BPG is committed to discovery and dissemination of knowledge
Review
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
Table 1 Different types of epigenetic changes and their impact on gene[6-9]
Mechanism
How it works
Effect on genes with example
DNA methylation[6]Addition of methyl groups (-CH3) to DNA (usually at CpG islands)MLH1 silenced: Leads to microsatellite instability increased mutation rate
Histone modification[7]Acetylation, methylation, phosphorylation of histone proteinsCDH1 (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 chromatinLoss 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 expressionCRC (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 disordersMutations in a single gene that often follow Mendelian inheritance patternsHereditary hemochromatosis (HFE gene), Wilson disease (ATP7B gene), alpha-1 antitrypsin deficiency
Polygenic and multifactorial disordersInvolve multiple genes and environmental interactionsInflammatory bowel disease (over 200 loci have been identified), celiac disease (HLA-DQ2 and HLA-DQ8)
Cancer predisposition syndromesInherited mutations in tumor suppressor genes or DNA repair genes increase GI cancer riskLynch syndrome (HNPCC) (MLH1, MSH2), familial adenomatous polyposis (APC) gene
MosaicismTwo or more genetically distinct cell populations within the same individual, derived from a single zygoteMosaic APC gene mutations may cause attenuated forms of FAP. Very early changes in IBD
Table 3 Classification of genes based on their function and their association with gastrointestinal disorders[10-16]
Genes
Function
Associated disorders
Genes involved in DNA repair and genomic stability[10]
MLH1, MSH2, MSH6, PMS2, EPCAMMismatch repair (MMR) systemLynch syndrome colorectal, gastric, pancreatic cancer
MUTYHBase excision repairMUTYH-associated polyposis
BRCA1/BRCA2, ATM, PALB2Double-strand break repairFamilial pancreatic and gastric cancers
TP53Tumor suppressor, DNA damage responseCRC, gastric, pancreatic, hepatocellular carcinoma
Genes involved in cell adhesion and structural integrity[11]
CDH1E-cadherin (cell-cell adhesion)Hereditary diffuse gastric cancer
CTNNA1Catenin alpha-1 (adherens junctions)HDGC
SMAD4, BMPR1ATGF-β pathway mediatorsJuvenile polyposis syndrome, pancreatic cancer
Genes regulating inflammation and immune response[12]
NOD2Innate immunity, bacterial sensingCrohn’s disease
IL23R, IL10, IL12BCytokine signalingIBD susceptibility
IRGM, ATG16 L1Autophagy genesCrohn’s disease
HLA-DQA1/HLA-DQB1Antigen presentationCeliac disease
TLR4, TLR9Pattern recognition receptorsFunctional dyspepsia, IBD
Genes involved in bile acid transport and cholestasis[13]
ABCB11Bile salt export pumpPFIC2, BRIC
ABCC2 (MRP2)Bile excretionDubin-Johnson syndrome
ATP8B1Phospholipid transporterPFIC1
TJP2Tight junction proteinPFIC4
Genes in neuronal/gut motility and enteric nervous system[14]
RET, EDNRB, GDNFENS developmentHirschsprung’s disease
SCN5ASodium channel in ICCs/ENSIBS with constipation
NEUROG3Enteroendocrine differentiationCongenital malabsorptive diarrhoea
Genes affecting nutrient absorption and metabolism[15]
LCTLactase enzymeLactose intolerance
SAR1BChylomicron transportChylomicron retention disease
SLC26A3Cl-/HCO3- exchangeCongenital chloride diarrhea
SLC5A1 (SGLT1)Glucose transportGlucose-galactose malabsorption
Genes in oncogenic signaling and growth factors[15]
KRAS, NRASMAPK signalingCRC, pancreatic cancer
BRAFDownstream of KRASCRC (BRAF V600E in MSI tumors)
PIK3CAPI3K/AKT pathwayCRC, gastric cancer
EGFR, HER2 (ERBB2)Receptor tyrosine kinasesGastric, colorectal cancers
FGFR2, IDH1/IDH2Growth factor pathwaysCholangiocarcinoma
Genes related to epigenetic and transcriptional regulation[16]
ARID1AChromatin remodelingBiliary cancer, CRC, gastric
MLH3, MSH3Mismatch repair (minor MMR genes)Polyposis syndromes
TET2, DNMT3ADNA methylation regulationCRC and inflammatory epigenetic signatures
Table 4 Genetic pathways, genes, and gastrointestinal disorders[17-32]
Pathway
Key genes
Associated disorders
Mechanism/role
Ref.
