Systematic Reviews
Copyright ©The Author(s) 2021.
World J Gastroenterol. Nov 28, 2021; 27(44): 7716-7733
Published online Nov 28, 2021. doi: 10.3748/wjg.v27.i44.7716
Table 1 Summary of basic research studies (animal models)
AuthorAim of studyPPI investigatedSpecies strain, genderMethods of CRC inductionPPI treatmentExperimental/ control groupOutcome measureMain findingsMechanism studiedRole of PPI in CRC
Graffner et al[16] 1992To determine the influence of PPI-induced endogenous hypergastrinemia on growth in CRC-implanted miceOMEBALB/C mice, MMC-26 tumor cells injected SC in epigastric regionDaily for 19 d, 400 μmol/kg, PO18/18Tumor size, survival5-fold higher serum gastrin levels in OME-treated animals than controls. No differences in tumor size, tumor weight, survival and metastatic potential (61% vs 72%, P = NR) between tumor-bearing treated and control groupTrophic effect of gastrinNE
Penman et al[20] 1993To assess the influence of OME-induced hypergastrinemia on CRC development in animal modelsOMESprague-Dawley rats, F12 (weekly) SC azoxymethane (10 mg/kg/wk)Daily for 27 wk, 40 μmol /kg, PO19/20Number of tumors, position, volume; metastatic disease9-10-fold higher gastrin levels in OME-treated groups than control groups. Significantly fewer OME-treated rats developed tumors compared to control group (63% vs 95%, P < 0.02). Number of tumors were also significantly lower in OME-treated rats. Average tumor size and invasiveness of CRC was similar for both groupsTrophic effect of gastrinPE
Hurwitz et al[18] 1995To evaluate effect of omeprazole-induced hypergastrinemia on carcinogen-induced CRC in ratsOMESprague-Dawley rats, MSix (weekly) IP methylazoxymethanol (30 mg/kg) Daily for 10 wk, 40 mg/kg, gastric gavageNRNumber of tumors, volume and total tumor burden, biochemical and histological analysisSerum gastrin levels were elevated 6-fold in OME-treated animals vs controls. No differences in tumor number, tumor volume, and total tumor burden between treated and control group. No histological (crypt/mucosal height) or biochemical features in CRC-free regions of colonTrophic effect of gastrinNE
Pinson et al[17] 1995To assess if hypergastrinemia enhances progression or invasiveness of CRCOMESprague-Dawley rats, MSix (weekly) IP methylazoxymethanol (30 mg/kg) Daily for 10 wk, 14 or 40 mg/kg, gastric gavage162/108Number of tumors, volume and total tumor burden, histological analysisPlasma gastrin levels in the treated groups (low-dose OME, high-dose OME, ranitidine) were 3–5-fold higher than controls. Crypt height/mucosal height ratio of CRC-free colonic mucosa was similar between all groups. No significant differences in tumor number, tumor burden and invasiveness between OME-treated and control groups. Trophic effect of gastrinNE
Chen et al[19] 1998To examine trophic effects of endogenous hypergastrinemia colonic mucosa and transplanted colon adenocarcinoma in ratsOMESprague-Dawley rats, MInjection of K-12 cell line (Established in syngeneic BDIX rats via induction using 1,2-dimethylhydrazine)Daily for 10 d, 400 μmol/kg, PONRTumor weight and volume, histological analysis, labelling indexOME treatment and fundectomy raised serum gastrin levels by 4-5-fold. OME-treatment did not stimulate growth of transplanted tumor (K-12) cells, while fundectomy suppressed CRC growth (decreased labelling index, weight and volume of tumor) Sustained hypergastrinemia did not affect the thickness and labelling index of normal colon mucosaTrophic effect of gastrinNE
