT3A | DNA methylation regulation | CRC and inflammatory epigenetic signatures |
| Pathway | Key genes | Associated disorders | Mechanism/role | Ref. |
| Wnt/β-catenin | APC, CTNNB1, AXIN2 | Colorectal cancer, hepatocellular carcinoma (HCC), familial adenomatous polyposis | Controls cell proliferation and differentiation; mutation leads to uncontrolled growth | Li et al[17] |
| NF-κB | NFKB1, TNFAIP3, IKK complex | IBD (Crohn’s, UC), gastric cancer, colorectal cancer | Regulates inflammation, cell survival, immunity; chronic activation promotes inflammation and tumorigenesis | Peng et al[18] |
| TGF-β/SMAD | TGFBR2, SMAD4 | Juvenile polyposis, CRC, pancreatic cancer | Controls growth inhibition and apoptosis; mutations cause evasion of tumor suppression | Hata and Chen[19] |
| JAK/STAT | JAK2, STAT3, STAT1 | IBD, colitis-associated cancer | Regulates immune cell signaling and cytokine responses | Hu et al[20] |
| MAPK/ERK | KRAS, BRAF, EGFR | CRC, pancreatic cancer, gastric cancer | Regulates cell proliferation and survival; mutations oncogenic signaling | Guo et al[21] |
| PI3K/AKT/mTOR | PIK3CA, PTEN, AKT1, MTOR | CRC, gastric cancer, IBD | Promotes cell growth, metabolism, and angiogenesis; dysregulation contributes to tumor growth and inflammation | Glaviano et al[22] |
| Mismatch repair | MLH1, MSH2, MSH6, PMS2 | Lynch syndrome, CRC, gastric cancer | Repairs DNA replication errors; loss leads to microsatellite instability (MSI) | Li[23] |
| P53 pathway | TP53 | CRC, esophageal, gastric, HCC | Controls cell cycle arrest, apoptosis, DNA repair; mutations common in late cancer stages | Harris and Levine[24] |
| Hedgehog signaling | PTCH1, GLI1 | Gastric cancer, GI developmental disorders | Controls tissue patterning and stem cell maintenance | Briscoe and Thérond[25] |
| Notch signaling | NOTCH1, DLL1, HES1 | Colitis, CRC, esophageal cancer | Regulates differentiation, especially goblet cells; dysregulation affects intestinal homeostasis | Kopan[26] |
| Autophagy pathway | ATG16 L1, IRGM | Crohn’s disease, IBD-associated cancer | Maintains intracellular bacterial clearance and mucosal homeostasis | Yu et al[27] |
| Immune checkpoint pathway | PD-L1, CTLA4 | MSI-high CRC, gastric cancer, IBD | Immune evasion in cancer; dysregulated tolerance in autoimmune GI diseases | He and Xu[28] |
| ER stress/UPR | XBP1, IRE1, PERK | IBD, Paneth cell dysfunction, CRC | Regulates response to unfolded proteins; chronic ER stress leads to inflammation and epithelial damage | Chen et al[29] |
| IL-23/Th17 pathway | IL23R, STAT3, RORC | Crohn’s disease, UC, CRC | Inflammatory cytokine signaling driving chronic inflammation | Bunte and Beikler[30] |
| Apoptosis/FAS-FASL | FAS, BAX, CASP8 | Colitis-associated cancer, gastric cancer | Regulates programmed cell death; evasion supports tumor survival | Waring and Müllbacher[31] |
| DNA repair pathways (base/nucleotide excision) | OGG1, XPA, POLB | CRC, gastric cancer | Repair oxidative and chemical DNA damage; defects genomic instability | Kumar et al[32] |
Table 5 Various types of clinical genetic testing
| Classification | Type | Purpose |
| Mutation origin | Germline testing | Detects inherited mutations; used for familial risk, carrier status, and predisposition |
| Somatic testing | Identifies acquired mutations in specific tissues (e.g., tumors); guides cancer therapy | |
| Mosaicism testing | Identify mosaicism in FAP, IBD | |
| Clinical purpose | Diagnostic testing | Confirms or rules out a specific genetic disorder in symptomatic individuals |
| Prognostic testing | Predicts disease course, severity, or likelihood of complications | |
| Predictive/screening | Identifies asymptomatic individuals at risk of developing a genetic disorder | |
| Carrier testing | Identifies individuals who carry one copy of a gene mutation (relevant for recessive conditions) | |
| Pharmacogenetic testing | Assesses genetic variants affecting drug metabolism and response | |