Wnt/β-cateninAPC, CTNNB1, AXIN2Colorectal cancer, hepatocellular carcinoma (HCC), familial adenomatous polyposisControls cell proliferation and differentiation; mutation leads to uncontrolled growthLi et al[17]
NF-κBNFKB1, TNFAIP3, IKK complexIBD (Crohn’s, UC), gastric cancer, colorectal cancerRegulates inflammation, cell survival, immunity; chronic activation promotes inflammation and tumorigenesisPeng et al[18]
TGF-β/SMADTGFBR2, SMAD4Juvenile polyposis, CRC, pancreatic cancerControls growth inhibition and apoptosis; mutations cause evasion of tumor suppressionHata and Chen[19]
JAK/STATJAK2, STAT3, STAT1IBD, colitis-associated cancerRegulates immune cell signaling and cytokine responsesHu et al[20]
MAPK/ERKKRAS, BRAF, EGFRCRC, pancreatic cancer, gastric cancerRegulates cell proliferation and survival; mutations oncogenic signalingGuo et al[21]
PI3K/AKT/mTORPIK3CA, PTEN, AKT1, MTORCRC, gastric cancer, IBDPromotes cell growth, metabolism, and angiogenesis; dysregulation contributes to tumor growth and inflammationGlaviano et al[22]
Mismatch repairMLH1, MSH2, MSH6, PMS2Lynch syndrome, CRC, gastric cancerRepairs DNA replication errors; loss leads to microsatellite instability (MSI)Li[23]
P53 pathwayTP53CRC, esophageal, gastric, HCCControls cell cycle arrest, apoptosis, DNA repair; mutations common in late cancer stagesHarris and Levine[24]
Hedgehog signalingPTCH1, GLI1Gastric cancer, GI developmental disordersControls tissue patterning and stem cell maintenanceBriscoe and Thérond[25]
Notch signalingNOTCH1, DLL1, HES1Colitis, CRC, esophageal cancerRegulates differentiation, especially goblet cells; dysregulation affects intestinal homeostasisKopan[26]
Autophagy pathwayATG16 L1, IRGMCrohn’s disease, IBD-associated cancerMaintains intracellular bacterial clearance and mucosal homeostasisYu et al[27]
Immune checkpoint pathwayPD-L1, CTLA4MSI-high CRC, gastric cancer, IBDImmune evasion in cancer; dysregulated tolerance in autoimmune GI diseasesHe and Xu[28]
ER stress/UPRXBP1, IRE1, PERKIBD, Paneth cell dysfunction, CRCRegulates response to unfolded proteins; chronic ER stress leads to inflammation and epithelial damageChen et al[29]
IL-23/Th17 pathwayIL23R, STAT3, RORCCrohn’s disease, UC, CRCInflammatory cytokine signaling driving chronic inflammationBunte and Beikler[30]
Apoptosis/FAS-FASLFAS, BAX, CASP8Colitis-associated cancer, gastric cancerRegulates programmed cell death; evasion supports tumor survivalWaring and Müllbacher[31]
DNA repair pathways (base/nucleotide excision)OGG1, XPA, POLBCRC, gastric cancerRepair oxidative and chemical DNA damage; defects genomic instabilityKumar et al[32]
Table 5 Various types of clinical genetic testing
Classification
Type
Purpose
Mutation originGermline testingDetects inherited mutations; used for familial risk, carrier status, and predisposition
Somatic testingIdentifies acquired mutations in specific tissues (e.g., tumors); guides cancer therapy
Mosaicism testingIdentify mosaicism in FAP, IBD
Clinical purposeDiagnostic testingConfirms or rules out a specific genetic disorder in symptomatic individuals
Prognostic testingPredicts disease course, severity, or likelihood of complications
Predictive/screeningIdentifies asymptomatic individuals at risk of developing a genetic disorder
Carrier testingIdentifies individuals who carry one copy of a gene mutation (relevant for recessive conditions)
Pharmacogenetic testingAssesses genetic variants affecting drug metabolism and response
Somatic/tumor profilingDetects actionable mutations in cancer cells to guide targeted therapy and prognosis
Newborn screeningEarly 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
1Lynch syndrome (hereditary nonpolyposis colorectal cancer)ACG, NCCN, ESMOMLH1, MSH2, MSH6, PMS2, EPCAMPersonal/family history of colorectal, endometrial, or other LS-associated cancers; tumor MSI or IHC abnormality
2Familial adenomatous polyposis (FAP)ACG, NCCNAPC> 100 colorectal adenomas or family history of FAP
3Attenuated FAPACGAPCPatients with 10-99 adenomas
4MUTYH-associated polyposisACGMUTYH (biallelic)Multiple adenomas and autosomal recessive inheritance
5Peutz-Jeghers syndromeNCCN, ESMOSTK11Mucocutaneous pigmentation and hamartomatous polyps; family history
6Juvenile polyposis syndromeACG, NCCNSMAD4, BMPR1A≥ 5 juvenile polyps or family history
7Cowden syndrome/PTEN hamartoma tumor syndromeNCCNPTENGI polyps with mucocutaneous lesions or macrocephaly
8Hereditary pancreatic cancerNCCNBRCA1/BRCA2, PALB2, ATM, CDKN2A, STK11Family history of pancreatic cancer or known mutation
9Hereditary diffuse gastric cancerNCCNCDH1Family history of diffuse gastric cancer or lobular breast cancer
10Serrated polyposis syndromeWHO, ACGNo known high-penetrance genes; RNF43 under investigationMultiple serrated polyps meeting WHO criteria
Table 7 Methods for genetic testing and its clinical implication[35-55]
Test
Detects
Clinical use
Benefit
Limitation
Ref.
