Review Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Dec 15, 2021; 13(12): 1956-1980
Published online Dec 15, 2021. doi: 10.4251/wjgo.v13.i12.1956
Solid extraintestinal malignancies in patients with inflammatory bowel disease
Anastasia Mala, Department of Medical Oncology, University Hospital of Heraklion, Heraklion 71110, Crete, Greece
Kalliopi Foteinogiannopoulou, Ioannis E Koutroubakis, Department of Gastroenterology, University Hospital of Heraklion, Heraklion 71110, Crete, Greece
ORCID number: Anastasia Mala (0000-0003-4289-7840); Kalliopi Foteinogiannopoulou (0000-0003-0554-5256); Ioannis E Koutroubakis (0000-0002-2773-2709).
Author contributions: Mala A, Foteinogiannopoulou K and Koutroubakis IE wrote the paper.
Conflict-of-interest statement: Mala A and Foteinogiannopoulou K have no conflict of interest. Koutroubakis IE is an Αdvisory board member for Abbvie, Astellas, Genesis, Janssen, MSD, Pharmacosmos, Pfizer, Shire and Takeda; Speaker for AbbVie, Astellas, Genesis, Janssen, MSD, Takeda and Mylan; research support Abbvie and Ferrin.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ioannis E Koutroubakis, MD, PhD, Professor, Department of Gastroenterology, University Hospital of Heraklion, Stavrakia-Voutes, PO BOX:1352, Heraklion 71110, Crete, Greece. ikoutroubakis@gmail.com
Received: April 9, 2021
Peer-review started: April 9, 2021
First decision: June 24, 2021
Revised: July 6, 2021
Accepted: August 13, 2021
Article in press: August 13, 2021
Published online: December 15, 2021

Abstract

Malignancies constitute the second cause of death in patients with inflammatory bowel diseases (IBD), after cardiovascular diseases. Although it has been postulated that IBD patients are at greater risk of colorectal cancer compared to the general population, lately there has been evidence supporting that this risk is diminishing over time as a result of better surveillance, while the incidence of extraintestinal cancers (EICs) is increasing. This could be attributed either to systemic inflammation caused by IBD or to long-lasting immunosuppression due to IBD treatments. It seems that the overall risk of EICs is higher for Crohn’s disease patients and it is mainly driven by skin cancers, and liver-biliary cancers in patients with IBD and primary sclerosing cholangitis. The aims of this review were first to evaluate the prevalence, characteristics, and risk factors of EICs in patients with IBD and second to raise awareness regarding a proper surveillance program resulting in early diagnosis, better prognosis and survival, especially in the era of new IBD treatments that are on the way.

Key Words: Extraintestinal malignancies, Crohn’s disease, Ulcerative colitis, Thiopurines, Anti-tumor necrosis factor

Core Tip: Patients with inflammatory bowel disease (IBD) are at risk of malignancies. The incidence of colorectal cancer is decreasing over time as a result of screening surveillance and better endoscopic techniques. Although IBD patients are not at high risk of overall extraintestinal malignancies compared to the general population, they have an increased risk of certain cancers. This should be kept in mind in the management of IBD patients especially in the context of immunosuppressive treatment that constitutes the cornerstone of treatment. Guidelines should incorporate all the preventive measures and be applied to everyday clinical practice for early diagnosis and better prognosis.



INTRODUCTION

Inflammatory bowel diseases (IBD), encompassing Crohn’s disease (CD) and ulcerative colitis (UC), are chronic diseases of unknown etiology. It has been proposed that in genetically predisposed patients, several environmental factors and altered gut microbiota interact, resulting in immune system dysregulation and finally to chronic intestinal inflammation[1]. IBD mainly affects the gastrointestinal tract but not exclusively, as 50% of patients experience at least one extraintestinal manifestation. In Western countries, IBD is not uncommon as the prevalence of CD is 246.7/100000 and UC is 286.3/100000[2].

Cancer is the second cause of death in IBD patients, after cardiovascular diseases[3] and 30% of IBD patients are diagnosed with malignancy at some point during the course of IBD[4]. A multicenter European study showed that the prevalence of cancer in IBD patients, during a 15-year follow-up period, was 9.1%, the majority of them were extraintestinal cancers (EICs) and the overall cancer frequency was equal to the background population[5].

However, it was recognized that patients with IBD are at increased risk of certain EICs and was also shown in other reports[6,7].

The risk of colorectal cancer (CRC) in IBD patients is 1.7-fold higher than that in the general population[3]. Lately, a reduction in the incidence of CRC in IBD has been reported that could be attributed not only to successful inflammation control due to new IBD treatments, but also to better surveillance strategies, colonoscopy techniques and implementation of guidelines supporting colectomy for high-grade dysplasia[3,4,8]. On the other hand, there is evidence that IBD patients have a higher risk of certain EICs[9-11]. EICs in IBD patients could be related either to chronic inflammation or to longstanding immunosuppressive treatment. As the incidence of EICs is increasing among IBD patients, awareness should be raised for more scrupulous surveillance for early diagnosis and curative treatment.

The aim of this review was to evaluate the prevalence, characteristics, and risk factors of EICs in patients with IBD, and to provide possible measures for prevention and early diagnosis.

METHODOLOGY

A review of the literature in PubMed using the MESH terms Inflammatory Bowel Disease AND neoplasms NOT colorectal cancer NOT intestinal neoplasms was conducted. Only studies in English language involving humans were included.

HEAD AND NECK CANCERS

Head and neck cancers (HNCs) include mainly squamous cancers of the oral cavity (OSCC), oropharynx (OPSCC), nasopharynx, hypopharynx and larynx (LSCC). Approximately 75% of HNCs are caused by alcohol and tobacco use and the remaining are mainly human papilloma virus (HPV) 16 related (HPV + HNCs)[12-17].

Only a few studies on HNCs and IBD have been published[18-21]. Danish cohorts showed no increased risk for OSCC and OPSCC[22,23], while a recent US retrospective cohort by Katsanos et al[18] showed for the first time an increased risk for OSCC [standardized incidence ratio (SIR) 9.77 (95% confidence interval (CI): 5.14-16.98)] in IBD patients. The risk was more pronounced in females (12-fold increase) compared to males (8-fold increase) and for tongue cancer (22-fold and 17-fold increase in females and males, respectively). A population-based cohort study by Mosher et al[24] also showed an increased 20-year risk of OSCC and OPSCC [20 yr relative risk (RR) 2.00 (0.95-4.22)] but not of LSCC [20 yr RR 0.48 (0.07-3.43)]. Indeed, a retrospective case-control Dutch study showed that IBD patients had a higher rate of HPV + HNCs (52.2%) compared to the general population and an impaired OSCC survival (P = 0.018), not related to immunosuppression[19]. Older age and UC diagnosis were risk factors for OSCC and OPSCC. A recent Dutch study showed that older age at IBD diagnosis (P < 0.001) as well as male sex (P < 0.001) were risk factors for LSCC in UC patients. On the other hand, tobacco use (P < 0.001), structuring (P = 0.006) and penetrating (P = 0.008) disease were identified as risk factors in CD patients who developed LSCC, while immunosuppressive medication did not influence survival[20]. Currently, studies on the prevalence of HNCs, risk factors and outcome in IBD patients are limited. Education on modifiable risk factors, such as smoking cessation, and safe sexual behavior, is the cornerstone of prevention of HNCs. In addition, given that IBD patients may have reduced immunosurveillance resulting in persisting HPV infections and cancer, oral screening and prophylactic HPV vaccines could be considered.

THYROID CANCER

Thyroid cancer (TC) is the most common endocrine cancer, and is more common in Caucasians, and young adults, predominantly females. Thyroid dysfunction, radiation exposure at a young age and heredity are known risk factors. Over 80% of all cases are of the papillary type[25].

A recent prospective multicenter Italian study showed that the prevalence of TC in IBD patients was 5.2%[26]. Several studies, have shown that TC risk was not significantly higher in IBD patients compared with the healthy population[27-29]. On the other hand, two studies reported an increased risk of TC in UC patients, the first in both sexes and the latter only in males[30,31]. A recent meta-analysis of 8 case-control studies with 334015 patients, confirmed that patients with UC had an increased risk of TC, while patients with CD did not, independent of sex and race[32]. On the contrary, a case-control study in Ohio, United States with 289935 IBD patients[33] and an Italian cohort study of 3664 IBD patients[9], reported a higher risk for TC only in CD patients [odds ratio (OR) 2.3 (1.06-5.1), P = 0.034 and SIR 5.58 (95%CI: 2.41-11.00), respectively].

Regarding the risk of immunosuppressive treatment for the development of TC in IBD, data are conflicting, with two studies reporting an increased risk in patients treated with immunosuppressants[29,31] and another two studies did not observe this relationship[28,32].

Most of the evidence is based on population-based studies[9,30-32] with many potential confounding factors, and limited studies are focused especially on the risk of TC in IBD patients[29,32,33]. There is not enough evidence to show that IBD is an independent risk factor for TC, but IBD might provide an appropriate inflammatory environment and promote development of the cancer. Further well-adjusted and population-based studies should be conducted to confirm this speculation, in order that screening strategies for TC can be recommended.

LUNG CANCER

Lung cancer is one of the most common cancers and the major cause of mortality worldwide. Several population-based studies[10,34-36] and a recent meta-analysis by Lo in 2020 showed an increased risk [incidence rate ratio (IRR), 1.53 (95%CI: 1.23-1.91), P = 0.00][37] of lung cancer in CD patients. Masala et al[38] also reported increased mortality only in CD patients [SMR 4.00 (95%CI: 1.60-8.24)]. On the contrary, in UC patients no significant risk was found in any study[31,34,36,37,39] and actually a significantly lower risk of lung cancers was reported in two studies[29,40]. The difference between CD and UC could be explained by the different association of the two diseases with smoking. In conclusion, further studies focused on lung cancer are needed and efforts should be made to guide patients to quit smoking, an important risk factor for both lung cancer and CD.

CHOLANGIOCARCINOMA

Cholangiocarcinoma (CCA) is a relatively rare EIC that mainly affects IBD patients in the context of primary sclerosing cholangitis (PSC)[41]. PSC is a chronic inflammatory, immune mediated, liver disease causing fibrosis of the intrahepatic and extrahepatic bile ducts finally leading to strictures[42]. The mechanisms of carcinogenesis in PSC are not well understood but it seems that CCA is the result of DNA damage caused by chronic biliary inflammation and bile acids in IBD patients with altered DNA repair ability[39].

Several studies have shown that IBD patients have an increased risk of biliary cancer (Table 1). A meta-analysis that included 17052 IBD patients showed that CD patients had a borderline significant increased risk of liver-biliary cancer (SIR 2.47, 95%CI: 0.95-6.46), while UC patients are at significantly increased risk (SIR 2.58, 95%CI: 1.58-4.22) and this is attributed to the occurrence of PSC in patients with IBD[10]. The incidence of CCA in PSC patients ranges between 0.5-1.5/100 person-years[42,45,46]. The risk of CCA is increased by 160-fold and PSC patients have a cumulative life-time risk of 5%-10%[44,47]. Approximately 30%-50% of CCAs are diagnosed within the first year of PSC diagnosis[45,46,48]. In another meta-analysis, it was shown that IBD patients were at increased risk of CCA (RR 2.63; 95%CI: 1.47-4.72, CD 2.69, 95%CI: 1.59-4.55 and UC 3.40, 95%CI: 2.50-4.62). In addition, further analyses concerning the site of CCA revealed that IBD patients are notably at increased risk of intrahepatic (RR 2.61, 95%CI: 1.72-3.95) and to a lesser extent extrahepatic CCA (RR 1.47, 95%CI: 1.10-1.97)[49].

Table 1 Studies on cholangiocarcinoma in patients with inflammatory bowel disease.
Ref.
Country
Type of study
Patients
Follow up time
Results
Limitations
Jussila et al[30], 2013FinlandPopulation-based study21964 IBD vs general populationMean 10.8 yrBiliary Ca UC (SIR 7.26, 95%CI: 4.37-11.1); CD (SIR 4.93, 95%CI: 1.02-14.4)Patients diagnosed in 1987-1993 and 2000-2007 were only included
Kappelman et al[35], 2014DenmarkPopulation-based cohort13756 CD, 35152 UC vs general populationCD 7.6 yr, UC 7.8 yrCD: GBC-biliary Ca (SIR 2.4, 95%CI: 1.1-4.5); UC: liver Ca (SIR 1.6, 95%CI: 1.1-2.2) GBC (SIR 2.5, 95%CI: 1.8-3.5), IBD-PSC: Liver Ca 80.0 (95%CI: 32.1-164.8), GBC 129.1 (95%CI: 47.4-281.5), IBD-non-PSC: Liver Ca 1.3 (95%CI: 0.9-1.9), GBC 2.1 (1.4-3.0)Exclusion of patients with very mild disease, no inpatient encounters prior to 1995
Ananthakrishnan et al[43], 2014United StatesMulti-institutional IBD cohort of IBD5506 CD 5522 UC 224 IBD-PSCNACCA in IBD-PSC patient (OR 55.31, 95%CI: 22.20-137.80) compared to IBD non-PSC patientsPSC diagnosis was predicted with a model. Did not separately examine the risk of large-duct over small-duct PSC
Bernstein et al[39], 2001CanadaPopulation-based study2857 CD and 2672 UC patients vs with non-IBD (1:10)14 yrLiver and biliary Ca: CD (IRR 5.22; 95%CI: 0.96-28.5, P = 0.06), UC (IRR 3.96; 95%CI: 1.05-14.9)Low percentage of the patients received IMMs
Sørensen et al[25], 2018DenmarkPopulation-based study222 PSC-IBD patients vs 8.231 IBD controlsPSC-IBD 7.4 yr, non-PSC IBD 8.4 yrCCA PSC-IBD (HR; 190; 95%CI: 54.8-660)Small number of PSC-IBD patients. Small duct PSC (8%) were included
Scharl et al[7], 2019Cohort study (IBD-Ca vs IBD-no Ca)3119 IBD patients5 yrIBD Biliary Ca (SIR 6.3, 95%CI: 1.27-18.41)Did not compute multivariate regression for Ca subtypes, IBD phenotypes

As for the risk factors of developing CCA in patients with PSC, those with prolonged IBD duration and those who underwent colectomy due to CRC or colonic dysplasia have an increased risk of CCA[50-52]. On the other hand, in a Scandinavian cohort, although IBD duration was associated with increased risk of CCA, colectomy and CRC were not[48]. CCA develops in chronic inflammation in patients with PSC-IBD and particularly in those with dominant biliary stenosis, something that seems to be a predisposing factor[41]. The definition of a dominant stenosis (DS) in PSC patients is a stricture less than 1.5 mm in diameter in the common bile duct or less than 1 mm in the left or right main hepatic duct[53]. Approximately 10%-62% of PSC patients develop a DS at some point during their disease course[54,55]. In a 25-year study of 128 PSC patients in the United Kingdom, the mean survival of the patients with DS was worst (13.7 years) than for those without a DS (23 years) and this difference was related to a 26% risk of CCA, which developed only in patients with DS. In 50% of patients with CCA, the diagnosis of CCA was made within 4 mo of the diagnosis of PSC[54]. A German study of 171 PSC patients, who were followed prospectively for 20 years, confirmed that the presence of DS in PSC is associated with a worse prognosis due to increased risk of CCA and CRC. Furthermore, this risk was directly related to the presence of underlying IBD. In total, 97 patients had DS, 20 at entry and 77 developed DS over the follow-up period. In patients with DS without IBD, no CCA developed and the survival free of transplantation was 77.8% at 18 years. On the contrary, the 18-year survival was only 23% in the PSC with a DS and IBD. On the other hand, the presence of IBD had no impact on survival in those without a DS[56,57]. The finding that the risk of CCA is related to the presence of IBD was not confirmed in a cohort of 241 Dutch PSC patients followed for a mean of 6 years[58]. As far as small duct PSC is concerned, it has not been associated with an increased risk of both malignancies (CRC, CCA)[59].