Kim et al[23] 2010To evaluate chemo-preventive properties of omeprazole in a colitis-associated CRC mouse model OMEC57BL/6 mice, FColitis induction - 15 cycles of 0.7% DSS in drinking waterNR, 10 mg/kg, IP12/24Tumor burden, biochemical and histological analysisOME-treated group developed significantly lower number of colon tumors than control groups. OME administration also resulted in decreased inflammatory markers (TNF-α, serum NO, and colon TBA-RS levels), attenuated expression of MMP, COX-2, NO synthase, and β-catenin, and greater apoptotic indexCytostatic propertiesPE
Patlolla et al[22] 2012To assess chemo-preventive effects of omeprazoleOMEF344 rats, MTwo (weekly) SC azoxymethane (15 mg/kg)9 wk, 200/400 ppm, PO30/18Aberrant crypt foci incidenceOmeprazole inhibited the AOM-induced colonic foci formation in a dose-dependent mannerCytostatic propertiesPE
Han et al[24] 2014To study the effects of PPI on colitis-associated carcinogenesisPANAPCMin/+ mice, MGenetically engineered mutation in APC geneThrice weekly for 10 wk, 8 mg/kg, IPNR/8Number and size of intestinal polypsGastrin + PPI exerted significant anti-polyposis effect through β-catenin inactivation, increased apoptosis, anti-angiogenic, and MAPK inactivation relevant to decreased levels of pro-inflammatory mediatorsCytostatic propertiesPE
Zeng et al[26] 2016To evaluate the effect of pantoprazole as TOPK inhibitor in vivo and in vitro PANNon-obese diabetic-SCID miceHCT 116 cells inoculated SC into left flankEvery 2 d for 19 d, 100 mg/kg, IP 8/8Tumor volume, immunohistochemical analysisTumors treated with PAN grew significantly more slowly, and the size of tumors was smaller compared with the control group. PAN-treated group had lower average tumor volume per mouse compared to controls (111 mm3 vs 285 mm3, P < 0.05). Average body weight was similar throughout the study indicating no toxic effects of PAN in the mice IMHC for phosphorylated histone H3 revealed substantially decreased expression in PAN-treated group compared to controlTOPK inhibitionPE
Zheng et al[27] 2017To evaluate the effect of PPI as a TOPK inhibitor in vivo and in vitro ILACB-17/Icr-scid miceHCT 116 cells inoculated SC into left flankDaily for 19 d, 150 mg/kg, PO8/8Tumor volume, immunohistochemical analysisEstimated tumor volumes of treatment groups were less than that of the control group. No toxicity or differences in body weight were observed. Expression levels of phosphorylated histone H3 were substantially decreased in ilaprazole-treated groups compared with the control groupTOPK inhibitionPE
Wang et al[25] 2017To investigate the chemosensitizing potential of PPI in CRCPANBALB/C mine, FHT29 cells injected SCWeekly for 4 wk, 30 mg/kg, IPNRTumor burdenPAN combined with 5-FU demonstrated greater inhibition of tumor growth and smaller tumor sizes compared to 5-FU aloneChemosensitizing propertiesPE
Table 2 Summary of basic research studies (colorectal cancer cell lines)
AuthorAim of studyPPI investigatedCell lines studiedOutcome measureMain findingMechanismsRole of PPI in CRC
Tobi et al[21] 1995To assess the direct effects of gastrin and OME on growth of CRC origin cells separately and in combinationOMENCI-H716, LCC-18, DLD-1Proliferation of cell linesOME treatment resulted in cytostatic effect on 1 of the 3 cell (NCI-H716) lines tested. Dose-dependent decrease in cell proliferation noted compared to controls (P < 0.05). Effect seen with gastrin (low concentration), OME, or both in combination. Gastrin increased proliferation of NCI-H716 cells only at high concentrationsTrophic effect of gastrinPE
Kim et al[23] 2010To evaluate chemo-preventive properties of omeprazole in a colitis-associated CRC mouse model OMEHT29Cell viability and growthSignificant cleavage of capsase-3 in presence of 500 μmol/L omeprazole, but effect attenuated with gastrin pre-treatment, signifying that gastrin could attenuate the cytotoxicity of PPI by decreasing apoptosis. Compared with the gastrin-treated group, cell proliferation was significantly attenuated in the presence of omeprazole (P < 0.05), suggesting that PPI could offset the trophic action of gastrin on colon cellsCytostatic propertiesPE