Cytogenetic testing
Karyotyping (conventional cytogenetics)Detects large chromosomal abnormalities: Trisomies, translocations, deletions, G-banding of metaphase chromosomesDown syndrome, Turner syndromeWhole-genome overview, identifies balanced/unbalanced rearrangementsLow resolution, cannot detect small deletions/duplications, requires dividing cellsGenetic Alliance[35]
Fluorescence in situ hybridization (FISH)Fluorescent probes bind specific DNA sequences on chromosomesDetects gene amplifications, deletions, rearrangements (e.g., HER2 in gastric cancer, ALK in GI stromal tumors)Rapid, targeted, works on interphase cellsLimited to known targets, one probe/test, cannot assess whole genomeYilmaz and Demiray[36]
Comparative genomic hybridization (aCGH)DNA from patient and control hybridized to a microarrayDetects copy number variations (e.g., deletions in polyposis syndromes, microdeletion syndromesHigh-resolution, genome-wide, detects sub microscopic CNVCannot detect balanced rearrangements (e.g., translocations), limited to CNVs onlyWeiss et al[37]
Chromosomal microarray analysisaCGH + SNP arrayUsed in syndromic GI diseases, unexplained developmental delay, congenital anomaliesGenome-wide, detects CNVs, uniparental disomy, mosaicismCannot detect balanced rearrangements, may report VUSMyllykangas et al[38]
Spectral karyotypingWhole chromosome painting with multicolor FISHIdentifies complex chromosomal rearrangements, often in cancersDetects complex karyotypes, color-coded analysisExpensive, not used for routine diagnostics, lower resolution than aCGHGuo et al[39]
Molecular genetic testing
Sanger sequencingSNV, small insertions/deletionsConfirmatory testing (e.g., known APC, MLH1 mutationsHigh accuracy for point mutation or small deletion/duplication/SNV, cost effective for single genetic testingOnly identify small subset of gene or single gene, not precisely quantifiableHerpich et al[40]
NGSPanel, exome, or genome-wide variantsMultigene panels for IBD, polyposis, CRC, gastric cancer, GISTMultiple, individually produced readings of the target area mosaism, quantitative, whole exome or genome sequencingLimited in their ability to detect copy number variations, incidental findings need to be verified by sanger sequencingSatam et al[41]
Targeted gene panelsFocused sequencing of disease-specific genesPanel specific to GIST, IBD, hereditary colorectal cancer panel, gist panelAccurate diagnosis focus on specific genes cost-effective and efficient: Can be customized according to disorderLimited coverage not detect structural rearrangements or copy number variants cannot identify novel or new gene related to diseaseMálaga et al[42]
Whole exome sequencingAll coding regionsEarly-onset or monogenic IBD, congenital diarrheal disorders (e.g., DGAT1, EPCAM mutations). Hereditary pancreatitis (e.g., PRSS1, SPINK1) colorectal cancerCost-effective WES allows deeper sequencies
WES captures approximately 85% of known disease-causing mutations
Misses non-coding variants incomplete exome coverageRabbani et al[43]; Uhlig et a[[44]
WGSCoding and non-coding genome variantIdentification of colorectal cancer genes. Undiagnosed complex diseaseCover both coding and non-coding reason detection of structural variant both germline and somatic mutationHigh cost difficult to pathogenic variant from benign variantde Voer et al[45]
MLPALarge deletions/duplicationsDetects large deletions, especially EPCAM deletions causing MSH2 inactivationEfficient CNV detection cost-effective and high throughput applicable on degraded DNACannot detect point mutations or small indels limited to pre-designed probesKuiper et al[46]; Schouten et al[47]