CCA is considered to result in a poor prognosis and the surveillance strategy has not been proven to be beneficial. However, it has been suggested that patients with IBD and PSC should undergo magnetic resonance cholangiopancreatography (MRCP) annually, serum CA 19-9 testing periodically and annual colonoscopy as they also have a higher risk of CRC.

HEPATOCELLULAR CARCINOMA

Hepatocellular carcinoma (HCC) mostly occurs in IBD on the background of PSC liver cirrhosis[60] or other established chronic liver diseases such as viral hepatitis. The annual incidence of HCC in cirrhotic patients is 5%, and justifies the 6-mo surveillance with liver ultrasound ± alpha-fetoprotein. Nevertheless, there have been sporadic case reports of HCC in non-cirrhotic IBD patients, mostly in CD patients treated with azathioprine[61]. Furthermore, it is of note that HCC develops more frequently in PSC-IBD patients compared to IBD non-PSC [hazard ratio (HR), 21.00][62]. However, most population-based studies address the risk of liver cancer without separating the histologic type (HCC or CCA) under the term liver or liver-biliary cancers.

GALLBLADDER CANCER

The risk of gallbladder cancer (GBC) mostly affects IBD patients in the context of PSC. Many studies have confirmed that PSC patients are at increased risk of GBC[62-70], and the prevalence is 3%-14% vs 0.35% in the general population[63]. The 10-year cumulative risk was 3% (95%CI: 1-7) in PSC-IBD compared to 0% in non-PSC IBD patients[45]. Furthermore, a study that evaluated 72 gallbladders from 100 liver explants-o (OLT) due to PSC showed that GBC was associated with intrahepatic bile duct dysplasia (P = 0.001), CCA (P = 0.023), and IBD (P = 0.03) and gallbladder dysplasia was associated with hilar/intrahepatic bile duct dysplasia (P = 0.0006), CCA (P = 0.028), IBD (P = 0.0014), and older age at OLT (P = 0.007)[71].

A Danish study showed that CD patients had a significant increased risk of liver-GBC and UC patients had a more notable risk (SIR 2.5, 95%CI: 1.8-3.5) compared to the general population. Furthermore, this was profound for UC-PSC (SIR 129.1; 95%CI: 47.4-281.5) than in those without PSC (SIR 2.1; 95%CI: 1.4-3.0)[35]. This indicates the need for annual surveillance with ultrasound of the gallbladder in PSC-IBD patients.

PANCREATIC CANCER

Pancreatic cancer (PC) is the seventh leading cause of cancer death worldwide with increasing incidence and mortality, and is considered to have an unfavorable outcome and prognosis.

In a recent large Scandinavian study with 161926 IBD patients, 442 (0.27%) were diagnosed with PC compared with 3386 (0.21%) of the 1599024 IBD-free individuals. The 20-year cumulative incidence was 0.34% (95%CI: 0.30-0.38) vs 0.29% (95%CI: 0.28-0.30), while the IR was 22.1 (20.1-24.2)/100000 person-years in IBD patients. The overall HR was 1.43 (1.30-1.58) [CD 1.44 (1.18-1.74), UC 1.35 (1.19-1.53), IBD unclassified was 1.99 (1.50-2.64)], whereas in IBD-PSC it was 7.55 (4.94-11.5)[72]. In a Korean study, the risk was only increased in females with CD (SIR 8.58; CI: 1.04-31.00)[30]. On the contrary, a previous meta-analysis and several population-based cohort studies have shown that the risk of PC is non-significantly increased in IBD patients [pooled SIR 0.51 (0.06-4.57) and 0.75 (0.30-1.87) accordingly][7-10]. IBD-PSC patients should be referred separately since they are at a higher risk of PC[62]. A Swedish study, which included PSC patients (79% had concurrent IBD), showed that they had a 14-fold greater risk of PC than the background population[46]. In another study of 224 PSC-IBD patients, a significant increased risk was confirmed (OR 11.22, 95%CI: 4.11-30.62) compared to IBD non-PSC patients[43]. Given that higher CCA risk in PSC is well established, we could assume that the increased PC risk might be due to misdiagnosis of periampullary cancer.

GASTRIC MALIGNANCIES

The cause of gastric cancer (GC) in IBD patients is uncertain. GC usually develops on the background of intestinal metaplasia and dysplasia due to Helicobacter pylori (H. pylori) infection and chronic inflammation[73]. Nevertheless, the prevalence of H. pylori in IBD patients is low[74], while a possible causative relation could be attributed to the upper gastrointestinal involvement of CD. A previous review and meta-analysis study, found that CD patients had a significantly increased risk of cancer of the upper gastrointestinal tract (SIR 2.87, 95%CI: 1.66-4.96) and of the stomach per se (SIR 2.05, 95%CI: 1.06-3.97)[10]. However, most recent population-based studies[7,11,30,35,39,75] did not confirm this finding. With regard to the risk factors and survival of IBD patients with GC, a study that included 59 GC cases in IBD individuals, showed that UC was more frequent among these patients (69.5% vs 51.4%; P < 0.01) compared with 177 IBD controls, and IBD patients with GC showed reduced survival (P = 0.035; HR 1.385, CI: 1.023-1.875) compared with 1534 individuals of the general population with the same malignancy[76]. Studies on GC in IBD patients are limited and no secure conclusions can be drawn.

BREAST CANCER

Breast cancer (BC) is the second most common malignancy diagnosed worldwide and its mortality is decreasing over time due to screening protocols and improved therapy. Increasing age, prolonged estrogen exposure, obesity in postmenopausal women, reproductive and genetic factors, western life-style, smoking and alcohol are known risk factors[77].

Limited data are available on the pathogenesis of BC in IBD[78]. IBD patients seem to have a shorter period of estrogen exposure than the general population, with later onset of menarche and earlier menopause, that could be related to a lower BC risk[78,79].

No studies primarily investigating the risk of BC in IBD are available. Data, generally not adjusted for confounding factors, can be derived mainly from population-based cohort studies investigating the general and type-specific risk of malignancies[6,15,30,36,39,75,80-88]. In 2012, Hemminki et al[84] showed a decreased risk for BC only for CD patients (SIR 0.85, 95%CI: 0.75-0.97) and a decreased mortality rate (HR 0.75, 95%CI: 0.58-0.98), and similar results were seen in the Dutch IBD-SL cohort (n = 1157; SIR 0.11 (95%CI: 0.00-0.64)[15] and the TREAT registry cohort study [n = 2975; SIR 0.28 (95%CI: 0.08-0.72)][86]. On the other hand, a Swedish cohort study conducted in 21788 CD patients showed a 30% higher BC risk in hospitalized patients between 45 and 64 years, possibly reflecting the advanced risk at older age or the exposure to IBD treatment or unregistered confounders[83]. Similarly, a retrospective Taiwanese cohort study by Tsai in 2014 indicated that IBD is not associated with increased BC risk (aHR 0.95 (95%CI: 0.66-1.36); however, there was an association between the frequency of IBD-related hospitalizations and BC risk in patients less than 65 years old (aHR 8.45; 95%CI: 4.64-15.4)[87]. An increased risk of BC in both UC and CD was reported by the IBSEN study in 2016[88]. A large cohort study[36], a Finish register study[30], and two meta-analyses, by Pedersen et al[10] and by Lo et al[37], showed no difference in the occurrence of BC in both UC, CD and the general population, but data on treatment modalities were lacking.

No association between the use of thiopurines and BC has been reported by cohort studies and small series[22,75,89-93]. The correlation between BC and the use of biologics in IBD patients has been addressed in several reports[94-98]. A large nationwide Danish register-based cohort study with 56146 IBD patients and a long follow-up, showed no increased BC risk for patients treated with infliximab, adalimumab or certolizumab[94]. In the TREAT Registry cohort study consisting of CD patients, a decrease in the occurrence of BC was observed in both patients exposed and non-exposed to infliximab [SIR 0.50 (95%CI: 0.24-0.92) and SIR 0.32 (95%CI: 0.12-0.70), respectively], compared to the SEER database of the general United States population[86]. No association for vedolizumab was also reported in a study involving 2830 IBD patients with a follow-up period up to 5 years[95]. In addition, no increased risk of BC in IBD patients exposed to adalimumab monotherapy or combination therapy with thiopurine or methotrexate was suggested in a pooled analysis of 1594 CD patients[96]. Similar results were shown in a pooled analysis of 5 small studies and 5 landmark trials comprising 2385 IBD patients[99] and by a meta-analysis, involving 22 randomized controlled trials comparing anti-tumor necrosis factor (TNF) therapy vs placebo, even if only 4 trials had a low risk of bias[100].

In conclusion, the risk of BC is probably decreased in CD patients and seems to be similar to the background population in UC patients. IBD and anti-TNF therapy do not seem to affect BC incidence; however, for thiopurines and combination therapy, this is still uncertain. Future studies on the pathophysiological association between BC and IBD may help identify populations at high risk for BC, who might therefore benefit from close surveillance.

CERVICAL NEOPLASIA

Cervical neoplasia (CN) consists of dysplasia or cervical intra-epithelial neoplasia (CIN) and invasive cervical cancer (CC)[101]. High-risk HPV infection (types 16, 18, 45, 31) is considered the causal agent of CN, whereas smoking, sexual, socioeconomic, and immunologic co-factors may contribute to persistent infection[102]. Most low-grade lesions regress spontaneously but most high-grade dysplasia and CIN 2/3 do not[103,104]. While HPV vaccines and Papanicolaou (Pap) smear have substantially reduced CC incidence and mortality, these are still high in developing countries and CC remains the second most common cancer in women worldwide[105-107].

The risk of CN in patients with IBD remains controversial and the role of immunosuppressants, a known risk factor for cervical dysplasia in other immune-mediated diseases, is not clear[108,109]. It may be hypothesized that the underlying immunologic changes in IBD or immunosuppressive drugs may reactivate HPV from a latent status or decrease HPV clearance and CIN regression or make HPV vaccination less effective.

Data on the association between IBD, treatment and CN are conflicting[31,75,91,110-119] (Table 2). Also, non-conclusive are the results of two meta-analyses. Allegretti in 2015, in a meta-analysis of 8 studies with 77116 IBD patients but also heterogeneity, found that IBD patients had an increased risk of high-grade dysplasia/cancer compared to healthy controls (OR 1.34, 95%CI: 1.23-1.46) and the risk was greater with the use of immunomodulators, corticosteroids and 5-aminosalicylic acids, but not with anti-TNF[120]. A recent meta-analysis by Lo et al[37] in 2020 reported no statistically increased risk for CC in IBD patients.