Patlolla et al[22] 2012To assess chemo-preventive effects of OMEOMEHCA-7, HCT-116Cell viability, cytotoxicity assays, apoptotic assaysDose-dependent suppression of cell growth and induction of apoptosis seen in both cell linesCytostatic propertiesPE
Han et al[24] 2014To study the effects of PPI on colitis-associated carcinogenesisPANHCT116Proliferation rate, apoptosis, and molecular analysisPPI antagonizes trophic actions of gastrin, causes dose-dependent suppression of cellular viability. Combination of PPI and gastrin had higher cytotoxic activity than PPI alone. PPI alone or in combination with gastrin induces apoptosis and blocks gastrin-CCKBR binding. PPI may possess anti-angiogenic activity, which inhibits the expression of angiogenic factors induced by gastrinCytostatic propertiesPE
Zeng et al[26] 2016To evaluate the effect of pantoprazole as TOPK inhibitor in vivo and in vitro PANHCT116, SW480, WiDrCell viability, TOPK assay analysis, cytotoxicity assaysPantoprazole had different cytotoxicity toward different colon cancer cells. It inhibits anchorage-independent growth of colon cancer cells. Cell line with high TOPK activity (HCT116) was more sensitive to pantoprazole. The study suggests that TOPK is a direct target for pantoprazole to suppress colon cancer cell growthTOPK inhibitionPE
Zheng et al[27] 2017To evaluate the effect of PPI as TOPK inhibitor in vivo and in vitro ILAHCT116Cell viability, TOPK assay analysis, cytotoxicity assaysIlaprazole exhibited potent inhibitory effect on growth and induced apoptosis in HCT116 cells in a dose-dependent manner. The study suggests that TOPK was a direct target for ilaprazole to suppress cancer cell growth and its anticancer activities were dependent on the TOPK expression. Inhibition of TOPK by ilaprazole is dependent on TOPK abundance in cancer cellsTOPK inhibitionPE
Wang et al[25] 2017To investigate the chemosensitizing potential of PPI in CRCPANHT29, RKOCell inhibition ratePPI in combination with 5-FU had a higher inhibitory effect on CRC cell line growth compared to controls. The study suggests that PPI may increase sensitivity of CRC tumors to 5-FU in vitroChemosensitizing propertiesPE
Table 3 Summary of epidemiological studies assessing the exposure of proton pump inhibitors in colorectal cancer patients
Author, year, placeAccrual yearStudy designGroupingNumberExposedUnexposedOR (95%CI)AdjustmentsRisk of CRC
Robertson et al[28] 2007 (Denmark)1989-2005CC CRC patients558929552941.11 (0.97-1.27)Age, sex, place of residence (matched), H2 blocker use, aspirin/NSAIDs, statins/diabetics use, history of cholecystectomy, alcoholNo increased risk
Non-CRC control 55890269253198
Van Soest et al[29] 2008 (Netherland)1996-2005CC CRC patients594535410.85 (0.63-1.16)Age, sex, calendar time, follow-up duration (matched), comorbiditiesNo increased risk
Non-CRC control 77907257065
Yang et al[30] 2007 (United Kingdom)1987-2002CC CRC patients443276936631.2 (0.8-1.9)Age, sex, alcohol, smoking, BMI, H2 blocker use, aspirin/NSAID use, calendar time, follow-up, general practice site (matched), HRT use, history of colonoscopy/flexible sigmoidoscopyNo increased risk
Non-CRC control 44292513339159
Lee et al[31] 2020 (San Francisco, United States)1996-2016CC CRC patients18595140617189NRAge, sex, ethnicity, general practitioner site, enrolment duration, smoking, alcoholism, BMI, history of colonoscopy, family history of CRC, Crohn’s disease, Ulcerative colitisNo increased risk
Non-CRC control 16012210813149309