qPCRCopy number variations or known mutationsRapid screening for common mutations, detects bacterial, viral, and parasitic DNA/RNA rapidly and accurately, bacterial load determination in gastro intestinal disorderHigh sensitivity and specificity, rapid turnaround, quantitativeRequires prior sequence knowledgeShah et al[48]; Bamias et al[49]
Array comparative genomic hybridization (aCGH)Sub microscopic deletions/duplications, germline CNVs in genes like APC, SMAD4, and BMPR1AGenome-wide coverage, germline CNVs in genes like APC, SMAD4 and BMPR1AHigh resolution can detect CNVs as small as 50-100 kbInability to detect balanced chromosomal rearrangements difficulties in interpreting CNVs of uncertain significanceMcKay et al[50]; Assämäki et al[51]
HLA typing (PCR-SSP, NGS-based)HLA allele identificationCeliac disease, IBD pharmacogenetics IBD, primary sclerosing cholangitis drug-induced GI injury, idiosyncratic reactions to drugs causing hepatic/GI damage. Transplant compatibilityCost-effective, simple requires minimal computational supportLimited resolution may not differentiate similar alleles. May yield ambiguous resultsMegiorni and Pizzuti[52]
FISHLarge chromosomal rearrangements, gene fusionsIn 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 regionsTargeted approach only. Limited genomic coverageBrankley et al[53]
PCRSpecific known mutationsQuick detection (e.g., PRSS1 in hereditary pancreatitis), KRAS in CRCHigh sensitivity and specificity can detect minute amounts of target DNA/RNA. Rapid turnaround time. Typically, within a few hours. Quantitative provides absolute or relative quantificationRequires prior sequence knowledge. Primers must be designed for specific known targets. Cannot differentiate live from dead organisms, detects DNA from bothTol et al[54]
RNA-seqGene expression, fusion transcriptsDetects 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 cancerUnbiased and comprehensive: Captures all RNA species (mRNA, lncRNA, miRNA, circular RNA), high resolution. Detects single-nucleotide changes, splicing variants, and gene fusionsExpensive and resource-intensive, requires advanced sequencing and computational infrastructure, data analysis is complex, needs bioinformatics expertise and robust pipelinesBailey et al[55]
Table 8 Genes that help in prediction and poor prognostications[56-89]
Disease
Prediction and prognostication
Genes
Ref.
FAPProfuse polyposisAPC codon 1250-1464, 1250-1311, 1309-1324Nagase et al[56]; Enomoto et al[57]; Ficari et al[58]; Walon et al[59]; Gebert et al[60]
Desmoid tumorsAPC codon 1924, 1962, 1444-1560, 1403-1987Caspari et al[61]
Upper gastrointestinal polyps1445-1578Davies et al[62]
Gastric adenomas1403-1987Caspari et al[61]
Multiple extracolonic manifestations3’14451995, 3’1403Caspari et al[61]
CHRPE311-1444, 413-1387, 542-1309Caspari et al[61]
Crohn’s diseaseStenotic/structuring behaviorNOD2, TLR4, IL-12B, CX3CR1, IL-10, IL-6Tsianos et al[63]
Penetrating/fistulizing behaviorNOD2, IRGM, TNF, HLADRB1, CDKAL1Tsianos et al[63]
Inflammatory behaviorHLATsianos et al[63]
Granulomatous diseaseTLR4/CARD15Tsianos et al[63]
Upper gastrointestinalNOD2, MIFTsianos et al[63]
IlealIL-10, CRP, NOD2, ZNF365, STAT3Tsianos et al[63]
IleocolonicATG16 L1, TCF-4 (TCF7 L2)Tsianos et al[63]
ColonicHLA, TLR4, TLR1, TLR2, TLR6
Crohn’s disease activityHSP70-2, NOD2, PAI-1, CNR1Tsianos et al[63]
SurgeryNOD2, HLA-GTsianos et al[63]