Table 2 Studies on cervical neoplasia in patients with inflammatory bowel disease.
Ref.
Country
Type of study
Patients
Results
Limitations
Connell et al[91], 1994United KingdomSingle centre-registry study, Prospective (1962-1991)755 IBD patients taking AZACC: SIR 4.00Small power of the study to detect an increased risk (expected 0.5). No variables analysed
Bhatia et al[110], 2006United StatesSingle centre-cohort study, Retrospective116 IBD patients 116 age-matched healthy controlsAbnormal Pap smears: 18% vs 5%; P = 0.004. Non association with IBD type or treatment exposureInter-observer variability in the smear interpretation. Recall bias (questionnaires)
Kane et al[111], 2008United StatesSingle centre-cohort study (2004-2005)8 UC; 32 CD. 120 controls (age, race, parity-matched). 58% exposed to ≥ 1 treatment (prednisone, AZA, 6-MP and/or IFX)Abnormal Pap smear 42.5% IBD vs 7% controls; OR 3.4 (95%CI: 1.7-12.1, aP < 0.001), low-grade lesions OR 2.2 (95%CI: 1.7-4.4, aP < 0.001), high-grade lesions OR 3.1 (95%CI: 1.3-8.7, aP < 0.001). Exposed vs non-exposed: Abnormal Pap smear OR 1.5 (95%CI: 1.2-7.1, bP = 0.02), High-grade lesions OR 6.5 (95%CI: 1.43-30.1, bP < 0.05), IMMs > 6 mo OR 1.9 (1.1-12.1, bP < 0.001)Small population study. Level of immune suppression not assessed
Hutfless et al[112], 2008United StatesNested case- control study (1996-2006)UC 778; CD 476; Controls 12124. TP monotherapy (AZA, 6-MP, MTX)CC: aOR 1.45 (95%CI: 0.74-2.84), ASA: OR 1.65 (0.34-7.98), corticosteroids: OR 2.79 (0.71-11.00), IMMs: OR 3.45 (0.82-14.45)Missing data for race, ethnicity, smoking status. No adjustment for therapy or disease severity. Diagnostic or screening Pap smears are not distinguishable
Marehbian et al[113], 2009United StatesInsurance claims-based United States population (2002-2005)CD 22310; controls 111550IBD vs controls: Cervical dysplasia/HPV: RR 1.35 (1.28, 1.43). Exposed vs non exposed to treatment: aHR Steroids 1.11 (0.59, 2.09), IS 1.77 (1.26, 2.49), anti-TNF 1.30 (0.68, 2.51), Combination 1.81 (1.10, 3.10)Disease severity is a possible confounding factor. Limited amount of person-time on various treatments
Lees et al[114], 2009ScotlandTertiary centre, case-control study RetrospectiveUC 178; CD 184; Healthy controls 1448Abnormal Pap smear: IBD patients OR 0.82 (95%CI: 0.59-1.1.3), IBD patients current smokers vs ex or no smokers OR 2.95 (95%CI: 1.55-5.50); P = 0.001, Women < 20 yr at IBD diagnosis vs 20-39 yr vs > 40 yr: 27% vs 13.4% vs 5.0% (P = 0.001). No effect of IS therapyNo data on HPV status or exposure to corticosteroids. Small number of patients on MTX or anti-TNF. Median time exposed to IS 2.4 yr. Smoking may be a confounding factor
Singh et al[115], 2009CanadaPopulation-nested case-control (2002-2006)UC 233; CD 292; Controls 57898Cervical abnormalities: OR 1.41 (1.09-1.81), IS+ steroids 1.41 (1.09-1.81). High risk lesions: IS monotherapy aOR 1.23 (0.57-2.63), IS + steroids aOR 1.28 (0.77-2.12), CD patients exposed to > 10 prescriptions of oral contraceptives OR, 1.66 (1.08-2.54), UC OR 1.03 (0.77-1.38)Administrative databases. Possible bias is smoking, parity and sexual factors. Small number of patients exposed to IMMs alone. No data on HPV infection. CN confirmed histologically in 19% of cases
Jess et al[75], 2013DenmarkPopulation-nested case-control (2002-2006)1437 UC; 774 CD. UC median F.U. 15 yr (0-33); CD 14 yr (0-33). 1978-2010 Population-based cohort study (1978-2010). 25176 IBD patients; UC 1437; CD 774(UC median 15yrs/22 582 pt-yrsCD median 14yrs/11 261 pt-yrs)TP (18% of UC pts,45% of CD pts everused TP)Cervical dysplasia/CC: SIR 1.65 (95%CI: 1.10-2.37), CD diagnosed at age 0-19 yr: SIR 2.52 (95%CI: 1.26-4.51), Smokers: SIR, 2.15 (95%CI: 1.27-3.40), 5-ASA: SIR, 1.69 (95%CI: 1.08-2.51), Thiopurines: SIR, 2.47; (95%CI: 1.54-3.73)No detailed pharmaco-epidemiological analyses. Cervical dysplasia was analysed along with CC resulting in higher CC incidence
Rungoe et al[116], 2015DenmarkNationwide population-based cohort (1979-2011)27408 IBD patients (UC 18691; CD 8717), controls 1508334, median F.U: UC. 7.8 yr; CD 8.3 yrCD: CC IRR 1.53 (95%CI: 1.04-2.27), High grade lesions IRR, 1.28 (95%CI: 1.13-1.45), low grade lesions IRR, 1.26 (95%CI: 1.07-1.48), CN significantly higher risk in CD patients diagnosed at young age and treated with AZA. UC: CC IRR 0.78 (0.53-1.13), High-grade lesions IRR 1.12 (95%CI: 1.01-1.25), Low-grade lesions: IRR 1.15 (95%CI: 1.00-1.32)Data for smoking not available. Possible confounding factor is disease severity. Vaccination policies and screening may influence risk estimation
Kim et al[119], 2015United StatesCohort U.S. insurance data (2001-2008 and 2003-2012)133333 SID patients, including 25176 IBDHigh grade dysplasia/CC: aHR 1.72 (0.66-4.45). IS: aHR 1.72 (95%CI: 0.66-4.45)Confounding factors (race, ethnicity, socioeconomic status, sexual behavioural, gynaecologic history). Study not designed to determine the comparative effect of IS drugs. Short follow-up (mean 2.1 yr)
Jung et al[31], 2017KoreaNational Health Insurance claims (2011-2014)IBD (5595 CD and 10049 UC)CC: UC 5.65 (2.44-11.13)Did not focus on cancer occurred during IBD treatment. Missing data (disease diagnosis, phenotype). Short follow-up
Segal et al[117], 2021United KingdomHospital Episode Statistics database (1997-2012)837 with IBD, 61648 control patientsIBD vs controls: CC: 5.2 of 100000 vs 4.6 of 100000; P = 0.042Other possible mechanism of carcinogenesis (other than HPV) not evaluated. Database accuracy. HPV-related cancers were not considered separately in CD and UC
Li et al[118], 2019ChinaProspective study (2014-2017)124 IBD patients and 372 controlsHPV 16/18 infection OR 29.035 (3.64-210.988) P = 0.001, HPV-infection rate: MTX OR 4.76 (1.471-15.402) P = 0.005, > 2 IS OR 3.64 (1.255-10.562) P = 0.013, CIN prevalence: 3.2 vs 0.0%, P = 0.004No data on sexual behaviour in control group

Regarding screening adherence some studies indicated that this is influenced by various factors such as the state system, increased age and use of immunosuppressants and there is no significant difference with non-IBD patients[121-123]. Singh showed that women with IBD in Canada had a low (54%) screening adherence and older age, lower socioeconomic status, lower intensity of healthcare utilization, CD and exposure to immunosuppressant medications were independent predictors of lower use of Pap testing[121]. In the United States, Long et al[122] and Xu et al[123] showed higher adherence (70%).

In many health care systems, it is not clear who has the primary responsibility for CC prevention. As incidence and mortality of CN are highly dependent on screening and treatment of precursor lesions, an inter-specialist cooperation is suggested to guide IBD patients to follow all preventive measures such as HPV vaccination, between 9 and 26 years and prior to the initiation of the sexual activity, in both men and women, annual screening testing for chronically immunocompromised patients and smokers starting at age 21, safe sexual practices and smoking cessation[124].

OTHER GYNAECOLOGICAL CANCERS

No studies primarily investigating the incidence and mortality risk of endometrial, ovarian, or vulvar-vaginal carcinoma in IBD have been identified. Data extracted from large cohort studies investigating the general risk of malignancies, an older meta-analysis and a recent meta-analysis in 2020 showed no increased risk of these cancers even when immunosuppressive medication was used[10,29,31,34,35,37,39,40,80-86,125-127].

URINARY TRACT CANCERS

The urinary tract cancers (UTC) include bladder cancer, that occurs at advanced age, predominantly in males, smokers or in association with chronic inflammation, and renal cell carcinoma (RCC), that is related to smoking, obesity, and hypertension[128-131]. Elevated levels of TNF, a key mediator of cancer-related inflammation, have been reported in the early stages of RCC due to loss of the Von Hippel-Lindau tumor-suppressor gene[132,133].

The prevalence of UTC in IBD patients in a recent, multicenter, prospective Italian study was 9.6% (6.3% in CD and 13% in UC) with main risk factors being the disease duration (in UC) and the use of immunomodulators[134,135]. There are some reports (Table 3) and a recent meta-analysis of 15 studies by Lo et al[37] of the non-association between IBD and UTC [Urological: CD IRR 1.34 (0.91-1.98) P = 0.14, UC IRR 1.01 (0.82-1.25) P = 0.92; RCC: CD IRR 1.93 (0.80-4.65) P = 0.14, UC IRR 0.84 (0.33-2.10) P = 0.71][30,35,37,39,75]. However, a case-control study conducted in veteran patients in Texas found an increased risk of RCC in IBD patients[24]. Young IBD patients with a severe disease course also had an increased risk of RCC in a large Dutch cohort study[136]. These patients had a better outcome compared to the general population, probably related to incidental RCC diagnosis at lower stage and younger age due to frequent abdominal imaging. Another key finding of this study was that the use of immunosuppressive or anti-TNFα therapy did not adversely affect RCC disease-free survival and overall survival[136].

Table 3 Studies on urinary tract cancer in patients with inflammatory bowel disease.
Ref.
Country
Type of study
Patients
Results
Limitations
Bernstein et al[39], 2001CanadaPopulation-based-cohort study (1984-1997)5529 IBD patients; Median F.U. 7.9 yr (3.5-12)Bladder Ca: CD IRR 1.30 (0.51-3.30) UC IRR 0.67 (0.24-1.85) IBD IRR 0.92 (0.47-1.82). RCC: CD IRR 1.02 (0.31-3.34) UC IRR O.8 (0.25-2.58) IBD IRR 0.89 (0.39-2.06)Possible confounding factor is socioeconomic status. Maximum F.U 12 yr. Data not analysed by extent of disease
Pasternak et al[22], 2013DenmarkCohort study (1997-2008)45986 patientsAZA: Current users RR 2.8 (1.24-6.51), former users: RR 1.73 (0.70-4.24)Possibly confounded by indication
Jess et al[75], 2013DenmarkPopulation-nested case-control (2002-2006)1437 UC; 774 CD; Median F.U. UC 15 yr (0-33); CD 14 yr (0-33)CD SIR 1.69 (0.68-3.49); UC SIR 1.08 (0.56-1.89)No detailed pharmaco-epidemiological analyses
Jussila et al[30], 2013FinlandCohort study (1987-1993 and 2000-2007 and followed up to 2010)21964 patients with IBDUrological CD IRR 1.56 (0.58-4.21), RCC CD IRR 1.61 (0.62-4.17)Possibility of misclassification of IBD, CD, UC, and Ca
Kappelman et al[35], 2014DenmarkPopulation-based-cohort study (1978-2010)48908 IBD patientsBladder Ca CD SIR 1.1 (0.8-1.6), RCC CD SIR 0.98 (0.77-1.23)Not age-and sex specific estimates of absolute Ca risk. Detection bias. Data possible lacking. No inpatient encounters prior to 1995
Algaba et al[85], 2015SpainProspective-cohort study (2005-2011)590 IBD patients; Controls 222219Bladder Ca RR 5.23 (1.95-13.87)Small number of cases and limited period of follow-up
Nyboe Andersen et al[94], 2014DenmarkCohort study (1999-2012)56146 IBD patients; Median F.U. 9.3 yr (4.2-14)Anti-TNF: aHR 1.60 (0.61-4.19)Confounding by indication, smoking, missing data. Short median F.U. of anti-TNF exposed (3.7 yr). Small number of Ca did not permit subgroup analysis
Bourrier et al[138], 2016 (CESAME study)FranceProspective-cohort study (2004-2005)19486 IBD patients (30.1% receiving TP)Bladder Ca SIR 1.20 (0.44-2.61); RCC SIR 2.05 (0.98-3.77), AZA > 65 yr vs < 50 yr HR 13.26 (3.52-50.03, P = 0.0001), Current users: SIR 3.40 (1.47-6.71; P = 0.006), Ex-users: SIR 0.64 (0.01-3.56)Smoking is a possible confounding factor. Risk of anti-TNFs not assessed. Short follow-up
Wauters et al[139], 2017BelgiumRetrospective case–control study (1990-2014)RCC; Exposed to anti-TNF: 2083 IBD patients (952 men and 1131 women); Un-exposed to anti-TNF: 1952 (977 men and 975 women)Un-exposed to anti-TNF males SIR 5.4 (2.9-9.2), females SIR 8.5 (3.7-16.8) Exposed to anti-TNF males SIR 7.1(2.3-16.5), females SIR 4.8 (0.6-17.3)Potential confounding factors were not adjusted. Disease type, severity, and drug exposure of hospitalized patients may not be comparable with the global patient population. Different agents and dose-response for anti-TNF were not studied
Mosher et al[24], 2018United StatesCase-control study Veteran population (1996-2015)2080 patients with IBD; 271898 without IBDBladder Ca 20 yr RR 1.72 (0.86-3.45); RCC 20 yr RR 2.90 (1.46-5.84)Administrative data. Possible underestimation of Ca incidence (newly diagnosed Ca treated in other centres)
Derikx et al[136], 2015HollandCase-control study (1991-2013)Case control study A: 180 IBD patients with RCC vs 1800 IBD patients; Case control study B: 180 IBD patients with RCC vs 4388 patients with RCC in the general populationCase control A: Montreal E3 UC OR 1.8-2.5 (95%CI: 1.0-5.3), penetrating IBD-CD OR 2.8 (95%CI: 1.3-5.8), IBD related surgery OR 3.7-4.5 (95%CI: 1.6-8.2), male gender OR 3.2-5.0 (95%CI: 1.7-13.2). Case control B: lower age at diagnosis RCC (P < 0.001), lower N-stage (P = 0.025), lower M-stage (P = 0.020), more frequent surgical treatment for RCC (P < 0.001), better survival (P = 0.026; HR 0.7)Retrospective data collection. Selection bias (different registries and databases)
Biancone et al[26], 2020ItalyMulti-centre nested case-control study prospective (2011-2017)403 IBD patients; 806 IBD controlsUC: OR 3.79 (1.27-16.2)Referral IBD centres included more severe patients compared with community-based centres

With regard to bladder cancer, an increased risk in IBD patients was clearly demonstrated by a meta-analysis of eight population-based cohort studies with 17052 IBD patients in 2010 (SIR 2.03, 1.14-3.63)[10] and a prospective Spanish cohort study[85]. Moreover, long-standing IBD and use of immunosuppressive medication were found to be associated with the development of bladder cancer[137]. It is noteworthy that a reversible increased risk of UTC in patients treated with azathioprine has been reported by two large studies[22,138].

Cancer risk assessment in patients under anti-TNF therapy is problematic due to frequent combined treatment with thiopurines. Anti-TNF monotherapy does not increase either UTC risk, according to the results of a population-based prospective cohort nationwide Danish study of more than 56000 IBD patients[94] or RCC risk, independent of diagnosis age, type, and duration of IBD or coexistence of known RCC risk factors[139].

There are currently no screening guidelines for UTC in any population, while incidental detection of RCC has been identified as a positive prognostic index[140,141]. Whether patients with a long IBD duration may require different surveillance programs for preventing UTC needs to be confirmed, but all CD patients should be encouraged to quit smoking as tobacco could be a key risk factor[128,142,143]. Limited evidence is available on the role of immunosuppressive therapy in the development of UTC; however, elderly men on thiopurine therapy, should be closely evaluated for UTC.

PROSTATE CANCER

Prostate cancer (PC) is the second most common cancer in men worldwide, with screening tools available for early diagnosis[144]. The most important risk factors are age, African American ethnicity, genetic and possibly dietary factors.

Some studies have indicated an increased risk of PC in IBD, especially in UC, while others did not confirm this[24,29-31,35,36,39,40,75,83,84,144-147] (Table 4). Intensive surveillance in IBD patients with digital rectal and prostate examination may be a reason for the high incidence of PC, but also potential pathophysiological mechanisms of IBD and PC association have been recognized. In principle, the distinct microbiome level between UC and CD may be related to the elevated risk of PC in UC[148-150]. In addition, simultaneous elevation of IBD-related inflammatory markers, such as C-reactive protein, and of prostate-specific antigen (PSA), known prostatic inflammation or PC index has been reported[151-155]. Receptors of pro-inflammatory cytokines, such as interleukin 6 (IL-6)[156-158], and folate hydrolase 1 (FOLH1)/prostate specific membrane antigen (PSMA) are up-regulated in both IBD and PC and in fact FOLH1/PSMA appears overexpressed in cases of biochemical recurrence with an increase of PSA and metastatic disease[159-163].