| Somatic/tumor profiling | Detects actionable mutations in cancer cells to guide targeted therapy and prognosis | |
| Newborn screening | Early identification of treatable genetic disorders in neonates |
Table 6 Germline testing available for screening
| No. | GI disorder/syndrome | Guideline source | Genes recommended for testing | Testing criteria |
| 1 | Lynch syndrome (hereditary nonpolyposis colorectal cancer) | ACG, NCCN, ESMO | MLH1, MSH2, MSH6, PMS2, EPCAM | Personal/family history of colorectal, endometrial, or other LS-associated cancers; tumor MSI or IHC abnormality |
| 2 | Familial adenomatous polyposis (FAP) | ACG, NCCN | APC | > 100 colorectal adenomas or family history of FAP |
| 3 | Attenuated FAP | ACG | APC | Patients with 10-99 adenomas |
| 4 | MUTYH-associated polyposis | ACG | MUTYH (biallelic) | Multiple adenomas and autosomal recessive inheritance |
| 5 | Peutz-Jeghers syndrome | NCCN, ESMO | STK11 | Mucocutaneous pigmentation and hamartomatous polyps; family history |
| 6 | Juvenile polyposis syndrome | ACG, NCCN | SMAD4, BMPR1A | ≥ 5 juvenile polyps or family history |
| 7 | Cowden syndrome/PTEN hamartoma tumor syndrome | NCCN | PTEN | GI polyps with mucocutaneous lesions or macrocephaly |
| 8 | Hereditary pancreatic cancer | NCCN | BRCA1/BRCA2, PALB2, ATM, CDKN2A, STK11 | Family history of pancreatic cancer or known mutation |
| 9 | Hereditary diffuse gastric cancer | NCCN | CDH1 | Family history of diffuse gastric cancer or lobular breast cancer |
| 10 | Serrated polyposis syndrome | WHO, ACG | No known high-penetrance genes; RNF43 under investigation | Multiple serrated polyps meeting WHO criteria |
| Test | Detects | Clinical use | Benefit | Limitation | Ref. |
| Cytogenetic testing | |||||
| Karyotyping (conventional cytogenetics) | Detects large chromosomal abnormalities: Trisomies, translocations, deletions, G-banding of metaphase chromosomes | Down syndrome, Turner syndrome | Whole-genome overview, identifies balanced/unbalanced rearrangements | Low resolution, cannot detect small deletions/duplications, requires dividing cells | Genetic Alliance[35] |
| Fluorescence in situ hybridization (FISH) | Fluorescent probes bind specific DNA sequences on chromosomes | Detects gene amplifications, deletions, rearrangements (e.g., HER2 in gastric cancer, ALK in GI stromal tumors) | Rapid, targeted, works on interphase cells | Limited to known targets, one probe/test, cannot assess whole genome | Yilmaz and Demiray[36] |
| Comparative genomic hybridization (aCGH) | DNA from patient and control hybridized to a microarray | Detects copy number variations (e.g., deletions in polyposis syndromes, microdeletion syndromes | High-resolution, genome-wide, detects sub microscopic CNV | Cannot detect balanced rearrangements (e.g., translocations), limited to CNVs only | Weiss et al[37] |
| Chromosomal microarray analysis | aCGH + SNP array | Used in syndromic GI diseases, unexplained developmental delay, congenital anomalies | Genome-wide, detects CNVs, uniparental disomy, mosaicism | Cannot detect balanced rearrangements, may report VUS | Myllykangas et al[38] |
| Spectral karyotyping | Whole chromosome painting with multicolor FISH | Identifies complex chromosomal rearrangements, often in cancers | Detects complex karyotypes, color-coded analysis | Expensive, not used for routine diagnostics, lower resolution than aCGH | Guo et al[39] |
| Molecular genetic testing | |||||
| Sanger sequencing | SNV, small insertions/deletions | Confirmatory testing (e.g., known APC, MLH1 mutations | High accuracy for point mutation or small deletion/duplication/SNV, cost effective for single genetic testing | Only identify small subset of gene or single gene, not precisely quantifiable | Herpich et al[40] |
| NGS | Panel, exome, or genome-wide variants | Multigene panels for IBD, polyposis, CRC, gastric cancer, GIST | Multiple, individually produced readings of the target area mosaism, quantitative, whole exome or genome sequencing | Limited in their ability to detect copy number variations, incidental findings need to be verified by sanger sequencing | Satam et al[41] |