Dysplasia and cancerFHIT
Extraintestinal manifestationsCARD15, FcRL3, HLADRB103Tsianos et al[63]
Ulcerative colitis
Extensive colitis and increased colectomy riskHLA-DRB1 alleles, CASP9 gene on 1p36, ATG16 L1 T300ANam et al[64]
May influence severity and steroid dependenceIL23R, STAT3, HSP70-2, MDR1Nam et al[64]
Early response to infliximabIL23R higher gene expression IL-17A and IFN-γJürgens et al[65]; Rismo et al[66]
Good response to therapyTNF ALPHA expressionOlsen et al[67]
Non response to infliximabPR3-ANCAYoshida et al[68]
Favorable response to treatmentFCGR3A, TNFRSF1A, IL-6, and IL-1B
Failure of steroid therapyMDR1 (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 correlationSazonovs et al[69]
Development of ADA against infliximab and adalimumabHLA-DQA1 05Sazonovs et al[69]
Development of ADA against infliximabHLA-DRB1
Celiac disease
Increase severity of diseaseDQA1 05 and DQB1 02, homozygous for DQ2.5 haplotype, second copy of the DQB1 0201Murray et al[70]; Stanković et al[71]
Hereditary pancreatitisIncreased risk of diseasePRSS1 pathogenic variants include p.Asn29Ile and p.Arg122His, p.Asn29Ile and p.Arg122HisAvanthi et al[72]; Whitcomb[73]
Increased severity and early onset of diseaseSPINK1, c.101A>G p.Asn34Ser and SPINK1, c.56-37T>CAbass et al[74]
GIST
Increase severity and relapseExon 11, 13, 17, c-KIT mutation; SDH deficient, BRAF mutationZhang and Liu[75]
Colorectal cancerIncreased severity and predict recurrenceP53, KRAS codon 12, loss of 18qAndreyev et al[76]; Walther et al[77]
HCCIncreased severityEZH2, STAT3, YB-1, ANLN, NLRC5
Poor prognosisOverexpression of CDCA5Wang 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 CDCA5Tian et al[84]
Gall bladder cancerIncreased severity of diseaseSERPINB5 (maspin) KRAS, E-cadherin/beta-catenin, PML, P53, CDKN21 lossKim et al[85]; Hirata et al[86]; Chang et al[87]
Intra hepatic cholangiocarcinomaIncreased severity and large tumor sizeBRAFXin et al[88]
Pancreatic cancerPoor prognosisKRAS (G12D/G12V/G12R), CDKN2A (p16), SMAD4 (DPC4)Zhou et al[89]
Table 9 Pharmacogenetics in gastrointestinal disorders[90-100]
Disorder
Gene
Drug(s)
Clinical impact
Ref.
CRC, gastric, pancreatic cancersDPYD5-fluorouracil, capecitabineDeficiency life-threatening toxicity (mucositis, myelosuppression)De Moraes et al[90]; Ruzzo et al[91]
CRC, pancreatic cancerUGT1A1IrinotecanUGT1A1 28/28 reduced glucuronidation increased toxicity (neutropenia, diarrhea)Maitland et al[92]
IBD, autoimmune hepatitisTPMT/NUDT15Azathioprine, 6-MPTPMT or NUDT15 deficiency risk of myelosuppressionMoriyama et al[93]
IBDHLA-DQA102:01, HLA-DQB102:02ThiopurinesIncrease risk of thiopurine-induced pancreatitisÅs et al[94]
IBDHLA-DQ2InfliximabIncreased formation of antibody formation against infliximabBrun et al[95]
GERD, H. pylori, ulcersCYP2C19PPIs (omeprazole, lansoprazole)Poor metabolizers increase drug levels; rapid metabolizers treatment failure in H. pyloriEl Rouby et al[96]
NAFLD, metabolic syndromeSLCO1B1Statins (e.g., simvastatin)Variants statin-induced myopathy riskSEARCH Collaborative Group[97]
IBDABCB1Various (e.g., corticosteroids)Associated with glucocorticoid resistance in some patientsLi et al[98]
Autoimmune hepatitis, liver transplantCYP3A5TacrolimusExpressors need higher doses; non-expressors risk overexposureKim et al[99]
IBDG6PD deficiencySulfasalazine, dapsoneIncrease risk of hemolysisDore et al[100]
Table 10 Genes which regulates response to targeted therapy[101-125]
Disorder
Gene/mutation
Role
Treatment/clinical implication
Ref.