Table 4 Studies on prostate cancer in patients with inflammatory bowel disease.
Ref.
Country
Type of study
Patients
Results
Limitations
Karlén et al[40], 1999SwedenCohort (1955-1989)1547 UCUC 7/1547; SIR 0.7 (0.3-1.5)Missing data. Closer monitoring of UC patients may lead to higher frequency and early detection
Bernstein et al[39], 2001CanadaCohort (1984 -1997)5529 IBD-1151000 controlsIBD 26/5529 SIR 0.86 (0.59-1.26), 6293/1151000 controlsPossible confounding factor is socioeconomic status. Maximum F.U 14 yr. Data not analyzed by extend of disease
Winther et al[145], 2004DenmarkCohort (1962-1997)1160 UC patients; F.U. median 19 yr4/1160 UC; UC SMR 0.74 (0.20-1.88)The treatment principles remained unchanged during the entire follow-up period
Hemminki et al[84], 2012SwedenCohort (1964-2004)27606 UC patientsUC 277/27606 SIR 1.14 (1.01-1.28), All + 1; SIR 1.08 (0.95-1.22)Possible incidental finding of PC in older UC patients
Hemminki et al[83], 2009SwedenCohort (1964-2004)21788 CD patientsCD 152/21788; SIR 1.19 (1.01-1.4), All + 1 SIR 1.12 (0.94-1.32)The sparseness of individual cancers did not allow conclusions about the trends
Jess et al[75], 2013DenmarkCohort (1978-2010)1437 UC; 774 CDUC SIR 1.82 (1.17-2.71)No detailed pharmaco-epidemiological analysis
Jussila et al[30], 2013FinlandCohort (1987-1993 and 2000-2007)21964 IBD (16649 UC; 5315 CD); 5351000 controlsIBD 176/21964; 51045/5351000 controls; IBD SIR 0.84 (0.73-0.97) UC 150/16649 SIR 0.85 (0.72-0.99) P < 0.05; CD 26/5315 SIR 0.79 (0.52-1.16)Possibility of mis-classification of IBD, CD, UC, and Ca. Patients diagnosed 1987-1993 and 2000-2007 were only included
Kappelman et al[35], 2014DenmarkCohort (1978-2010)42717 IBD (35152 UC; 13756 CD); 5554844 controls. F.U. CD for 7.6 yr, UC for 7.8 yrIBD 316/42717; controls 33960/5554844. IBD SIR 1.21 (1.08-1.35); UC 258/35152 SIR 1.2 (1.1-1.4); CD 58/13756 SIR 1.2 (0.9-1.6)No age-estimates of absolute cancer risk. Detection bias. Data possibly missing. No inpatient encounters prior to 1995
Wilson et al[36], 2016SwitzerlandCase-control (1995-2012)19647 IBD (7850 CD; 11797 UC); 19647 controlsIBD 79/19647; 67/19647 controls. IBD aHR 1.19 (0.86-1.65); CD 17/7850; 16/7850 controls; CD aHR 1.08 (0.54-2.15); UC 62/11797; 51/11797 controls UC aHR 1.22 (0.84-1.77)Exposure misclassification. Potential bias in multivariate analysis (smoking, alcohol, BMI)
Jung et al[31], 2017KoreaCohort (2011-2014)9785 UC; 5506 CD; 50750000 controls19/15291 IBD; 20607/50750000 controls. IBD SIR 3.5 (2.1-5.5); UC SIR 3.47 (2.06-5.48); CD SIR O.99 (0.03-5.54)The study did not focus on IBD treatment. Data for disease diagnosis, phenotype not available. Short follow-up
So et al[29], 2017ChinaCohort (1990-2016)2621 IBD; 1603 UC; 7392000 controls. Median F.U. 8 yr CD, 10 yr UC8/2621 IBD; 11115/7392000 controls. IBD SIR 2.03 (1.03-4.06); 8/1603 UC; UC SIR 2.47 (1.24-4.95)The 25% of the cohort was followed up for < 5 yr. Small size of PC cases. Lead-time and detection bias. Exposure not evaluated
Mosher et al[24], 2018United StatesCase-control study Veteran population (1996-2015)2080 IBD patients; 271898 without IBD574/2080 IBD; 337/271898 IBD free; 20 yr RR 1.70 (1.28-2.27)Administrative data. Heterogeneity of the Ca types. Ca incidence rates may be underestimated
Burns et al[146], 2019United StatesCohort (1996-2017)1033 IBD; 9306 IBD freeIBD 30/1033; IBD free 29/9306, 10 yr HR 4.44 (2.98-6.62) P < 0.001; clinically significant PC: 10 yr HR 3.72 (2.15-6.42) P < 0.001; RR 9.32 (5.62-15.46)Variables for IBD missing. Academic medical centre. PC morbidity, mortality, IBD treatments and healthcare utilization not assessed
Meyers et al[147], 2020United StatesProspective Population-based United Kingdom Biobank cohort (2006 and 2010, with follow-up through mid-2015)2311 IBD (1488 UC; 643 CD); 215773 IBD free; Men aged 40 to 69 at study entryUC 49/1488; aHR 1.47 (1.11-1.95) P = 0.0070, ≤ 20 yr: 1.29 (0.89, 1.85), > 20 yr: 1.87 (1.21-2.91) P = 0.0052, BMI ≤ 30: 1.48 (1.07, 2.03), BM1 > 30: 1.35 (0.72, 2.51); CD 14/643 aHR 1.06 (0.63-1.80), ≤ 20 yr: 1.11 (0.58, 2.14), > 20 yr: 0.98 (0.41, 2.37), BMI ≤ 30: 0.83 (0.43, 1.59), BMI > 30: 2.25 (0.93, 5.41)Number of prior PSA test, DRE, PC morbidity and mortality and IBD treatments were not reported. No data for PC grade or stage. Selection bias

A meta-analysis of 9 studies involving 17052 IBD patients by Pedersen et al[10] in 2010 showed no association between IBD and PC [IBD: SIR 1.16 (0.88-1.52), UC: SIR 1.14 (0.85-1.42) CD: SIR 0.77 (0.41-1.45)]. These results were not confirmed by recent studies and on the other hand, an increased risk of PC in patients with UC was reported (Table 4). Actually, an increased risk worldwide only for UC patients was shown in another meta-analysis of nine population-based studies by Ge et al[164] in 2020, with a large sample and a long follow-up of patients [Cohort studies: IBD RR 1.33 (1.03-1.71); UC RR 1.58 (1.08-2.30); CD RR 1.12 (0.97-1.31); Case control studies: RR 1.81 (1.43-2.29)]. In addition, a recent meta-analysis of eight studies by Chen et al[165] in 2020 reported two interesting results. For the first time a higher risk was also found for CD patients [IBD RR 1.78 (1.32-2.41); UC RR 1.76 (1.06-2.91); CD RR 1.29 (1.04-1.61)] and a higher risk in Asian patients with IBD compared to Caucasians was shown [RR 3.02 (95%CI: 2.03-4.48) and 1.6 (95%CI: 1.17-2.19), respectively], in contrast with the known lower risk of PC in the Asian general population[165]. It is noteworthy that two other meta-analyses in 2020, by Carli et al[166] [IBD RR 1.71 (1.16-2.51) P = 0.007; UC RR 1.21 (0.98-1.51) P = 0.07; CD RR 1.10 (0.98-1.25) P = 0.12] and by Lo et al[37] [UC SIR 1.08 (0.9-1.3); CD SIR 1.04 (0.8-1.35)] did not demonstrate a higher risk of PC in the subgroup analysis of UC or CD.

In conclusion, data for PC risk in IBD patients are conflicting. Further prospective studies are needed to evaluate the impact of concomitant medications on the risk of developing PC and the possible utility of screening programs for early diagnosis of PC in patients with IBD.

SKIN MALIGNANCIES

Several population-based studies have shown an increased prevalence of skin malignancies among patients with IBD[10,167-169]. Furthermore, the incidence of skin cancer among those receiving immunosuppressants is rising over time[170]. On the contrary, other population-based studies showed that there is no per se increased risk of skin cancer in IBD patients[34,39,75]. The skin cancers include melanoma (MSC) and non-melanoma skin cancers (NMSC) with the latter further divided into squamous cell carcinoma (SCC) and basal cell carcinoma (BCC). It is well established that anti-TNF exposure is associated with the development of MSC[169], whereas NMSC is associated with thiopurines exposure (azathioprine, 6-mercaptopurine)[171]. With regard to the mechanism by which thiopurines may predispose patients to skin cancer the most prominent theory supports that thiopurines increase the vulnerability to UVA radiation. In detail, they cause the accumulation of 6-thioguanine in the DNA of patients and further UVA radiation causes production of reactive oxygen species that leads to DNA mutations and oxidative stress, resulting in oncogenesis[172]. On the other hand, MSC is an immunogenic aggressive tumor, in which suppression of the immune response allows these tumors to grow and metastasize[173]. For this reason, immunosuppressants may cause down-regulation of the tumor surveillance mechanisms, increased susceptibility to infection with oncogenic viruses (e.g., melanoma associated retroviruses), or direct pharmacologic effects of these medications on DNA metabolism[174].

There is evidence from several studies of an increased risk of NMSC in IBD associated with thiopurines use (Table 5). These findings were not confirmed by two other studies that found no increased NMSC risk among IBD thiopurine users[89,175]. A meta-analysis that pooled 60351 IBD patients to assess the risk of NMSC in those treated with thiopurines, showed that the risk was only modestly elevated (aHR 2.28; 95%CI: 1.50-3.45)[184]. There is also evidence of an increased risk of MSC in IBD and this risk is associated with anti-TNF use (Table 5). In a review and meta-analysis that included 172837 IBD patients, 179 MSC cases were reported (pooled crude IR 27.5/100000 person-years; 95%CI: 19.9-37.0) and a 37% increased risk of MSC, independent of biologic therapy[185]. On the contrary, a Danish Cochrane review and meta-analysis (56146 IBD patients 8.1% anti-TNF exposed) did not show increased risk of MSC in those treated with anti-TNF (HR 0.62; CI: 0.08-4.57)[94], which was further confirmed by a recent systematic review and meta-analysis (comprising 34029 biologic-treated and 135370 biologic-naive patients), where biologics did not significantly increase the risk of MSC[186]. In a recent study that included IBD patients with MSC, IBD extent was found to be a risk factor, both in UC (pancolitis OR: 3.09; 95%CI: 1.670-5.727) and CD (ileocolonic disease: OR: 1.98; 95%CI: 1.009-3.882), corticosteroids were risk factors in UC (OR: 1.41-3.72) whereas anti-TNFs were protective factors in UC (OR: 0.15-0.88) and CD (0.27-0.92), but this was attributed to in-situ melanoma only. Moreover, no association between survival of IBD-MSC patients and anti-TNF or immunosuppressants after MSC diagnosis was found[181].