| Targeted gene panels | Focused sequencing of disease-specific genes | Panel specific to GIST, IBD, hereditary colorectal cancer panel, gist panel | Accurate diagnosis focus on specific genes cost-effective and efficient: Can be customized according to disorder | Limited coverage not detect structural rearrangements or copy number variants cannot identify novel or new gene related to disease | Málaga et al[42] |
| Whole exome sequencing | All coding regions | Early-onset or monogenic IBD, congenital diarrheal disorders (e.g., DGAT1, EPCAM mutations). Hereditary pancreatitis (e.g., PRSS1, SPINK1) colorectal cancer | Cost-effective WES allows deeper sequencies WES captures approximately 85% of known disease-causing mutations | Misses non-coding variants incomplete exome coverage | Rabbani et al[43]; Uhlig et a[[44] |
| WGS | Coding and non-coding genome variant | Identification of colorectal cancer genes. Undiagnosed complex disease | Cover both coding and non-coding reason detection of structural variant both germline and somatic mutation | High cost difficult to pathogenic variant from benign variant | de Voer et al[45] |
| MLPA | Large deletions/duplications | Detects large deletions, especially EPCAM deletions causing MSH2 inactivation | Efficient CNV detection cost-effective and high throughput applicable on degraded DNA | Cannot detect point mutations or small indels limited to pre-designed probes | Kuiper et al[46]; Schouten et al[47] |
| qPCR | Copy number variations or known mutations | Rapid screening for common mutations, detects bacterial, viral, and parasitic DNA/RNA rapidly and accurately, bacterial load determination in gastro intestinal disorder | High sensitivity and specificity, rapid turnaround, quantitative | Requires prior sequence knowledge | Shah et al[48]; Bamias et al[49] |
| Array comparative genomic hybridization (aCGH) | Sub microscopic deletions/duplications, germline CNVs in genes like APC, SMAD4, and BMPR1A | Genome-wide coverage, germline CNVs in genes like APC, SMAD4 and BMPR1A | High resolution can detect CNVs as small as 50-100 kb | Inability to detect balanced chromosomal rearrangements difficulties in interpreting CNVs of uncertain significance | McKay et al[50]; Assämäki et al[51] |
| HLA typing (PCR-SSP, NGS-based) | HLA allele identification | Celiac disease, IBD pharmacogenetics IBD, primary sclerosing cholangitis drug-induced GI injury, idiosyncratic reactions to drugs causing hepatic/GI damage. Transplant compatibility | Cost-effective, simple requires minimal computational support | Limited resolution may not differentiate similar alleles. May yield ambiguous results | Megiorni and Pizzuti[52] |
| FISH | Large chromosomal rearrangements, gene fusions | In Barretts esophagus identifies chromosomal instability (e.g., 20q gain, 18q loss), and BRAF rearrangements; detection of HER2 gene amplification (ERBB2 at 17q12) predicts response to trastuzumab therapy (gastric cancer) | High specificity and sensitivity for targeted chromosomal regions | Targeted approach only. Limited genomic coverage | Brankley et al[53] |
| PCR | Specific known mutations | Quick detection (e.g., PRSS1 in hereditary pancreatitis), KRAS in CRC | High sensitivity and specificity can detect minute amounts of target DNA/RNA. Rapid turnaround time. Typically, within a few hours. Quantitative provides absolute or relative quantification | Requires prior sequence knowledge. Primers must be designed for specific known targets. Cannot differentiate live from dead organisms, detects DNA from both | Tol et al[54] |