Chubak et al[32] 2009 (Washington State, United States)2000-2003CCCRC patients641164821.7 (0.8-4.0)Age, sex, calendar time, follow-up duration (matched)No increased risk
Non-CRC control 6419471
Kuiper et al[33] 2020(Netherlands)2007-2014CCCRC patients197810419371.08 (0.97-1.21)Age, sex, calendar time, H2 blocker use, aspirin, NSAIDs, statins, antidiabetics useNo increased risk
Non-CRC control791241613751
Table 4 Summary of epidemiological studies assessing the effect of proton pump inhibitors exposure on the risk of developing colorectal cancer
Author, year, placeAccrual yearStudy designGroupingNumber of patientsDeveloped CRCDid not develop CRCHR (95%CI)AdjustmentsPPI use and CRC risk
Hwang et al 2017[34] (Korea)2007-2013PPPI users49520Total cases (including PPI users and non-PPI users) 5304NRNRSex, age, smoking, alcohol, BMI, consumption, physical activity, type 2 diabetes, CCI score, aspirin use, metformin use, stain use, socioeconomic statusNo association
Non-PPI users401764
Babic et al 2020[35] (United States)1988-2015PPPI users1320583131220.84 (0.67-1.04)Age, physical activity, BMI, family history of CRC, alcohol, smoking, history of lower endoscopy, caloric intake, vitamin D, calcium intake, regular aspirin use, folate intake, menopausal hormone therapy use, and red meatNo association
Non-PPI users1626541172161482
Lei et al 2020[36] (Taiwan)1999-2011RPPI users45382172452102.03 (1.56-2.63)Sex, age, year of index date, diabetes, coronary artery disease, HTN, dyslipidemia, COPD, cirrhosis, CCI, aspirin/NSAID use, statin use, antidiabetic useIncreased risk
Non-PPI users453829345289
Table 5 Summary of treatment studies
AuthorCenterStudy designCancer stage and typeCancer treatmentPPI use (definition)No. of patientsResults
Zhang et al[39] 2017Guangzhou, ChinaRStage II-III Rectal cancerLCRT (46 Gy, Oxaliplatin + Capecitabine (2 cycles)EOU = OME: 20 mg PO, min. OD for 6 d / 40 mg IVI, daily). EOG = total OME dose ≥ 200 mg1125EOG vs non-EOG: 1DFS (3-year) = 77.1% vs 96.6%, P = 0.032, DFS (5-year) = 69.6% vs 46.7%, P = 0.032, OS (3-year) = 82.3% vs 96.6%, P = 0.092, OS (5-year) = 76.9% vs 89.5%, P = 0.092 EOU vs non-EOU: 1DFS (3-year) = 85.5% vs 77.8%, P = 0.658, DFS (5-year) = 75.6% vs 74.6%, P = 0.658, OS (3-year) = 90.3% vs 82.5%, P = 0.754, OS (5-year) = 82% vs 77.6%, P = 0.754
Sun et al[40] 2016Edmonton, CanadaRStage I-III CRCAdjuvant Capecitabine monotherapyAny use during treatment (based on prescription data)298PPI-user vs non-users: RFS (5 years) = 74% vs 83%, P = 0.03; OS (5-year) = 81% vs 78%, P = 0.7. Multivariate RFS (5-year): HR (95%CI) = 1.65 (0.93-2.94), P = 0.09
Wong et al[41] 2019Alberta, CanadaRStage II-III CRCAdjuvant CapeOx or FOLFOX Any use during treatment (based on prescription data)389PPI-users vs non-users, RFS (3-year): CapeOX = 69.5% vs 82.6%, P = 0.03; FOLFOX = 82.9% vs 61.7%, P = 0.7; Multivariate RFS: HR (95%CI) = 2.20 (1.14-4.25) P = 0.018; OS (3-year): CapeOX = 90.1% vs 91.2%, P = 0.345, FOLFOX = 77.4% vs 80.1%, P = 0.929
Kichenadasse et al[42] 20216 clinical trialsRetrospective post-hoc analysis of RCTStage IV CRC Fluoropyrimidine-based chemotherapy (± additional agents). Regimens differed across included trialsMinimum 7 d of use during study period 5633OS: Significantly worse in PPI-users [HR (95%CI) = 1.20 (1.03-1.40)], P = 0.02; PFS: Significantly worse in PPI-users [HR (95%CI) = 1.20 (1.05-1.37)], P = 0.009 Various treatment subgroups did not influence OS and PFS
Kim et al[43] 2021China, Japan, South Korea (98 centers)Retrospective post-hoc analysis of RCTStage IV CRC mXELIRI or FOLFIRI (± Bevacizumab)Use for ≥ 20% of study period 620mXELIRI arm: No difference in OS or PFSFOLFIRI arm: Significantly better OS [HR (95%CI) = 0.5 (0.3-0.85), P = 0.11] and PFS [HR (95%CI) = 0.55 (0.33-0.91), P = 0.20] in PPI users