Table 5 Studies on skin cancer in patients with inflammatory bowel disease.
Ref.
Type of study
Country
Patients
Follow up time
Results
Limitations
Armstrong et al[89], 2010Nested case controlUnited Kingdom16663 IBD patients; 392 developed Ca vs 1914 IBD controls6.4 yrNMSC with AZA use (OR 0.99, CI: 0.35-2.81)AZA users were included but not 6MP
Long et al[167], 2010Retrospective cohort; nested case controlUnited States53377 IBD patients vs 160037 non-IBD; 742 IBD NMSC cases vs 2968 IBD controls1.32 yrNMSC (IRR, 1.64; 95%CI: 1.51-1.78), NMSC recent TP use (OR, 3.56; 95%CI: 2.81-4.50), recent biologics in CD (OR, 2.07; 95%CI: 1.28-3.33), persistent TP use (OR, 4.27; 95%CI: 3.08-5.92), persistent biologic use in CD (OR, 2.18; 95%CI: 1.07-4.46)Patients aged < 64 yr, no exposure dose, short follow-up
Singh et al[168], 2011Retrospective cohort; case controlCanada9618 IBD patients vs 91378 non-IBD; 237 IBD NMSC cases vs 948 IBD controls11.7 yrBCC (HR, 1.20; 95%CI: 1.03-1.40). TP use SCC (HR, 5.40; 95%CI: 2.00-14.56) BCC (HR, 1.12; CI 0.68-1.85). Case-control: TP use SCC (OR, 20.52; 95%CI: 2.42-173.81), BCC (OR: 2.07; 95%CI: 1.10-3.87)Do not include use of IMMs before 1995
Peyrin-Biroulet et al[171], 2011Prospective observational cohort study (CESAME)France19486 IBD patients2.55 yrNMSC (SIR 2.89, 95%CI: 1.98-4.08) MSC (SIR 0.64, 95%CI: 0.17-1.63). NMSC: ongoing TP use (HR, 5.9; 95%CI: 2.1-16.4; P = 0.0006), past TP use (HR, 3.9; 95%CI: 1.3-12.1; P = 0.02), age per 1-yr increase (HR, 1.08; 95%CI: 1.05-1.11; P < 0.0001)Younger patients
van Schaik et al[175], 2011Retrospective cohortHolland2887 IBD patients6.46 yrNMSC AZA use (HR 0.85, 95%CI: 0.51-1.41)Small study sample size
Long et al[169], 2012Retrospective cohort; nested case-controlUnited States108579 IBD vs 434 233 non-IBD controls; 209 MSC cases vs 823 IBD non-MSC controls, 3288 NMSC cases vs 12945 IBD non-NMSC controls2 yrMSC (HR, 1.15; 95%CI: 0.97-1.36) NMSC (HR, 1.34; 95%CI: 1.28-1.40). MSC anti-TNF (OR, 1.88; 95%CI: 1.08-3.29), long-term vs non-long-term use (OR 3.93, 95%CI: 1.82-8.50), no association with TP or 5-ASA. NMSC any TP use (OR, 1.85; 95%CI: 1.66-2.05), anti-TNF (OR, 1.14; 95%CI: 0.95-1.36), combination treatment (OR, 3.89; 95%CI: 2.33-6.46)Study population aged < 64 yr, no dose information about treatments, short mean follow-up
Peyrin-Biroulet et al[176], 2012Prospective observational cohort study (CESAME)France19486 IBD patients2.55 yrMSC previously TP treated (SIR: 0; 95%CI: 0-3.11), current TP users (SIR: 1.09; 95%CI: 0.13-3.94)Younger patient population
Abbas et al[177], 2014Retrospective cohort; nested case controlUnited States14527 patients; 421 NMSC and 45 MSC cases8.1 yrNMSC current AZA use (HR 2.1, 95%CI: 1.6-2.6), previous AZA use (HR 0.7, 95%CI: 0.5-1.0). MSC current AZA use (HR 1.5, 95%CI: 0.6-3.4), previous AZA use (HR 0.5, 95%CI: 90.1-1.8)Patient population limited to VA health care system (older, white, male)
McKenna et al[178], 2014Database inquiry (AE- (FAERS)United States315 skin CaNAPRR, increased odds of MSC and NMSC for anti-TNF (P = 0.035 and 0.03, respectively) and combination treatment (P < 0.001 and P < 0.001)AE database (reporting bias) skewed towards CD
Kopylov et al[179], 2015Nested case controlCanada19582 patients; (MSC 102 vs IBD Controls 1014) (NMSC 474 IBD vs Controls 4684)No reported meanNMSC: TP treatment ≥ 3 yr (OR 1.41; 95%CI: 1.11-1.79), TP treatment ≥ 5 yr (OR: 2.07; 95%CI: 1.36-3.7), combination treatment (OR: 3.11; 95%CI: 1.33-7.27). After stopping TP, OR: 1.04 (0.69-1.55). IMMs-anti-TNF were not associated with MSCYounger, employed patients are underrepresented, not mentioned disease severity
Scott et al[180], 2016Retrospective cohortUnited States2788 IBD patients2.24 yrSecond NMSC with short-term TP treatment (HR 1.53, 95%CI: 0.87-2.70), with > 1 yr of TP therapy (HR 1.49, 95%CI: 0.98-2.27)Older patient population
Nissen et al[181], 20172 Retrospective case-control studiesThe Netherlands304 IBD patients with MSC, 1800 IBD controls, 8177 MSC non-IBD controlsMSC: UC (pancolitis OR 3.09; 95%CI: 1.670-5.727), CD (ileocolonic disease: OR 1.98; 95%CI: 1.009-3.882). Corticosteroids (OR 1.41-3.72), anti-TNF UC (OR 0.15-0.88), CD (0.27-0.92). (only attributed to the in situ MSC). Survival with anti-TNF (HR 0.32; 95%CI: 0.08-1.27) and TP (HR 0.72; 95%CI: 0.37-1.31). Survival after MSC diagnosis anti-TNF (HR 0.16, 95%CI: 0.02-1.21) and TP (HR 0.55, 95%CI: 0.25-1.23)Medication of patients after 1990 was included. Not informed about skin type, number of sun burns
Clowry et al[182], 2017Retrospective cohortIreland2053 patients with IBD9.8 yrNMSC under IMMs SIR 1.8 (95%CI: 1.0-2.7), TP exposure (OR: 5.26, 95%CI: 2.15-12.93, P < 0.001), TP and/or anti-TNF (OR: 6.45, 95%CI: 2.69-15.95, P <0.001)Small sample size, hospital database mostly severe IBD
Khan et al[183], 2020Retrospective cohortUnited States54919 patients with IBD; VAHS 518 patients with BCC5.71 yrRepeated BCC occurrences, compared with 5-ASA, under active TP use (HR 1.65, 95%CI: 1.24-2.19, P = 0.0005), 6 mo after TP discontinuation (HR 1.22, 95%CI: 0.86-1.74, P = 0.26), for anti-TNF use (HR 1.27, 95%CI: 0.84-1.90, P = 0.26), for combination treatment (HR 1.37, 95%CI: 0.90-2.08, P = 0.14)Study population mostly males. Prescriptions outside VAHS not included

The known risk factors for the development of skin cancers include smoking, older age, male sex, fair skin type, cumulative sun exposure (mostly for NMSC), sun-burn (mostly for MSC), family history of skin cancer, Caucasian race, geographical area and certain genetic factors (e.g., p53 polymorphisms). All these factors should be taken into account when immunosuppressant therapy is being considered for a patient with IBD. Given these findings, drugs other than anti-TNFs might be safer for use in patients after a melanoma diagnosis or in those at high risk for these tumors, whereas thiopurines should also be avoided in patients with a history of SCC, multiple BCCs, or premalignant skin lesions (e.g., solar keratosis)[187].

With regard to the maintenance of immunosuppressive treatment after the initial diagnosis of skin cancer, the results from various studies have been controversial. A large cohort study showed that thiopurines did not increase the risk of second NMSC when used in combination treatment compared with anti-TNF monotherapy (HR 0.79, 95%CI: 0.30-2.08)[180]. In addition, a previous study by Beaugerie concluded that exposure to immunosuppressants did not increase the risk of new or recurrent malignancy in individuals with previous cancer[188]. On the contrary, another recent cohort study with 54919 IBD patients, of whom 518 developed BCC and maintained their treatment, it was shown that repeated BCC occurrences were associated with active thiopurine use[183]. In line with this, a large meta-analysis that included 3706 IBD patients with 10332 person-years of follow-up after a cancer diagnosis showed, in a subgroup on skin cancer alone, that the risk was significantly higher in immunosuppressant-users (71.6 per 1000 person-year, 95%CI: 58.9-84.2, P = 0.035) compared to those not receiving immunosuppressants (50.8 per 1000 person-year, 95%CI: 43.7-57.8) and numerically higher than anti-TNFα therapy (55.5 per 1000 person-year, 95%CI: 44.7-66.3, P = 0.22)[189].

With regard to prevention, a recent study showed that only a small proportion of IBD patients (8.3% during the 7-year study period) were seeking dermatologic care[190].

In conclusion, it seems that there is an increased risk of NMSC and a slightly lower risk of MSC in IBD patients, which justifies the recommendations for routine screening for skin malignancies. Although there is some evidence of second NMSC after an index diagnosis with azathioprine, overall data are conflicting regarding maintaining immunosuppressive treatment after the diagnosis of a skin malignancy and the risk of recurrence or a second skin malignancy, so the decision should be individualized. Conclusively, primary prevention measures, such as sun-protection, annual dermatologic examination and smoking abstinence should be encouraged in all IBD patients, regardless of thiopurine and/or anti-TNFα use.

CONCLUSION

Cancer is the second cause of death in patients with IBD, after cardiovascular disease. Lately there has been evidence that the incidence of intestinal cancers is decreasing over time in IBD patients, as a result of the implementation of a tight screening strategy. Although recent data supporting that there is no excess overall risk of extraintestinal cancers, it seems that IBD patients are in higher risk of certain extraintestinal cancers. This indicates the need for close monitoring, as it seems that chronic systemic inflammation but also other factors such as older age, smoking, specific virus infections (e.g., HPV) and concurrent diseases (PSC) play a role in carcinogenesis. The IBD treatments, for both CD and UC, are aimed at clinical improvement, endoscopic remission and sustained mucosal healing. Although immunosuppressants, including azathioprine, methotrexate and anti-TNFs, constitute the cornerstone of IBD treatment, these drugs might have a carcinogenic effect either by directly altering cellular DNA or reducing tumor immunosurveillance. Lately there are more treatment choices available, such as anti-integrins, anti-IL-12/23 and JAK inhibitors. Although these new treatments are considered to have a better safety profile as far as cancer is concerned, data are limited and future studies are required.

Specialists who deal with the management of IBD patients must bear in mind the possibility of extraintestinal malignancies, especially in those with long standing immunosuppressive therapies and persistent systemic or local inflammation. Guidelines and recommendations should incorporate all the preventative measures [sun protection, dermatologic examination, vaccinations (HPV, hepatitis B virus), annual Pap smear, mammography, PSA testing, annual MRCP where appropriate] and applied in everyday clinical practice to diminish the risk of IBD-related extraintestinal malignancies, achieve an early diagnosis and improve the prognosis and overall outcome.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Specialty type: Oncology

Country/Territory of origin: Greece

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Sassaki LY S-Editor: Gao CC L-Editor: Webster JR P-Editor: Yu HG