| RNA-seq | Gene expression, fusion transcripts | Detects tumor-specific expression changes, fusion transcripts (e.g., NTRK fusions), and provides prognostic biomarkers in CRC reveals deregulated pathways (e.g., WNT, PI3K), tumor microenvironment features, and therapeutic target molecular marker of pancreatic cancer | Unbiased and comprehensive: Captures all RNA species (mRNA, lncRNA, miRNA, circular RNA), high resolution. Detects single-nucleotide changes, splicing variants, and gene fusions | Expensive and resource-intensive, requires advanced sequencing and computational infrastructure, data analysis is complex, needs bioinformatics expertise and robust pipelines | Bailey et al[55] |
| Disease | Prediction and prognostication | Genes | Ref. |
| FAP | Profuse polyposis | APC codon 1250-1464, 1250-1311, 1309-1324 | Nagase et al[56]; Enomoto et al[57]; Ficari et al[58]; Walon et al[59]; Gebert et al[60] |
| Desmoid tumors | APC codon 1924, 1962, 1444-1560, 1403-1987 | Caspari et al[61] | |
| Upper gastrointestinal polyps | 1445-1578 | Davies et al[62] | |
| Gastric adenomas | 1403-1987 | Caspari et al[61] | |
| Multiple extracolonic manifestations | 3’14451995, 3’1403 | Caspari et al[61] | |
| CHRPE | 311-1444, 413-1387, 542-1309 | Caspari et al[61] | |
| Crohn’s disease | Stenotic/structuring behavior | NOD2, TLR4, IL-12B, CX3CR1, IL-10, IL-6 | Tsianos et al[63] |
| Penetrating/fistulizing behavior | NOD2, IRGM, TNF, HLADRB1, CDKAL1 | Tsianos et al[63] | |
| Inflammatory behavior | HLA | Tsianos et al[63] | |
| Granulomatous disease | TLR4/CARD15 | Tsianos et al[63] | |
| Upper gastrointestinal | NOD2, MIF | Tsianos et al[63] | |
| Ileal | IL-10, CRP, NOD2, ZNF365, STAT3 | Tsianos et al[63] | |
| Ileocolonic | ATG16 L1, TCF-4 (TCF7 L2) | Tsianos et al[63] | |
| Colonic | HLA, TLR4, TLR1, TLR2, TLR6 | ||
| Crohn’s disease activity | HSP70-2, NOD2, PAI-1, CNR1 | Tsianos et al[63] | |
| Surgery | NOD2, HLA-G | Tsianos et al[63] | |
| Dysplasia and cancer | FHIT | ||
| Extraintestinal manifestations | CARD15, FcRL3, HLADRB103 | Tsianos et al[63] | |
| Ulcerative colitis | |||
| Extensive colitis and increased colectomy risk | HLA-DRB1 alleles, CASP9 gene on 1p36, ATG16 L1 T300A | Nam et al[64] | |
| May influence severity and steroid dependence | IL23R, STAT3, HSP70-2, MDR1 | Nam et al[64] | |
| Early response to infliximab | IL23R higher gene expression IL-17A and IFN-γ | Jürgens et al[65]; Rismo et al[66] | |
| Good response to therapy | TNF ALPHA expression | Olsen et al[67] | |
| Non response to infliximab | PR3-ANCA | Yoshida et al[68] | |
| Favorable response to treatment | FCGR3A, TNFRSF1A, IL-6, and IL-1B | ||
| Failure of steroid therapy | MDR1 (ABCB1), TNFα (-308/-857 SNPs), HLA-DQA1 05/DRB1, NOD2, ATG16 L1, IL13RA2, IL6, IL11, TNFAIP6 | ||
| Unfavorable response to therapy (IBD) | TLR2 and TLR9 show a negative correlation | Sazonovs et al[69] | |
| Development of ADA against infliximab and adalimumab | HLA-DQA1 05 | Sazonovs et al[69] | |
| Development of ADA against infliximab | HLA-DRB1 | ||
| Celiac disease | |||
| Increase severity of disease | DQA1 05 and DQB1 02, homozygous for DQ2.5 haplotype, second copy of the DQB1 0201 | Murray et al[70]; Stanković et al[71] | |
| Hereditary pancreatitis | Increased risk of disease | PRSS1 pathogenic variants include p.Asn29Ile and p.Arg122His, p.Asn29Ile and p.Arg122His | Avanthi et al[72]; Whitcomb[73] |
| Increased severity and early onset of disease | SPINK1, c.101A>G p.Asn34Ser and SPINK1, c.56-37T>C | Abass et al[74] | |
| GIST | |||
| Increase severity and relapse | Exon 11, 13, 17, c-KIT mutation; SDH deficient, BRAF mutation | Zhang and Liu[75] | |
| Colorectal cancer | Increased severity and predict recurrence | P53, KRAS codon 12, loss of 18q | Andreyev et al[76]; Walther et al[77] |
| HCC | Increased severity | EZH2, STAT3, YB-1, ANLN, NLRC5 | |
| Poor prognosis | Overexpression of CDCA5 | Wang and Lai[78]; Hashemi et al[79]; Svinka et al[80]; Chao et al[81]; Jia et al[82]; Peng et al[83] | |
| Overexpression of CDCA5 | Tian et al[84] | ||
| Gall bladder cancer | Increased severity of disease | SERPINB5 (maspin) KRAS, E-cadherin/beta-catenin, PML, P53, CDKN21 loss | Kim et al[85]; Hirata et al[86]; Chang et al[87] |