Colorectal cancerKRAS (codon 12/13)Predicts resistance to anti-EGFR therapyAvoid cetuximab/panitumumab in mutant casesZhu et al[101]
Colorectal cancerNRAS mutationsSimilar to KRASAlso predicts non-response to EGFR inhibitorsHu et al[102]
CRC, cholangiocarcinomaBRAF V600EPoor prognosis, targetableConsider BRAF + MEK inhibitorsRizzo et al[103]
Gastric, colorectal cancerHER2 (ERBB2) amplificationTargetable mutationResponds to trastuzumab, pertuzumabBang et al[104]
CRC, gastric, biliaryMSI-H/dMMRBiomarker for immunotherapy. Poor response to chemotherapy in stage 2 tumorEligible for checkpoint inhibitors (e.g., pembrolizumab)Le et al[105]
HCCCTNNB1 (β-catenin)Resistance to immunotherapyPoor response to immunotherapyShah et al[106]
EZH2Resistance to immunotherapyNegatively express PD-L1Xiao 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 superfamilyResistance to biologicsPoor response to immunotherapyCurci et al[108]; Yoon et al[109]; Ye and McGovern[110]
Polymorphisms in ATG16 L1 (C11orf30; rs7927894CC, CCNY; rs12777960CC) (rs10210302)Clinical response to adalimumabKoder et al[111]
Crohn’s disease (IBD)Polymorphisms in NOD2Loss of response to anti-TNFJuanola et al[112]
UCPolymorphisms in IL-23R
Early response to infliximabJürgens et al[65]; Golan et al[113]
Crohn’s diseaseATG16 L1, IRGMAutophagy pathway genesPredict disease course and microbiome interactionRioux et al[114]
Polymorphisms in FcγRIIIa, HLA-DRB1, HLA-DQA1 05Development of ADA against infliximab and adalimumabSalvador-Martín et al[115]; Billiet et al[116]
Polymorphisms in FAS, FASL, and CASP9 (apoptotic pharmacogenetic index)Clinical response to infliximab and adalimumabHlavaty et al[117]
Gene protein tyrosine phosphatase non-receptor type 2 (rs7234029AG + GG, CASP9)Non-response to anti-TNF and ustekinumabHlavaty et al[117]
HCCEZH2Negatively regulate PD-L1 expression. Less response to PD-L1 agonistMeng et al[118]
TOP2A, PRC1Resistance to chemotherapyMeng et al[118]; Wang et al[119]
IBSTJP1, TPH1, SERT (SLC6A4)Serotonin signaling, barrier dysfunctionMay guide use of 5-HT3 antagonists or SSRIsCamilleri et al[120]; Kerckhoffs et al[121]
Hereditary pancreatitisSPINK1, PRSS1, CTRCTrypsin regulation defectsMay influence early interventions and surveillancePanchoo et al[122]
Autoimmune hepatitisHLA-DRB103, 04Susceptibility and severityMay predict treatment response to steroids/immunosuppressants
Gastric, pancreatic, cholangiocarcinomaARID1A mutationsEpigenetic dysregulationMay predict response to EZH2 inhibitors or immunotherapy
Pancreatic cancerKRASAnti EGFR treatment in effectiveFotopoulos et al[123]
hENT1Good response to gemcitabine therapy
DCKIncrease active form of gemcitabine and increase survival
DPDLow DPD level associated with increase survival
hMLLH1/2Pancreatic cancer with MSI associated with less response to 5-FU
TSLower level of TS associated with better response to capecitabine and 5-FU
WOXXDecreased expression interferes with gemcitabine sensitivity
SMAD4 (DPC4)Poor response to chemotherapy
GBCARID1APotential sensitivity to EZH2 inhibitors or immunotherapyWardell et al[124]
CDKN2A loss/mutationResistant to chemotherapyNakamura et al[125]
Table 11 Gastrointestinal and hepatopancreatic biliary disorders targeted therapy based on genetic testing[128-148]
Gene/pathway
Targeted drug(s)
Clinical status and trial setting
Ref.