References
1.  de Souza HS, Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nat Rev Gastroenterol Hepatol. 2016;13:13-27.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Nieminen U, Färkkilä M. Malignancies in inflammatory bowel disease. Scand J Gastroenterol. 2015;50:81-89.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Lutgens MW, van Oijen MG, van der Heijden GJ, Vleggaar FP, Siersema PD, Oldenburg B. Declining risk of colorectal cancer in inflammatory bowel disease: an updated meta-analysis of population-based cohort studies. Inflamm Bowel Dis. 2013;19:789-799.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Jess T, Simonsen J, Jørgensen KT, Pedersen BV, Nielsen NM, Frisch M. Decreasing risk of colorectal cancer in patients with inflammatory bowel disease over 30 years. Gastroenterology. 2012;143:375-81.e1; quiz e13.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Katsanos KH, Tatsioni A, Pedersen N, Shuhaibar M, Ramirez VH, Politi P, Rombrechts E, Pierik M, Clofent J, Beltrami M, Bodini P, Freitas J, Mouzas I, Fornaciari G, Moum B, Lakatos PL, Vermeire S, Langholz E, Odes S, Morain CO, Stockbrügger R, Munkholm P, Tsianos EV. Cancer in inflammatory bowel disease 15 years after diagnosis in a population-based European Collaborative follow-up study. J Crohns Colitis. 2011;5:430-442.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Algaba A, Guerra I, Castaño A, de la Poza G, Castellano VM, López M, Bermejo F. Risk of cancer, with special reference to extra-intestinal malignancies, in patients with inflammatory bowel disease. World J Gastroenterol. 2013;19:9359-9365.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Scharl S, Barthel C, Rossel JB, Biedermann L, Misselwitz B, Schoepfer AM, Straumann A, Vavricka SR, Rogler G, Scharl M, Greuter T. Malignancies in Inflammatory Bowel Disease: Frequency, Incidence and Risk Factors-Results from the Swiss IBD Cohort Study. Am J Gastroenterol. 2019;114:116-126.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Loo SY, Vutcovici M, Bitton A, Lakatos PL, Azoulay L, Suissa S, Brassard P. Risk of Malignant Cancers in Inflammatory Bowel Disease. J Crohns Colitis. 2019;13:1302-1310.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Taborelli M, Sozzi M, Del Zotto S, Toffolutti F, Montico M, Zanier L, Serraino D. Risk of intestinal and extra-intestinal cancers in patients with inflammatory bowel diseases: A population-based cohort study in northeastern Italy. PLoS One. 2020;15:e0235142.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Pedersen N, Duricova D, Elkjaer M, Gamborg M, Munkholm P, Jess T. Risk of extra-intestinal cancer in inflammatory bowel disease: meta-analysis of population-based cohort studies. Am J Gastroenterol. 2010;105:1480-1487.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  van den Heuvel TR, Wintjens DS, Jeuring SF, Wassink MH, Romberg-Camps MJ, Oostenbrug LE, Sanduleanu S, Hameeteman WH, Zeegers MP, Masclee AA, Jonkers DM, Pierik MJ. Inflammatory bowel disease, cancer and medication: Cancer risk in the Dutch population-based IBDSL cohort. Int J Cancer. 2016;139:1270-1280.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Aupérin A. Epidemiology of head and neck cancers: an update. Curr Opin Oncol. 2020;32:178-186.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Hashibe M, Brennan P, Chuang SC, Boccia S, Castellsague X, Chen C, Curado MP, Dal Maso L, Daudt AW, Fabianova E, Fernandez L, Wünsch-Filho V, Franceschi S, Hayes RB, Herrero R, Kelsey K, Koifman S, La Vecchia C, Lazarus P, Levi F, Lence JJ, Mates D, Matos E, Menezes A, McClean MD, Muscat J, Eluf-Neto J, Olshan AF, Purdue M, Rudnai P, Schwartz SM, Smith E, Sturgis EM, Szeszenia-Dabrowska N, Talamini R, Wei Q, Winn DM, Shangina O, Pilarska A, Zhang ZF, Ferro G, Berthiller J, Boffetta P. Interaction between tobacco and alcohol use and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Cancer Epidemiol Biomarkers Prev. 2009;18:541-550.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Whiteman DC, Wilson LF. The fractions of cancer attributable to modifiable factors: A global review. Cancer Epidemiol. 2016;44:203-221.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  de Martel C, Plummer M, Vignat J, Franceschi S. Worldwide burden of cancer attributable to HPV by site, country and HPV type. Int J Cancer. 2017;141:664-670.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Sabatini ME, Chiocca S. Human papillomavirus as a driver of head and neck cancers. Br J Cancer. 2020;122:306-314.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Ndiaye C, Mena M, Alemany L, Arbyn M, Castellsagué X, Laporte L, Bosch FX, de Sanjosé S, Trottier H. HPV DNA, E6/E7 mRNA, and p16INK4a detection in head and neck cancers: a systematic review and meta-analysis. Lancet Oncol. 2014;15:1319-1331.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Katsanos KH, Roda G, McBride RB, Cohen B, Colombel JF. Increased Risk of Oral Cancer in Patients With Inflammatory Bowel Diseases. Clin Gastroenterol Hepatol. 2016;14:413-420.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Nissen LHC, Derikx LAAP, Jacobs AME, van Herpen CM, Kievit W, Verhoeven R, van den Broek E, Bekers E, van den Heuvel T, Pierik M, Rahamat-Langendoen J, Takes RP, Melchers WJG, Nagtegaal ID, Hoentjen F; Dutch Initiative on Crohn and Colitis (ICC);  Dutch Head and Neck Society, PALGA group;  IBD/HNC group. Risk Factors and Clinical Outcomes of Head and Neck Cancer in Inflammatory Bowel Disease: A Nationwide Cohort Study. Inflamm Bowel Dis. 2018;24:2015-2026.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  van de Ven SEM, Derikx LAAP, Nagtegaal ID, van Herpen CM, Takes RP, Melchers WJG, Pierik M, van den Heuvel T, Verhoeven RHA, Hoentjen F, Nissen LHC. Laryngeal Carcinoma in Patients With Inflammatory Bowel Disease: Clinical Outcomes and Risk Factors. Inflamm Bowel Dis. 2020;26:1060-1067.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Giagkou E, Christodoulou DK, Katsanos KH. Mouth cancer in inflammatory bowel diseases. Oral Dis. 2016;22:260-264.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Pasternak B, Svanström H, Schmiegelow K, Jess T, Hviid A. Use of azathioprine and the risk of cancer in inflammatory bowel disease. Am J Epidemiol. 2013;177:1296-1305.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Nyboe Andersen N, Pasternak B, Friis-Møller N, Andersson M, Jess T. Association between tumour necrosis factor-α inhibitors and risk of serious infections in people with inflammatory bowel disease: nationwide Danish cohort study. BMJ. 2015;350:h2809.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Mosher CA, Brown GR, Weideman RA, Crook TW, Cipher DJ, Spechler SJ, Feagins LA. Incidence of Colorectal Cancer and Extracolonic Cancers in Veteran Patients With Inflammatory Bowel Disease. Inflamm Bowel Dis. 2018;24:617-623.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  National Cancer Institute  SEER cancer stat facts: thyroid cancer. [cited 21 August 2018]. In: National Cancer Institute [Internet]. Available from: https://seer.cancer.gov/statfacts/html/thyro.html.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Biancone L, Armuzzi A, Scribano ML, Castiglione F, D'Incà R, Orlando A, Papi C, Daperno M, Vecchi M, Riegler G, Fries W, Alvisi P, Meucci G, Mocciaro F, Rogai F, Festa S, Guidi L, Testa A, Spina L, Renna S, Viola A, Patturelli M, Di Mitri R, Frankovic I, Calabrese E, Petruzziello C, De Cristofaro E, Sena G, Ruffa A, Neri B, Rossi A. Cancer Risk in Inflammatory Bowel Disease: A 6-Year Prospective Multicenter Nested Case-Control IG-IBD Study. Inflamm Bowel Dis. 2020;26:450-459.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Yano Y, Matsui T, Hirai F, Okado Y, Sato Y, Tsurumi K, Ishikawa S, Beppu T, Koga A, Yoshizawa N, Higashi D, Futami K. Cancer risk in Japanese Crohn's disease patients: investigation of the standardized incidence ratio. J Gastroenterol Hepatol. 2013;28:1300-1305.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Sonu IS, Blonski W, Lin MV, Lewis J, Aberra F, Lichtenstein GR. Papillary thyroid cancer and inflammatory bowel disease: is there a relationship? World J Gastroenterol. 2013;19:1079-1084.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  So J, Tang W, Leung WK, Li M, Lo FH, Wong MTL, Sze ASF, Leung CM, Tsang SWC, Shan EHS, Chan KH, Lam BCY, Hui AJ, Chow WH, Lam TY, Lam V, Lee TW, Lo HHH, Tang CM, Wong CL, Wu JCY, Chan FKL, Sung JJY, Harbord M, Ng SC. Cancer Risk in 2621 Chinese Patients with Inflammatory Bowel Disease: A Population-based Cohort Study. Inflamm Bowel Dis. 2017;23:2061-2068.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Jussila A, Virta LJ, Pukkala E, Färkkilä MA. Malignancies in patients with inflammatory bowel disease: a nationwide register study in Finland. Scand J Gastroenterol. 2013;48:1405-1413.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Jung YS, Han M, Park S, Kim WH, Cheon JH. Cancer Risk in the Early Stages of Inflammatory Bowel Disease in Korean Patients: A Nationwide Population-based Study. J Crohns Colitis. 2017;11:954-962.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Cao L. Assessment of thyroid cancer risk in more than 334,000 patients with inflammatory bowel disease: a case-control study and a meta-analysis. World J Surg Oncol. 2018;16:182.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Wadhwa V, Lopez R, Shen B. Crohn's Disease Is Associated with the Risk for Thyroid Cancer. Inflamm Bowel Dis. 2016;22:2902-2906.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Jess T, Winther KV, Munkholm P, Langholz E, Binder V. Intestinal and extra-intestinal cancer in Crohn's disease: follow-up of a population-based cohort in Copenhagen County, Denmark. Aliment Pharmacol Ther. 2004;19:287-293.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Kappelman MD, Farkas DK, Long MD, Erichsen R, Sandler RS, Sørensen HT, Baron JA. Risk of cancer in patients with inflammatory bowel diseases: a nationwide population-based cohort study with 30 years of follow-up evaluation. Clin Gastroenterol Hepatol. 2014;12:265-73.e1.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Wilson JC, Furlano RI, Jick SS, Meier CR. A population-based study examining the risk of malignancy in patients diagnosed with inflammatory bowel disease. J Gastroenterol. 2016;51:1050-1062.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Lo B, Zhao M, Vind I, Burisch J. The Risk of Extraintestinal Cancer in Inflammatory Bowel Disease: A Systematic Review and Meta-analysis of Population-based Cohort Studies. Clin Gastroenterol Hepatol. 2021;19:1117-1138.e19.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Masala G, Bagnoli S, Ceroti M, Saieva C, Trallori G, Zanna I, D'Albasio G, Palli D. Divergent patterns of total and cancer mortality in ulcerative colitis and Crohn's disease patients: the Florence IBD study 1978-2001. Gut. 2004;53:1309-1313.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Bernstein CN, Blanchard JF, Kliewer E, Wajda A. Cancer risk in patients with inflammatory bowel disease: a population-based study. Cancer. 2001;91:854-862.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Karlén P, Löfberg R, Broström O, Leijonmarck CE, Hellers G, Persson PG. Increased risk of cancer in ulcerative colitis: a population-based cohort study. Am J Gastroenterol. 1999;94:1047-1052.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Chapman RW, Williamson KD. Are Dominant Strictures in Primary Sclerosing Cholangitis a Risk Factor for Cholangiocarcinoma? Curr Hepatol Rep. 2017;16:124-129.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Bonato G, Cristoferi L, Strazzabosco M, Fabris L. Malignancies in Primary Sclerosing Cholangitis--A Continuing Threat. Dig Dis. 2015;33 Suppl 2:140-148.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Ananthakrishnan AN, Cagan A, Gainer VS, Cheng SC, Cai T, Szolovits P, Shaw SY, Churchill S, Karlson EW, Murphy SN, Kohane I, Liao KP. Mortality and extraintestinal cancers in patients with primary sclerosing cholangitis and inflammatory bowel disease. J Crohns Colitis. 2014;8:956-963.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Sørensen JØ, Nielsen OH, Andersson M, Ainsworth MA, Ytting H, Bélard E, Jess T. Inflammatory bowel disease with primary sclerosing cholangitis: A Danish population-based cohort study 1977-2011. Liver Int. 2018;38:532-541.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Fevery J, Verslype C, Lai G, Aerts R, Van Steenbergen W. Incidence, diagnosis, and therapy of cholangiocarcinoma in patients with primary sclerosing cholangitis. Dig Dis Sci. 2007;52:3123-3135.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Bergquist A, Ekbom A, Olsson R, Kornfeldt D, Lööf L, Danielsson A, Hultcrantz R, Lindgren S, Prytz H, Sandberg-Gertzén H, Almer S, Granath F, Broomé U. Hepatic and extrahepatic malignancies in primary sclerosing cholangitis. J Hepatol. 2002;36:321-327.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Singh S, Talwalkar JA. Primary sclerosing cholangitis: diagnosis, prognosis, and management. Clin Gastroenterol Hepatol. 2013;11:898-907.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Boberg KM, Bergquist A, Mitchell S, Pares A, Rosina F, Broomé U, Chapman R, Fausa O, Egeland T, Rocca G, Schrumpf E. Cholangiocarcinoma in primary sclerosing cholangitis: risk factors and clinical presentation. Scand J Gastroenterol. 2002;37:1205-1211.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Huai JP, Ding J, Ye XH, Chen YP. Inflammatory bowel disease and risk of cholangiocarcinoma: evidence from a meta-analysis of population-based studies. Asian Pac J Cancer Prev. 2014;15:3477-3482.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Gulamhusein AF, Eaton JE, Tabibian JH, Atkinson EJ, Juran BD, Lazaridis KN. Duration of Inflammatory Bowel Disease Is Associated With Increased Risk of Cholangiocarcinoma in Patients With Primary Sclerosing Cholangitis and IBD. Am J Gastroenterol. 2016;111:705-711.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Burak K, Angulo P, Pasha TM, Egan K, Petz J, Lindor KD. Incidence and risk factors for cholangiocarcinoma in primary sclerosing cholangitis. Am J Gastroenterol. 2004;99:523-526.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Broomé U, Löfberg R, Veress B, Eriksson LS. Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology. 1995;22:1404-1408.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  European Association for the Study of the Liver. EASL Clinical Practice Guidelines: management of cholestatic liver diseases. J Hepatol. 2009;51:237-267.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Chapman MH, Webster GJ, Bannoo S, Johnson GJ, Wittmann J, Pereira SP. Cholangiocarcinoma and dominant strictures in patients with primary sclerosing cholangitis: a 25-year single-centre experience. Eur J Gastroenterol Hepatol. 2012;24:1051-1058.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Gotthardt DN, Rudolph G, Klöters-Plachky P, Kulaksiz H, Stiehl A. Endoscopic dilation of dominant stenoses in primary sclerosing cholangitis: outcome after long-term treatment. Gastrointest Endosc. 2010;71:527-534.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Rudolph G, Gotthardt D, Klöters-Plachky P, Kulaksiz H, Rost D, Stiehl A. Influence of dominant bile duct stenoses and biliary infections on outcome in primary sclerosing cholangitis. J Hepatol. 2009;51:149-155.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Rudolph G, Gotthardt D, Kloeters-Plachky P, Rost D, Kulaksiz H, Stiehl A. In PSC with dominant bile duct stenosis, IBD is associated with an increase of carcinomas and reduced survival. J Hepatol. 2010;53:313-317.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Janse M, Lamberts LE, Verdonk RC, Weersma RK. IBD is associated with an increase in carcinoma in PSC irrespective of the presence of dominant bile duct stenosis. J Hepatol. 2012;57:473-4; author reply 475.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Björnsson E, Olsson R, Bergquist A, Lindgren S, Braden B, Chapman RW, Boberg KM, Angulo P. The natural history of small-duct primary sclerosing cholangitis. Gastroenterology. 