| Intra hepatic cholangiocarcinoma | Increased severity and large tumor size | BRAF | Xin et al[88] |
| Pancreatic cancer | Poor prognosis | KRAS (G12D/G12V/G12R), CDKN2A (p16), SMAD4 (DPC4) | Zhou et al[89] |
| Disorder | Gene | Drug(s) | Clinical impact | Ref. |
| CRC, gastric, pancreatic cancers | DPYD | 5-fluorouracil, capecitabine | Deficiency life-threatening toxicity (mucositis, myelosuppression) | De Moraes et al[90]; Ruzzo et al[91] |
| CRC, pancreatic cancer | UGT1A1 | Irinotecan | UGT1A1 28/28 reduced glucuronidation increased toxicity (neutropenia, diarrhea) | Maitland et al[92] |
| IBD, autoimmune hepatitis | TPMT/NUDT15 | Azathioprine, 6-MP | TPMT or NUDT15 deficiency risk of myelosuppression | Moriyama et al[93] |
| IBD | HLA-DQA102:01, HLA-DQB102:02 | Thiopurines | Increase risk of thiopurine-induced pancreatitis | Ås et al[94] |
| IBD | HLA-DQ2 | Infliximab | Increased formation of antibody formation against infliximab | Brun et al[95] |
| GERD, H. pylori, ulcers | CYP2C19 | PPIs (omeprazole, lansoprazole) | Poor metabolizers increase drug levels; rapid metabolizers treatment failure in H. pylori | El Rouby et al[96] |
| NAFLD, metabolic syndrome | SLCO1B1 | Statins (e.g., simvastatin) | Variants statin-induced myopathy risk | SEARCH Collaborative Group[97] |
| IBD | ABCB1 | Various (e.g., corticosteroids) | Associated with glucocorticoid resistance in some patients | Li et al[98] |
| Autoimmune hepatitis, liver transplant | CYP3A5 | Tacrolimus | Expressors need higher doses; non-expressors risk overexposure | Kim et al[99] |
| IBD | G6PD deficiency | Sulfasalazine, dapsone | Increase risk of hemolysis | Dore et al[100] |
| Disorder | Gene/mutation | Role | Treatment/clinical implication | Ref. |
| Colorectal cancer | KRAS (codon 12/13) | Predicts resistance to anti-EGFR therapy | Avoid cetuximab/panitumumab in mutant cases | Zhu et al[101] |
| Colorectal cancer | NRAS mutations | Similar to KRAS | Also predicts non-response to EGFR inhibitors | Hu et al[102] |
| CRC, cholangiocarcinoma | BRAF V600E | Poor prognosis, targetable | Consider BRAF + MEK inhibitors | Rizzo et al[103] |
| Gastric, colorectal cancer | HER2 (ERBB2) amplification | Targetable mutation | Responds to trastuzumab, pertuzumab | Bang et al[104] |
| CRC, gastric, biliary | MSI-H/dMMR | Biomarker for immunotherapy. Poor response to chemotherapy in stage 2 tumor | Eligible for checkpoint inhibitors (e.g., pembrolizumab) | Le et al[105] |
| HCC | CTNNB1 (β-catenin) | Resistance to immunotherapy | Poor response to immunotherapy | Shah et al[106] |
| EZH2 | Resistance to immunotherapy | Negatively express PD-L1 | Xiao et al[107] | |
| Crohn’s disease (IBD) | SNP rs396991GG of gene FCGR3A, rs976881-AA + GA (TNFRSF1B), SNPs in loci DENND1B (rs2488397) and aryl hydrocarbon receptor (rs1077773) s1813443-CC and rs1568885-TT (CNTN5) from the immunoglobulin superfamily | Resistance to biologics | Poor response to immunotherapy | Curci et al[108]; Yoon et al[109]; Ye and McGovern[110] |
| Polymorphisms in ATG16 L1 (C11orf30; rs7927894CC, CCNY; rs12777960CC) (rs10210302) | Clinical response to adalimumab | Koder et al[111] | ||
| Crohn’s disease (IBD) | Polymorphisms in NOD2 | Loss of response to anti-TNF | Juanola et al[112] | |
| UC | Polymorphisms in IL-23R | Early response to infliximab | Jürgens et al[65]; Golan et al[113] | |
| Crohn’s disease | ATG16 L1, IRGM | Autophagy pathway genes | Predict disease course and microbiome interaction | Rioux et al[114] |
| Polymorphisms in FcγRIIIa, HLA-DRB1, HLA-DQA1 05 | Development of ADA against infliximab and adalimumab | Salvador-Martín et al[115]; Billiet et al[116] | ||
| Polymorphisms in FAS, FASL, and CASP9 (apoptotic pharmacogenetic index) | Clinical response to infliximab and adalimumab | Hlavaty et al[117] | ||
| Gene protein tyrosine phosphatase non-receptor type 2 (rs7234029AG + GG, CASP9) | Non-response to anti-TNF and ustekinumab | Hlavaty et al[117] | ||