KRAS G12CSotorasib, adagrasib (+ cetuximab)Colorectal cancer, FDA approvedRos et al[128]
EGFR (mAB)Cetuximab, panitumumab, necitumumabColorectal cancer, gastric, FDA approvedXie et al[129]
EGFR TKIErlotinib, gefitinib, afatinib, osimertinib, amivantamabColorectal cancer, gastric cancer, FDA approvedCorvaja et al[130]
VEGFBevacizumab, afliberceptColorectal cancer, gastric cancerMahaki et al[131]
BRAF V600EEncorafenib, dabrafenibColorectal cancer, gastric cancerElez et al[132]
CLDN18.2ZolbetuximabGastric/GEJ adenocarcinomaShitara et al[133]
NTRK fusion (NTRK1/NTRK2/NTRK3)Larotrectinib, entrectinibCRC, pancreatic, cholangiocarcinoma, gastric, othersManea et al[134]
PD-1 (CD274 gene, checkpoint pathway)Dostarlimab, camrelizumab1, nivolumab and pembrolizumab (keytruda)Hepatocellular carcinoma, gastric and esophagogastric cancerAbou-Alfa et al[135]
RET fusionSelpercatinib, pralsetinib, avelumabRare GI/HPB tumors (cholangiocarcinoma, pancreatic)Li et al[136]
FGFR2 fusion/rearrangementPemigatinib, futibatinib, infigratinib1Intrahepatic cholangiocarcinomaHyung et al[137]
IDH1 mutationIvosidenibCholangiocarcinomaCarosi 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, pazopanibHepatocellular carcinomaKim[140]
APC mutation/COX2 pathwayCelecoxib (COX2 inhibitor)FAPSteinbach et al[141]
NR1H4 (FXR nuclear receptor)Obeticholic acidPrimary biliary cholangitisFloreani et al[142]
AGXT mutation (glyoxylate metabolism)Lumasiran (RNAi against glycolate oxidase)Primary hyperoxaluria type 1Garrelfs et al[143]
SERPINA1 mutation (A1AT deficiency, liver disease)Fazirsiran, ARO-AAT (RNAi)Alpha-1 antitrypsin liver diseaseStrnad et al[144]
ATP7B mutationChelators (penicillamine, trientine); zincWilson disease
Anti-TNF agentsInfliximab, adalimumabIBDFeng et al[145]
IL-12/23 pathwayUstekinumab (anti-IL-12/23)IBDFeng et al[145]
α4β7 integrin/cell traffickingVedolizumab (gut-specific anti-integrin)IBDFeng et al[145]
JAK-STAT pathwayTofacitinib (pan-JAK), upadacitinib (JAK1)IBDLiu et al[146]
PD-L1 antibodyDurvalumab (imfinzi), atezolizumab, tislelizumabGBC, HCCLi et al[147]
MET amplification/overexpressionForetinib1, cabozantinib (multi-target TKIs), glumetinib1GBC, HCC, gastric, cholangiocarcinomaZhang et al[148]
Table 12 Different gene therapy under research/evaluation[150-164]
Therapy/product
Target
Ref.
Alicaforsen (antisense targeting ICAM-1) (phase III)Pouchitis, left-sided UCGreuter 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 cancerYan 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 cancersPal et al[154]
CAN2409 (HSV thymidine kinase gene + pro-drug) (phase 2a)Pancreatic cancerGarrett Nichols et al[155]
Mutogene cevumeran (personalized mRNA vaccine) (phase 1b)Pancreatic ductal adenocarcinomaLopez et al[156]
GENEGUT (preclinical settings)Crohn’s diseaseHoffmann et al[157]
AAVrh.10mAnti-Eos, a serotype rh.10 AAV vector coding for an anti-Siglec-F monoclonal antibody (preclinical)Eosinophill esophagitisCamilleri et al[158]
Local delivery of an adenoviral vector expressing the HSV-tk gene (aglatimagene besadenovec, AdV-tk) followed by anti-herpetic prodrugPancreatic cancerAguilar et al[159]
Thymidine kinase-based gene therapyHCCSangro et al[160]
Adenovirus-mediated double-suicide gene therapyPDACLee et al[161]
Oncolytic virus pelareorep (reolysin) (phase 1/2 trial)PDACNoonan et al[162]
GVAX pancreas prime and Listeria Monocytogenes expressing mesothelin (CRS-207) boost vaccines (preclinical)PDACLe et al[163]
TNF-erade biologic (phase 1)Esophageal cancerChang 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
TechniqueCRISPR-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 diseasesTargeted cells of a patient are extracted, isolated, edited, expanded, and delivered back to the same patient
ApplicationTreatment of monogenic genetic disordersCancer immunotherapy. Treatment of hereditary diseases. Viral infection inhibition
Table 14 Representative studies using clustered regularly interspaced short palindromic repeats in gastrointestinal disorders and malignancies[153,175-177]
Serial No.