2008;134:975-980.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Fevery J, Henckaerts L, Van Oirbeek R, Vermeire S, Rutgeerts P, Nevens F, Van Steenbergen W. Malignancies and mortality in 200 patients with primary sclerosering cholangitis: a long-term single-centre study. Liver Int. 2012;32:214-222.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Ishida M, Naka S, Shiomi H, Tsujikawa T, Andoh A, Nakahara T, Saito Y, Kurumi Y, Takikita-Suzuki M, Kojima F, Hotta M, Tani T, Fujiyama Y, Okabe H. Hepatocellular carcinoma occurring in a Crohn's disease patient. World J Gastroenterol. 2010;16:3215-3218.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Trivedi PJ, Crothers H, Mytton J, Bosch S, Iqbal T, Ferguson J, Hirschfield GM. Effects of Primary Sclerosing Cholangitis on Risks of Cancer and Death in People With Inflammatory Bowel Disease, Based on Sex, Race, and Age. Gastroenterology. 2020;159:915-928.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Brandt DJ, MacCarty RL, Charboneau JW, LaRusso NF, Wiesner RH, Ludwig J. Gallbladder disease in patients with primary sclerosing cholangitis. AJR Am J Roentgenol. 1988;150:571-574.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Ahrendt SA, Rashid A, Chow JT, Eisenberger CF, Pitt HA, Sidransky D. p53 overexpression and K-ras gene mutations in primary sclerosing cholangitis-associated biliary tract cancer. J Hepatobiliary Pancreat Surg. 2000;7:426-431.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Bergquist A, Glaumann H, Persson B, Broomé U. Risk factors and clinical presentation of hepatobiliary carcinoma in patients with primary sclerosing cholangitis: a case-control study. Hepatology. 1998;27:311-316.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Brandsaeter B, Isoniemi H, Broomé U, Olausson M, Bäckman L, Hansen B, Schrumpf E, Oksanen A, Ericzon BG, Höckerstedt K, Mäkisalo H, Kirkegaard P, Friman S, Bjøro K. Liver transplantation for primary sclerosing cholangitis; predictors and consequences of hepatobiliary malignancy. J Hepatol. 2004;40:815-822.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Buckles DC, Lindor KD, Larusso NF, Petrovic LM, Gores GJ. In primary sclerosing cholangitis, gallbladder polyps are frequently malignant. Am J Gastroenterol. 2002;97:1138-1142.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Campbell WL, Ferris JV, Holbert BL, Thaete FL, Baron RL. Biliary tract carcinoma complicating primary sclerosing cholangitis: evaluation with CT, cholangiography, US, and MR imaging. Radiology. 1998;207:41-50.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Campbell WL, Peterson MS, Federle MP, Siqueira ES, Slivka A, Grazioli L, Ichikawa T, Oliver JH 3rd, Kim T, Li W. Using CT and cholangiography to diagnose biliary tract carcinoma complicating primary sclerosing cholangitis. AJR Am J Roentgenol. 2001;177:1095-1100.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Dorudi S, Chapman RW, Kettlewell MG. Carcinoma of the gallbladder in ulcerative colitis and primary sclerosing cholangitis. Report of two cases. Dis Colon Rectum. 1991;34:827-828.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Lewis JT, Talwalkar JA, Rosen CB, Smyrk TC, Abraham SC. Prevalence and risk factors for gallbladder neoplasia in patients with primary sclerosing cholangitis: evidence for a metaplasia-dysplasia-carcinoma sequence. Am J Surg Pathol. 2007;31:907-913.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Everhov ÅH, Erichsen R, Sachs MC, Pedersen L, Halfvarson J, Askling J, Ekbom A, Ludvigsson JF, Sørensen HT, Olén O. Inflammatory bowel disease and pancreatic cancer: a Scandinavian register-based cohort study 1969-2017. Aliment Pharmacol Ther. 2020;52:143-154.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Correa P. Human gastric carcinogenesis: a multistep and multifactorial process--First American Cancer Society Award Lecture on Cancer Epidemiology and Prevention. Cancer Res. 1992;52:6735-6740.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Papamichael K, Konstantopoulos P, Mantzaris GJ. Helicobacter pylori infection and inflammatory bowel disease: is there a link? World J Gastroenterol. 2014;20:6374-6385.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Jess T, Horváth-Puhó E, Fallingborg J, Rasmussen HH, Jacobsen BA. Cancer risk in inflammatory bowel disease according to patient phenotype and treatment: a Danish population-based cohort study. Am J Gastroenterol. 2013;108:1869-1876.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Nissen LH, Assendorp EL, van der Post RS, Derikx LA, de Jong DJ, Kievit W, Pierik M, van den Heuvel T, Verhoeven R, Overbeek LI, Hoentjen F, Nagtegaal ID. Impaired Gastric Cancer Survival in Patients with Inflammatory Bowel Disease. J Gastrointestin Liver Dis. 2016;25:431-440.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87-108.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Lichtarowicz A, Norman C, Calcraft B, Morris JS, Rhodes J, Mayberry J. A study of the menopause, smoking, and contraception in women with Crohn's disease. Q J Med. 1989;72:623-631.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Bharadwaj S, Kulkarni G, Shen B. Menstrual cycle, sex hormones in female inflammatory bowel disease patients with and without surgery. J Dig Dis. 2015;16:245-255.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Ekbom A, Helmick C, Zack M, Adami HO. Extracolonic malignancies in inflammatory bowel disease. Cancer. 1991;67:2015-2019.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Mellemkjaer L, Olsen JH, Frisch M, Johansen C, Gridley G, McLaughlin JK. Cancer in patients with ulcerative colitis. Int J Cancer. 1995;60:330-333.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Mellemkjaer L, Johansen C, Gridley G, Linet MS, Kjaer SK, Olsen JH. Crohn's disease and cancer risk (Denmark). Cancer Causes Control. 2000;11:145-150.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Hemminki K, Li X, Sundquist J, Sundquist K. Cancer risks in Crohn disease patients. Ann Oncol. 2009;20:574-580.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Hemminki K, Liu X, Ji J, Försti A, Sundquist J, Sundquist K. Effect of autoimmune diseases on risk and survival in female cancers. Gynecol Oncol. 2012;127:180-185.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Algaba A, Guerra I, Marín-Jiménez I, Quintanilla E, López-Serrano P, García-Sánchez MC, Casis B, Taxonera C, Moral I, Chaparro M, Martín-Rodríguez D, Martín-Arranz MD, Manceñido N, Menchén L, López-Sanromán A, Castaño Á, Bermejo F. Incidence, management, and course of cancer in patients with inflammatory bowel disease. J Crohns Colitis. 2015;9:326-333.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Lichtenstein GR, Feagan BG, Cohen RD, Salzberg BA, Diamond RH, Langholff W, Londhe A, Sandborn WJ. Drug therapies and the risk of malignancy in Crohn's disease: results from the TREAT™ Registry. Am J Gastroenterol. 2014;109:212-223.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Tsai MS, Chen HP, Hung CM, Lee PH, Lin CL, Kao CH. Hospitalization for Inflammatory Bowel Disease is Associated with Increased Risk of Breast Cancer: A Nationwide Cohort Study of an Asian Population. Ann Surg Oncol. 2015;22:1996-2002.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Hovde Ø, Høivik ML, Henriksen M, Solberg IC, Småstuen MC, Moum BA. Malignancies in Patients with Inflammatory Bowel Disease: Results from 20 Years of Follow-up in the IBSEN Study. J Crohns Colitis. 2017;11:571-577.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Armstrong RG, West J, Card TR. Risk of cancer in inflammatory bowel disease treated with azathioprine: a UK population-based case-control study. Am J Gastroenterol. 2010;105:1604-1609.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Fraser AG, Orchard TR, Robinson EM, Jewell DP. Long-term risk of malignancy after treatment of inflammatory bowel disease with azathioprine. Aliment Pharmacol Ther. 2002;16:1225-1232.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Connell WR, Kamm MA, Dickson M, Balkwill AM, Ritchie JK, Lennard-Jones JE. Long-term neoplasia risk after azathioprine treatment in inflammatory bowel disease. Lancet. 1994;343:1249-1252.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Setshedi M, Epstein D, Winter TA, Myer L, Watermeyer G, Hift R. Use of thiopurines in the treatment of inflammatory bowel disease is associated with an increased risk of non-melanoma skin cancer in an at-risk population: a cohort study. J Gastroenterol Hepatol. 2012;27:385-389.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Gómez-García M, Cabello-Tapia MJ, Sánchez-Capilla AD, De Teresa-Galván J, Redondo-Cerezo E. Thiopurines related malignancies in inflammatory bowel disease: local experience in Granada, Spain. World J Gastroenterol. 2013;19:4877-4886.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Nyboe Andersen N, Pasternak B, Basit S, Andersson M, Svanström H, Caspersen S, Munkholm P, Hviid A, Jess T. Association between tumor necrosis factor-α antagonists and risk of cancer in patients with inflammatory bowel disease. JAMA. 2014;311:2406-2413.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Colombel JF, Sands BE, Rutgeerts P, Sandborn W, Danese S, D'Haens G, Panaccione R, Loftus EV Jr, Sankoh S, Fox I, Parikh A, Milch C, Abhyankar B, Feagan BG. The safety of vedolizumab for ulcerative colitis and Crohn's disease. Gut. 2017;66:839-851.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Osterman MT, Sandborn WJ, Colombel JF, Robinson AM, Lau W, Huang B, Pollack PF, Thakkar RB, Lewis JD. Increased risk of malignancy with adalimumab combination therapy, compared with monotherapy, for Crohn's disease. Gastroenterology. 2014;146:941-949.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Beigel F, Steinborn A, Schnitzler F, Tillack C, Breiteneicher S, John JM, Van Steen K, Laubender RP, Göke B, Seiderer J, Brand S, Ochsenkühn T. Risk of malignancies in patients with inflammatory bowel disease treated with thiopurines or anti-TNF alpha antibodies. Pharmacoepidemiol Drug Saf. 2014;23:735-744.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Fidder H, Schnitzler F, Ferrante M, Noman M, Katsanos K, Segaert S, Henckaerts L, Van Assche G, Vermeire S, Rutgeerts P. Long-term safety of infliximab for the treatment of inflammatory bowel disease: a single-centre cohort study. Gut. 2009;58:501-508.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Lichtenstein GR, Rutgeerts P, Sandborn WJ, Sands BE, Diamond RH, Blank M, Montello J, Tang L, Cornillie F, Colombel JF. A pooled analysis of infections, malignancy, and mortality in infliximab- and immunomodulator-treated adult patients with inflammatory bowel disease. Am J Gastroenterol. 2012;107:1051-1063.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Williams CJ, Peyrin-Biroulet L, Ford AC. Systematic review with meta-analysis: malignancies with anti-tumour necrosis factor-α therapy in inflammatory bowel disease. Aliment Pharmacol Ther. 2014;39:447-458.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Waggoner SE. Cervical cancer. Lancet. 2003;361:2217-2225.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Burd EM. Human papillomavirus and cervical cancer. Clin Microbiol Rev. 2003;16:1-17.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Wheeler CM. Natural history of human papillomavirus infections, cytologic and histologic abnormalities, and cancer. Obstet Gynecol Clin North Am. 2008;35:519-36; vii.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Dalstein V, Riethmuller D, Prétet JL, Le Bail Carval K, Sautière JL, Carbillet JP, Kantelip B, Schaal JP, Mougin C. Persistence and load of high-risk HPV are predictors for development of high-grade cervical lesions: a longitudinal French cohort study. Int J Cancer. 2003;106:396-403.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  US Preventive Services Task Force, Curry SJ, Krist AH, Owens DK, Barry MJ, Caughey AB, Davidson KW, Doubeni CA, Epling JW Jr, Kemper AR, Kubik M, Landefeld CS, Mangione CM, Phipps MG, Silverstein M, Simon MA, Tseng CW, Wong JB. Screening for Cervical Cancer: US Preventive Services Task Force Recommendation Statement. JAMA. 2018;320:674-686.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  FUTURE II Study Group. Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions. N Engl J Med. 2007;356:1915-1927.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Shrestha AD, Neupane D, Vedsted P, Kallestrup P.   Cervical Cancer Prevalence, Incidence and Mortality in Low and Middle Income Countries: A Systematic Review Asian Pac J Cancer Prev 2018; 19: 319-324.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Strickler HD, Burk RD, Fazzari M, Anastos K, Minkoff H, Massad LS, Hall C, Bacon M, Levine AM, Watts DH, Silverberg MJ, Xue X, Schlecht NF, Melnick S, Palefsky JM. Natural history and possible reactivation of human papillomavirus in human immunodeficiency virus-positive women. J Natl Cancer Inst. 2005;97:577-586.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  García-Piñeres AJ, Hildesheim A, Herrero R, Trivett M, Williams M, Atmetlla I, Ramírez M, Villegas M, Schiffman M, Rodríguez AC, Burk RD, Hildesheim M, Freer E, Bonilla J, Bratti C, Berzofsky JA, Pinto LA. Persistent human papillomavirus infection is associated with a generalized decrease in immune responsiveness in older women. Cancer Res. 2006;66:11070-11076.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Bhatia J, Bratcher J, Korelitz B, Vakher K, Mannor S, Shevchuk M, Panagopoulos G, Ofer A, Tamas E, Kotsali P, Vele O. Abnormalities of uterine cervix in women with inflammatory bowel disease. World J Gastroenterol. 2006;12:6167-6171.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Kane S, Khatibi B, Reddy D. Higher incidence of abnormal Pap smears in women with inflammatory bowel disease. Am J Gastroenterol. 2008;103:631-636.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Hutfless S, Fireman B, Kane S, Herrinton LJ. Screening differences and risk of cervical cancer in inflammatory bowel disease. Aliment Pharmacol Ther. 2008;28:598-605.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Marehbian J, Arrighi HM, Hass S, Tian H, Sandborn WJ. Adverse events associated with common therapy regimens for moderate-to-severe Crohn's disease. Am J Gastroenterol. 2009;104:2524-2533.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Lees CW, Critchley J, Chee N, Beez T, Gailer RE, Williams AR, Shand AG, Arnott ID, Satsangi J. Lack of association between cervical dysplasia and IBD: a large case-control study. Inflamm Bowel Dis. 2009;15:1621-1629.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Singh H, Demers AA, Nugent Z, Mahmud SM, Kliewer EV, Bernstein CN. Risk of cervical abnormalities in women with inflammatory bowel disease: a population-based nested case-control study. Gastroenterology. 2009;136:451-458.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Rungoe C, Simonsen J, Riis L, Frisch M, Langholz E, Jess T. Inflammatory bowel disease and cervical neoplasia: a population-based nationwide cohort study. Clin Gastroenterol Hepatol. 2015;13:693-700.e1.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Segal JP, Askari A, Clark SK, Hart AL, Faiz OD. The Incidence and Prevalence of Human Papilloma Virus-associated Cancers in IBD. Inflamm Bowel Dis. 2021;27:34-39.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Li M, Yang QF, Cao Q, Tang J, Gao Y, Zhi M, Chao K, Su ML, Huang WM, Yi Y, Xia SY, Huang LJ, Zhao Y, Wang XH, Liu XY, Lin L, Hu PJ, Gao X. High-risk human papilloma virus infection and cervical neoplasm in female inflammatory bowel disease patients: a cross-sectional study. Gastroenterol Rep (Oxf). 2019;7:338-344.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Kim SC, Glynn RJ, Giovannucci E, Hernández-Díaz S, Liu J, Feldman S, Karlson EW, Schneeweiss S, Solomon DH. Risk of high-grade cervical dysplasia and cervical cancer in women with systemic inflammatory diseases: a population-based cohort study. Ann Rheum Dis. 2015;74:1360-1367.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Allegretti JR, Barnes EL, Cameron A. Are patients with inflammatory bowel disease on chronic immunosuppressive therapy at increased risk of cervical high-grade dysplasia/cancer? Inflamm Bowel Dis. 2015;21:1089-1097.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Singh H, Nugent Z, Demers AA, Bernstein CN. Screening for cervical and breast cancer among women with inflammatory bowel disease: a population-based study. Inflamm Bowel Dis. 2011;17:1741-1750.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Long MD, Porter CQ, Sandler RS, Kappelman MD. Suboptimal rates of cervical testing among women with inflammatory bowel disease. Clin Gastroenterol Hepatol. 2009;7:549-553.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Xu F, Dahlhamer JM, Terlizzi EP, Wheaton AG, Croft JB. Receipt of Preventive Care Services Among US Adults with Inflammatory Bowel Disease, 2015-2016. Dig Dis Sci. 2019;64:1798-1808.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Parian A, Lazarev M. Who and how to screen for cancer in at-risk inflammatory bowel disease patients. Expert Rev Gastroenterol Hepatol. 2015;9:731-746.  [PubMed]  [DOI]  [Cited in This Article: ]