| HCC | EZH2 | Negatively regulate PD-L1 expression. Less response to PD-L1 agonist | Meng et al[118] | |
| TOP2A, PRC1 | Resistance to chemotherapy | Meng et al[118]; Wang et al[119] | ||
| IBS | TJP1, TPH1, SERT (SLC6A4) | Serotonin signaling, barrier dysfunction | May guide use of 5-HT3 antagonists or SSRIs | Camilleri et al[120]; Kerckhoffs et al[121] |
| Hereditary pancreatitis | SPINK1, PRSS1, CTRC | Trypsin regulation defects | May influence early interventions and surveillance | Panchoo et al[122] |
| Autoimmune hepatitis | HLA-DRB103, 04 | Susceptibility and severity | May predict treatment response to steroids/immunosuppressants | |
| Gastric, pancreatic, cholangiocarcinoma | ARID1A mutations | Epigenetic dysregulation | May predict response to EZH2 inhibitors or immunotherapy | |
| Pancreatic cancer | KRAS | Anti EGFR treatment in effective | Fotopoulos et al[123] | |
| hENT1 | Good response to gemcitabine therapy | |||
| DCK | Increase active form of gemcitabine and increase survival | |||
| DPD | Low DPD level associated with increase survival | |||
| hMLLH1/2 | Pancreatic cancer with MSI associated with less response to 5-FU | |||
| TS | Lower level of TS associated with better response to capecitabine and 5-FU | |||
| WOXX | Decreased expression interferes with gemcitabine sensitivity | |||
| SMAD4 (DPC4) | Poor response to chemotherapy | |||
| GBC | ARID1A | Potential sensitivity to EZH2 inhibitors or immunotherapy | Wardell et al[124] | |
| CDKN2A loss/mutation | Resistant to chemotherapy | Nakamura et al[125] |
| Gene/pathway | Targeted drug(s) | Clinical status and trial setting | Ref. |
| KRAS G12C | Sotorasib, adagrasib (+ cetuximab) | Colorectal cancer, FDA approved | Ros et al[128] |
| EGFR (mAB) | Cetuximab, panitumumab, necitumumab | Colorectal cancer, gastric, FDA approved | Xie et al[129] |
| EGFR TKI | Erlotinib, gefitinib, afatinib, osimertinib, amivantamab | Colorectal cancer, gastric cancer, FDA approved | Corvaja et al[130] |
| VEGF | Bevacizumab, aflibercept | Colorectal cancer, gastric cancer | Mahaki et al[131] |
| BRAF V600E | Encorafenib, dabrafenib | Colorectal cancer, gastric cancer | Elez et al[132] |
| CLDN18.2 | Zolbetuximab | Gastric/GEJ adenocarcinoma | Shitara et al[133] |
| NTRK fusion (NTRK1/NTRK2/NTRK3) | Larotrectinib, entrectinib | CRC, pancreatic, cholangiocarcinoma, gastric, others | Manea et al[134] |
| PD-1 (CD274 gene, checkpoint pathway) | Dostarlimab, camrelizumab1, nivolumab and pembrolizumab (keytruda) | Hepatocellular carcinoma, gastric and esophagogastric cancer | Abou-Alfa et al[135] |
| RET fusion | Selpercatinib, pralsetinib, avelumab | Rare GI/HPB tumors (cholangiocarcinoma, pancreatic) | Li et al[136] |
| FGFR2 fusion/rearrangement | Pemigatinib, futibatinib, infigratinib1 | Intrahepatic cholangiocarcinoma | Hyung et al[137] |
| IDH1 mutation | Ivosidenib | Cholangiocarcinoma | Carosi et al[138] |
| BRCA1/BRCA2, PALB2 (HRD pathway) | Olaparib (PARP inhibitor) | Pancreatic adenocarcinoma (germline BRCA) | Alhusaini et al[139] |
| VEGFR, FGFR, PDGFR, RAF (angiogenesis/multikinase) | Sorafenib, lenvatinib, regorafenib, cabozantinib, pazopanib | Hepatocellular carcinoma | Kim[140] |
| APC mutation/COX2 pathway | Celecoxib (COX2 inhibitor) | FAP | Steinbach et al[141] |
| NR1H4 (FXR nuclear receptor) | Obeticholic acid | Primary biliary cholangitis | Floreani et al[142] |
| AGXT mutation (glyoxylate metabolism) | Lumasiran (RNAi against glycolate oxidase) | Primary hyperoxaluria type 1 | Garrelfs et al[143] |
| SERPINA1 mutation (A1AT deficiency, liver disease) | Fazirsiran, ARO-AAT (RNAi) | Alpha-1 antitrypsin liver disease | Strnad et al[144] |
| ATP7B mutation | Chelators (penicillamine, trientine); zinc | Wilson disease | |
| Anti-TNF agents | Infliximab, adalimumab | IBD | Feng et al[145] |
| IL-12/23 pathway | Ustekinumab (anti-IL-12/23) | IBD | Feng et al[145] |