Model/sample size
Disease
CRISPR target
Key findings
Ref.
1Phase 1 trial; 12 patients with metastatic colorectal cancerMetastatic CRC (human trial)CISH knockout in autologous T cellsCRISPR-edited T cells were safe, feasible, and showed preliminary anti-tumor activityLou et al[153]
2Phase 1 trial; 3 patients with advanced cancers (incl 1 GI malignancy)Advanced solid tumorsKnockout of TRAC, TRBC, PD-1; insertion of NY-ESO-1 TCRDemonstrated safety and persistence of CRISPR-edited T cells in humans; proof of feasibilityStadtmauer et al[175]
3Ongoing; sample size approximately 20 plannedSolid tumors (GI cancers included)Endogenous TCR knockout + NY-ESO-1 TCR insertionDesigned to enhance adoptive T-cell therapy; early feasibility data availableClinical trial (No. NCT03399448)
4Human colon organoidsColorectal cancer modelingDNA repair genes (MLH1, MSH2, APC, TP53)Sequential CRISPR editing in organoids recapitulated colorectal tumorigenesisDrost et al[176]
5Human intestinal organoidsTumor suppressor modelingPTEN, APCHigh-efficiency CRISPR editing showed functional loss-of-gene effects; robust platform for GI cancer studiesSkoufou-Papoutsaki et al[177]
Table 15 Newer techniques of gene editing tools and their applications in gastrointestinal tract disorders
Technique
Mechanism
GIT applications
CRISPR-Cas9/12/13DNA or RNA targeting via guide RNA and nucleaseCancer mutations (APC, KRAS), viral hepatitis, IBD models
Base/prime editingPrecise base or sequence correction without DSBsCFTR mutations, APC mutations
ZFNsDNA-binding proteins fused to nucleasesHBV suppression (preclinical)
TALENsTALE DNA-binding fused to nucleasesCancer cell targeting, liver disease models
Epigenome editingdCas9 fused to activators/repressorsRegulation of PD-L1, IBD immune genes
RNAi (siRNA, ASO)Degrade/block specific mRNAsLumasiran (PH1), fazirsiran (A1AT deficiency)
Table 16 Newer concepts in genetic medicine[185-189]
Area
Key advancements
Ref.
Multi-omics integrationCombined use of genomics, transcriptomics, proteomics, and metabolomics to understand complex GI diseasesZhao et al[185]
Polygenic risk scoresUsing multiple low-risk variants to predict risk of diseases like IBD, colorectal cancerCross et al[186]
Single-cell sequencingHelps identify cell-specific pathways in diseases like IBD, gastric cancerMisra et al[187]
Organoid modelsPatient-derived GI organoids used for drug testing, personalized therapy, and gene editing studiesYang et al[188]
EpigenomicsStudying methylation, histone modifications, especially in GI cancers (e.g., MLH1 methylation in CRC)Struhl[189]
Artificial intelligenceAI-driven prediction models, imaging-genomics integration for early diagnosis and prognosis
Table 17 RNA therapies[150,190-193]
Type
Mechanism of action
Example use
Ref.
Antisense oligonucleotidesSingle-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 RNADouble-stranded RNA binds to target mRNA guides RISC complex degrades mRNASTNM01 in Crohn’s disease (fibrosis gene CHST15) (phase 1)Suzuki et al[190]
mRNA replacement therapySynthetic mRNA encoding a therapeutic protein is delivered translated into proteinmRNA vaccines, IL-10 mRNA for colitis. Arcturus “lunar” mRNA, IL-10 mRNA LNPs (phase 1/2 study)Qin et a[191]
CRISPR-Cas9 mRNAmRNA encodes Cas9 protein + guide RNA edits DNA directly via targeted cleavageCasgevy (CRISPR for β-thalassemia) (FDA approved)Parums[192]
RNA aptamersStructured RNA molecules bind and inhibit specific proteins or receptorsMacugen 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 cancerPhase 2b/3 trial undergoing
BioNTech iNeST/BNT-pipeline Personalized or fixed mRNA cancer vaccines for CRC, pancreatic, HCCPhase 1/2 trials
Gritstone GRANITEPersonalized neoantigen immunotherapy MSS colorectal cancerPhase 2 trial
MSK/investigator-initiated mRNA vaccinePersonalized mRNA neoantigen vaccine pancreatic adenocarcinomaEarly phase trial
OX40 L mRNA (LNP delivery)Immune costimulatory agonist mRNA for HCCPreclinical