| α4β7 integrin/cell trafficking | Vedolizumab (gut-specific anti-integrin) | IBD | Feng et al[145] |
| JAK-STAT pathway | Tofacitinib (pan-JAK), upadacitinib (JAK1) | IBD | Liu et al[146] |
| PD-L1 antibody | Durvalumab (imfinzi), atezolizumab, tislelizumab | GBC, HCC | Li et al[147] |
| MET amplification/overexpression | Foretinib1, cabozantinib (multi-target TKIs), glumetinib1 | GBC, HCC, gastric, cholangiocarcinoma | Zhang et al[148] |
| Therapy/product | Target | Ref. |
| Alicaforsen (antisense targeting ICAM-1) (phase III) | Pouchitis, left-sided UC | Greuter et al[150] |
| Glybera (AAV1-LPL) (withdrawn) | Lipoprotein lipase deficiency (severe pancreatitis) | Ferreira et al[151] |
| Oncolytic AAV-DC-CTL (phase 1) | Stage IV gastric cancer | Yan et al[152] |
| CRISPRedited TIL therapy (phase 1 completed) | Metastatic GI cancers (colorectal, pancreas, gallbladder, esophagus, stomach) | Lou et al[153] |
| CTX131 (allogeneic, CRISPR-engineered CD70-CAR-T) (phase 1/2 trial) | Pancreatic/oesophageal cancers | Pal et al[154] |
| CAN2409 (HSV thymidine kinase gene + pro-drug) (phase 2a) | Pancreatic cancer | Garrett Nichols et al[155] |
| Mutogene cevumeran (personalized mRNA vaccine) (phase 1b) | Pancreatic ductal adenocarcinoma | Lopez et al[156] |
| GENEGUT (preclinical settings) | Crohn’s disease | Hoffmann et al[157] |
| AAVrh.10mAnti-Eos, a serotype rh.10 AAV vector coding for an anti-Siglec-F monoclonal antibody (preclinical) | Eosinophill esophagitis | Camilleri et al[158] |
| Local delivery of an adenoviral vector expressing the HSV-tk gene (aglatimagene besadenovec, AdV-tk) followed by anti-herpetic prodrug | Pancreatic cancer | Aguilar et al[159] |
| Thymidine kinase-based gene therapy | HCC | Sangro et al[160] |
| Adenovirus-mediated double-suicide gene therapy | PDAC | Lee et al[161] |
| Oncolytic virus pelareorep (reolysin) (phase 1/2 trial) | PDAC | Noonan et al[162] |
| GVAX pancreas prime and Listeria Monocytogenes expressing mesothelin (CRS-207) boost vaccines (preclinical) | PDAC | Le et al[163] |
| TNF-erade biologic (phase 1) | Esophageal cancer | Chang et al[164] |
| GNT-0003 (phase III trial) | Crigler-Najjar syndrome | |
| Pexa-Vec (JX-594) (phase 3 trial) | HCC | |
| DTX401 (AAV8-G6Pase gene therapy) (phase 3 trial) | Glycogen storage disorder 1a | |
| DTX301 (avalotcagene ontaparvovec) (phase 3 trial) | Ornithine transcarbamylase deficiency | |
| UX701 (rivunatpagene miziparvovec) (AAV9) (phase 1/2 trial) | Wilson disease | |
| VTX-802 (preclinical study) | PFIC type 2 (BSEP) |
Table 13 Gene editing techniques and their application
| Gene editing techniques | In vivo gene editing | Ex vivo gene editing |
| Technique | CRISPR-Cas system is delivered by various vectors to disease-associated cells or organs of the body to correct the mutations or treat the cause of diseases | Targeted cells of a patient are extracted, isolated, edited, expanded, and delivered back to the same patient |
| Application | Treatment of monogenic genetic disorders | Cancer immunotherapy. Treatment of hereditary diseases. Viral infection inhibition |
| Serial No. | Model/sample size | Disease | CRISPR target | Key findings | Ref. |
| 1 | Phase 1 trial; 12 patients with metastatic colorectal cancer | Metastatic CRC (human trial) | CISH knockout in autologous T cells | CRISPR-edited T cells were safe, feasible, and showed preliminary anti-tumor activity | Lou et al[153] |
| 2 | Phase 1 trial; 3 patients with advanced cancers (incl 1 GI malignancy) | Advanced solid tumors | Knockout of TRAC, TRBC, PD-1; insertion of NY-ESO-1 TCR | Demonstrated safety and persistence of CRISPR-edited T cells in humans; proof of feasibility | Stadtmauer et al[175] |
| 3 | Ongoing; sample size approximately 20 planned | Solid tumors (GI cancers included) | Endogenous TCR knockout + NY-ESO-1 TCR insertion | Designed to enhance adoptive T-cell therapy; early feasibility data available | Clinical trial (No. NCT03399448) |
| 4 | Human colon organoids | Colorectal cancer modeling | DNA repair genes (MLH1, MSH2, APC, TP53) | Sequential CRISPR editing in organoids recapitulated colorectal tumorigenesis | Drost et al[176] |
| 5 | Human intestinal organoids | Tumor suppressor modeling | PTEN, APC | High-efficiency CRISPR editing showed functional loss-of-gene effects; robust platform for GI cancer studies | Skoufou-Papoutsaki et al[177] |
Table 15 Newer techniques of gene editing tools and their applications in gastrointestinal tract disorders
| Technique | Mechanism | GIT applications |
| CRISPR-Cas9/12/13 | DNA or RNA targeting via guide RNA and nuclease | Cancer mutations (APC, KRAS), viral hepatitis, IBD models |
| Base/prime editing | Precise base or sequence correction without DSBs | CFTR mutations, APC mutations |
| ZFNs | DNA-binding proteins fused to nucleases | HBV suppression (preclinical) |
| TALENs | TALE DNA-binding fused to nucleases | Cancer cell targeting, liver disease models |
| Epigenome editing | dCas9 fused to activators/repressors | Regulation of PD-L1, IBD immune genes |
| RNAi (siRNA, ASO) | Degrade/block specific mRNAs | Lumasiran (PH1), fazirsiran (A1AT deficiency) |
| Area | Key advancements | Ref. |
| Multi-omics integration | Combined use of genomics, transcriptomics, proteomics, and metabolomics to understand complex GI diseases | Zhao et al[185] |
| Polygenic risk scores | Using multiple low-risk variants to predict risk of diseases like IBD, colorectal cancer | Cross et al[186] |
| Single-cell sequencing | Helps identify cell-specific pathways in diseases like IBD, gastric cancer | Misra et al[187] |
| Organoid models | Patient-derived GI organoids used for drug testing, personalized therapy, and gene editing studies | Yang et al[188] |
| Epigenomics | Studying methylation, histone modifications, especially in GI cancers (e.g., MLH1 methylation in CRC) | Struhl[189] |
| Artificial intelligence | AI-driven prediction models, imaging-genomics integration for early diagnosis and prognosis |
| Type | Mechanism of action | Example use | Ref. |
| Antisense oligonucleotides | Single-stranded RNA/DNA binds mRNA blocks translation or triggers degradation (via RNase H) | Alicaforsen in IBD (targets ICAM-1 mRNA) (phase 2/3 study) | Greuter et al[150] |
| Small interfering RNA | Double-stranded RNA binds to target mRNA guides RISC complex degrades mRNA | STNM01 in Crohn’s disease (fibrosis gene CHST15) (phase 1) | Suzuki et al[190] |
| mRNA replacement therapy | Synthetic mRNA encoding a therapeutic protein is delivered translated into protein | mRNA vaccines, IL-10 mRNA for colitis. Arcturus “lunar” mRNA, IL-10 mRNA LNPs (phase 1/2 study) | Qin et a[191] |
| CRISPR-Cas9 mRNA | mRNA encodes Cas9 protein + guide RNA edits DNA directly via targeted cleavage | Casgevy (CRISPR for β-thalassemia) (FDA approved) | Parums[192] |
| RNA aptamers | Structured RNA molecules bind and inhibit specific proteins or receptors | Macugen for eye disease; potential GI targets in research (preclinical) | Nagpal et al[193] |
Table 18 Messenger RNA therapy in disorders of gastrointestinal tract
| Agent/platform | Target/indication (GIT) | Study type/phase |
| RNA-4157/V940 (Moderna) | Individualized neoantigen vaccine colorectal cancer | Phase 2b/3 trial undergoing |
| BioNTech iNeST/BNT-pipeline | Personalized or fixed mRNA cancer vaccines for CRC, pancreatic, HCC | Phase 1/2 trials |
| Gritstone GRANITE | Personalized neoantigen immunotherapy MSS colorectal cancer | Phase 2 trial |
| MSK/investigator-initiated mRNA vaccine | Personalized mRNA neoantigen vaccine pancreatic adenocarcinoma | Early phase trial |
| OX40 L mRNA (LNP delivery) | Immune costimulatory agonist mRNA for HCC | Preclinical |
- Citation: Kumar A, Sarangi Y, Kaw P. Gene, genetics and genetic medicines in gastroenterology: Current status and its future. World J Gastroenterol 2026; 32(1): 112496
- URL: https://www.wjgnet.com/1007-9327/full/v32/i1/112496.htm
- DOI: https://dx.doi.org/10.3748/wjg.v32.i1.112496