125.  Goldacre MJ, Wotton CJ, Yeates D, Seagroatt V, Collier J. Liver cirrhosis, other liver diseases, pancreatitis and subsequent cancer: record linkage study. Eur J Gastroenterol Hepatol. 2008;20:384-392.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Cheddani H, Dauchet L, Fumery M, Charpentier C, Marie Bouvier A, Dupas JL, Pariente B, Peyrin-Biroulet L, Savoye G, Gower-Rousseau C. Cancer in Elderly Onset Inflammatory Bowel Disease: A Population-Based Study. Am J Gastroenterol. 2016;111:1428-1436.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Yadav S, Singh S, Harmsen WS, Edakkanambeth Varayil J, Tremaine WJ, Loftus EV Jr. Effect of Medications on Risk of Cancer in Patients With Inflammatory Bowel Diseases: A Population-Based Cohort Study from Olmsted County, Minnesota. Mayo Clin Proc. 2015;90:738-746.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Boffetta P. Tobacco smoking and risk of bladder cancer. Scand J Urol Nephrol Suppl. 2008;45-54.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Burger M, Catto JW, Dalbagni G, Grossman HB, Herr H, Karakiewicz P, Kassouf W, Kiemeney LA, La Vecchia C, Shariat S, Lotan Y. Epidemiology and risk factors of urothelial bladder cancer. Eur Urol. 2013;63:234-241.  [PubMed]  [DOI]  [Cited in This Article: ]
130.  Rini BI, Campbell SC, Escudier B. Renal cell carcinoma. Lancet. 2009;373:1119-1132.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Chow WH, Dong LM, Devesa SS. Epidemiology and risk factors for kidney cancer. Nat Rev Urol. 2010;7:245-257.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Balkwill F. Tumour necrosis factor and cancer. Nat Rev Cancer. 2009;9:361-371.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Galbán S, Fan J, Martindale JL, Cheadle C, Hoffman B, Woods MP, Temeles G, Brieger J, Decker J, Gorospe M. von Hippel-Lindau protein-mediated repression of tumor necrosis factor alpha translation revealed through use of cDNA arrays. Mol Cell Biol. 2003;23:2316-2328.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Biancone L, Orlando A, Kohn A, Colombo E, Sostegni R, Angelucci E, Rizzello F, Castiglione F, Benazzato L, Papi C, Meucci G, Riegler G, Petruzziello C, Mocciaro F, Geremia A, Calabrese E, Cottone M, Pallone F. Infliximab and newly diagnosed neoplasia in Crohn's disease: a multicentre matched pair study. Gut. 2006;55:228-233.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Biancone L, Petruzziello C, Orlando A, Kohn A, Ardizzone S, Daperno M, Angelucci E, Castiglione F, D'Incà R, Zorzi F, Papi C, Meucci G, Riegler G, Sica G, Rizzello F, Mocciaro F, Onali S, Calabrese E, Cottone M, Pallone F. Cancer in Crohn's Disease patients treated with infliximab: a long-term multicenter matched pair study. Inflamm Bowel Dis. 2011;17:758-766.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  Derikx LA, Nissen LH, Drenth JP, van Herpen CM, Kievit W, Verhoeven RH, Mulders PF, Hulsbergen-van de Kaa CA, Boers-Sonderen MJ, van den Heuvel TR, Pierik M, Nagtegaal ID, Hoentjen F; Dutch Initiative on Crohn and Colitis;  PALGA Group;  IBD/RCC Group. Better survival of renal cell carcinoma in patients with inflammatory bowel disease. Oncotarget. 2015;6:38336-38347.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Madanchi M, Zeitz J, Barthel C, Samaras P, Scharl S, Sulz MC, Biedermann L, Frei P, Vavricka SR, Rogler G, Scharl M. Malignancies in Patients with Inflammatory Bowel Disease: A Single-Centre Experience. Digestion. 2016;94:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]
138.  Bourrier A, Carrat F, Colombel JF, Bouvier AM, Abitbol V, Marteau P, Cosnes J, Simon T, Peyrin-Biroulet L, Beaugerie L; CESAME study group. Excess risk of urinary tract cancers in patients receiving thiopurines for inflammatory bowel disease: a prospective observational cohort study. Aliment Pharmacol Ther. 2016;43:252-261.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Wauters L, Billiet T, Papamichael K, Ballet V, Joniau S, Verschueren P, Silversmit G, Van Assche G, Vermeire S, Ferrante M. Incidence of renal cell carcinoma in inflammatory bowel disease patients with and without anti-TNF treatment. Eur J Gastroenterol Hepatol. 2017;29:84-90.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Ficarra V, Prayer-Galetti T, Novella G, Bratti E, Maffei N, Dal Bianco M, Artibani W, Pagano F. Incidental detection beyond pathological factors as prognostic predictor of renal cell carcinoma. Eur Urol. 2003;43:663-669.  [PubMed]  [DOI]  [Cited in This Article: ]
141.  Palsdottir HB, Hardarson S, Petursdottir V, Jonsson A, Jonsson E, Sigurdsson MI, Einarsson GV, Gudbjartsson T. Incidental detection of renal cell carcinoma is an independent prognostic marker: results of a long-term, whole population study. J Urol. 2012;187:48-53.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Lakatos PL, Szamosi T, Lakatos L. Smoking in inflammatory bowel diseases: good, bad or ugly? World J Gastroenterol. 2007;13:6134-6139.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Mahid SS, Minor KS, Soto RE, Hornung CA, Galandiuk S. Smoking and inflammatory bowel disease: a meta-analysis. Mayo Clin Proc. 2006;81:1462-1471.  [PubMed]  [DOI]  [Cited in This Article: ]
144.  Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Winther KV, Jess T, Langholz E, Munkholm P, Binder V. Long-term risk of cancer in ulcerative colitis: a population-based cohort study from Copenhagen County. Clin Gastroenterol Hepatol. 2004;2:1088-1095.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Burns JA, Weiner AB, Catalona WJ, Li EV, Schaeffer EM, Hanauer SB, Strong S, Burns J, Hussain MHA, Kundu SD. Inflammatory Bowel Disease and the Risk of Prostate Cancer. Eur Urol. 2019;75:846-852.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Meyers TJ, Weiner AB, Graff RE, Desai AS, Cooley LF, Catalona WJ, Hanauer SB, Wu JD, Schaeffer EM, Abdulkadir SA, Kundu SD, Witte JS. Association between inflammatory bowel disease and prostate cancer: A large-scale, prospective, population-based study. Int J Cancer. 2020;147:2735-2742.  [PubMed]  [DOI]  [Cited in This Article: ]
148.  Sfanos KS, Yegnasubramanian S, Nelson WG, De Marzo AM. The inflammatory microenvironment and microbiome in prostate cancer development. Nat Rev Urol. 2018;15:11-24.  [PubMed]  [DOI]  [Cited in This Article: ]
149.  Sfanos KS, Joshu CE. IBD as a risk factor for prostate cancer: what is the link? Nat Rev Urol. 2019;16:271-272.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  Porter CM, Shrestha E, Peiffer LB, Sfanos KS. The microbiome in prostate inflammation and prostate cancer. Prostate Cancer Prostatic Dis. 2018;21:345-354.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  De Marzo AM, Platz EA, Sutcliffe S, Xu J, Grönberg H, Drake CG, Nakai Y, Isaacs WB, Nelson WG. Inflammation in prostate carcinogenesis. Nat Rev Cancer. 2007;7:256-269.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  Lippi G, Montagnana M, Guidi GC. Epidemiological association between C-reactive protein and prostate-specific antigen. Cancer. 2009;115:1132.  [PubMed]  [DOI]  [Cited in This Article: ]
153.  Lehrer S, Diamond EJ, Mamkine B, Droller MJ, Stone NN, Stock RG. C-reactive protein is significantly associated with prostate-specific antigen and metastatic disease in prostate cancer. BJU Int. 2005;95:961-962.  [PubMed]  [DOI]  [Cited in This Article: ]
154.  Beer TM, Lalani AS, Lee S, Mori M, Eilers KM, Curd JG, Henner WD, Ryan CW, Venner P, Ruether JD, Chi KN; ASCENT Investigators. C-reactive protein as a prognostic marker for men with androgen-independent prostate cancer: results from the ASCENT trial. Cancer. 2008;112:2377-2383.  [PubMed]  [DOI]  [Cited in This Article: ]
155.  Sevcenco S, Mathieu R, Baltzer P, Klatte T, Fajkovic H, Seitz C, Karakiewicz PI, Rouprêt M, Rink M, Kluth L, Trinh QD, Loidl W, Briganti A, Scherr DS, Shariat SF. The prognostic role of preoperative serum C-reactive protein in predicting the biochemical recurrence in patients treated with radical prostatectomy. Prostate Cancer Prostatic Dis. 2016;19:163-167.  [PubMed]  [DOI]  [Cited in This Article: ]
156.  Nguyen DP, Li J, Tewari AK. Inflammation and prostate cancer: the role of interleukin 6 (IL-6). BJU Int. 2014;113:986-992.  [PubMed]  [DOI]  [Cited in This Article: ]
157.  Culig Z, Puhr M. Interleukin-6 and prostate cancer: Current developments and unsolved questions. Mol Cell Endocrinol. 2018;462:25-30.  [PubMed]  [DOI]  [Cited in This Article: ]
158.  Shariat SF, Kattan MW, Traxel E, Andrews B, Zhu K, Wheeler TM, Slawin KM. Association of pre- and postoperative plasma levels of transforming growth factor beta(1) and interleukin 6 and its soluble receptor with prostate cancer progression. Clin Cancer Res. 2004;10:1992-1999.  [PubMed]  [DOI]  [Cited in This Article: ]
159.  Rais R, Jiang W, Zhai H, Wozniak KM, Stathis M, Hollinger KR, Thomas AG, Rojas C, Vornov JJ, Marohn M, Li X, Slusher BS. FOLH1/GCPII is elevated in IBD patients, and its inhibition ameliorates murine IBD abnormalities. JCI Insight. 2016;1.  [PubMed]  [DOI]  [Cited in This Article: ]
160.  Date AA, Rais R, Babu T, Ortiz J, Kanvinde P, Thomas AG, Zimmermann SC, Gadiano AJ, Halpert G, Slusher BS, Ensign LM. Local enema treatment to inhibit FOLH1/GCPII as a novel therapy for inflammatory bowel disease. J Control Release. 2017;263:132-138.  [PubMed]  [DOI]  [Cited in This Article: ]
161.  Zhang T, Song B, Zhu W, Xu X, Gong QQ, Morando C, Dassopoulos T, Newberry RD, Hunt SR, Li E. An ileal Crohn's disease gene signature based on whole human genome expression profiles of disease unaffected ileal mucosal biopsies. PLoS One. 2012;7:e37139.  [PubMed]  [DOI]  [Cited in This Article: ]
162.  Yao V, Parwani A, Maier C, Heston WD, Bacich DJ. Moderate expression of prostate-specific membrane antigen, a tissue differentiation antigen and folate hydrolase, facilitates prostate carcinogenesis. Cancer Res. 2008;68:9070-9077.  [PubMed]  [DOI]  [Cited in This Article: ]
163.  Kaittanis C, Andreou C, Hieronymus H, Mao N, Foss CA, Eiber M, Weirich G, Panchal P, Gopalan A, Zurita J, Achilefu S, Chiosis G, Ponomarev V, Schwaiger M, Carver BS, Pomper MG, Grimm J. Prostate-specific membrane antigen cleavage of vitamin B9 stimulates oncogenic signaling through metabotropic glutamate receptors. J Exp Med. 2018;215:159-175.  [PubMed]  [DOI]  [Cited in This Article: ]
164.  Ge Y, Shi Q, Yao W, Cheng Y, Ma G. The association between inflammatory bowel disease and prostate cancer risk: a meta-analysis. Prostate Cancer Prostatic Dis. 2020;23:53-58.  [PubMed]  [DOI]  [Cited in This Article: ]
165.  Chen M, Yuan C, Xu T. An increase in prostate cancer diagnosis during inflammatory bowel disease: A systematic review and meta-analysis. Clin Res Hepatol Gastroenterol. 2020;44:302-309.  [PubMed]  [DOI]  [Cited in This Article: ]
166.  Carli E, Caviglia GP, Pellicano R, Fagoonee S, Rizza S, Astegiano M, Saracco GM, Ribaldone DG. Incidence of Prostate Cancer in Inflammatory Bowel Disease: A Meta-Analysis. Medicina (Kaunas). 2020;56.  [PubMed]  [DOI]  [Cited in This Article: ]
167.  Long MD, Herfarth HH, Pipkin CA, Porter CQ, Sandler RS, Kappelman MD. Increased risk for non-melanoma skin cancer in patients with inflammatory bowel disease. Clin Gastroenterol Hepatol. 2010;8:268-274.  [PubMed]  [DOI]  [Cited in This Article: ]
168.  Singh H, Nugent Z, Demers AA, Bernstein CN. Increased risk of nonmelanoma skin cancers among individuals with inflammatory bowel disease. Gastroenterology. 2011;141:1612-1620.  [PubMed]  [DOI]  [Cited in This Article: ]
169.  Long MD, Martin CF, Pipkin CA, Herfarth HH, Sandler RS, Kappelman MD. Risk of melanoma and nonmelanoma skin cancer among patients with inflammatory bowel disease. Gastroenterology. 2012;143:390-399.e1.  [PubMed]  [DOI]  [Cited in This Article: ]
170.  Brin L, Zubair AS, Brewer JD. Optimal management of skin cancer in immunosuppressed patients. Am J Clin Dermatol. 2014;15:339-356.  [PubMed]  [DOI]  [Cited in This Article: ]
171.  Peyrin-Biroulet L, Khosrotehrani K, Carrat F, Bouvier AM, Chevaux JB, Simon T, Carbonnel F, Colombel JF, Dupas JL, Godeberge P, Hugot JP, Lémann M, Nahon S, Sabaté JM, Tucat G, Beaugerie L; Cesame Study Group. Increased risk for nonmelanoma skin cancers in patients who receive thiopurines for inflammatory bowel disease. Gastroenterology. 2011;141:1621-28.e1.  [PubMed]  [DOI]  [Cited in This Article: ]
172.  Magro F, Peyrin-Biroulet L, Sokol H, Aldeger X, Costa A, Higgins PD, Joyce JC, Katsanos KH, Lopez A, de Xaxars TM, Toader E, Beaugerie L. Extra-intestinal malignancies in inflammatory bowel disease: results of the 3rd ECCO Pathogenesis Scientific Workshop (III). J Crohns Colitis. 2014;8:31-44.  [PubMed]  [DOI]  [Cited in This Article: ]
173.  Kubica AW, Brewer JD. Melanoma in immunosuppressed patients. Mayo Clin Proc. 2012;87:991-1003.  [PubMed]  [DOI]  [Cited in This Article: ]
174.  Dillon P, Thomas N, Sharpless N, Collichio F. Regression of advanced melanoma upon withdrawal of immunosuppression: case series and literature review. Med Oncol. 2010;27:1127-1132.  [PubMed]  [DOI]  [Cited in This Article: ]
175.  van Schaik FD, van Oijen MG, Smeets HM, van der Heijden GJ, Siersema PD, Oldenburg B. Risk of nonmelanoma skin cancer in patients with inflammatory bowel disease who use thiopurines is not increased. Clin Gastroenterol Hepatol. 2011;9:449-50.e1; author reply 450.  [PubMed]  [DOI]  [Cited in This Article: ]
176.  Peyrin-Biroulet L, Chevaux JB, Bouvier AM, Carrat F, Beaugerie L. Risk of melanoma in patients who receive thiopurines for inflammatory bowel disease is not increased. Am J Gastroenterol. 2012;107:1443-1444.  [PubMed]  [DOI]  [Cited in This Article: ]
177.  Abbas AM, Almukhtar RM, Loftus EV Jr, Lichtenstein GR, Khan N. Risk of melanoma and non-melanoma skin cancer in ulcerative colitis patients treated with thiopurines: a nationwide retrospective cohort. Am J Gastroenterol. 2014;109:1781-1793.  [PubMed]  [DOI]  [Cited in This Article: ]
178.  McKenna MR, Stobaugh DJ, Deepak P. Melanoma and non-melanoma skin cancer in inflammatory bowel disease patients following tumor necrosis factor-α inhibitor monotherapy and in combination with thiopurines: analysis of the Food and Drug Administration Adverse Event Reporting System. J Gastrointestin Liver Dis. 2014;23:267-271.  [PubMed]  [DOI]  [Cited in This Article: ]
179.  Kopylov U, Vutcovici M, Kezouh A, Seidman E, Bitton A, Afif W. Risk of Lymphoma, Colorectal and Skin Cancer in Patients with IBD Treated with Immunomodulators and Biologics: A Quebec Claims Database Study. Inflamm Bowel Dis. 2015;21:1847-1853.  [PubMed]  [DOI]  [Cited in This Article: ]
180.  Scott FI, Mamtani R, Brensinger CM, Haynes K, Chiesa-Fuxench ZC, Zhang J, Chen L, Xie F, Yun H, Osterman MT, Beukelman T, Margolis DJ, Curtis JR, Lewis JD. Risk of Nonmelanoma Skin Cancer Associated With the Use of Immunosuppressant and Biologic Agents in Patients With a History of Autoimmune Disease and Nonmelanoma Skin Cancer. JAMA Dermatol. 2016;152:164-172.  [PubMed]  [DOI]  [Cited in This Article: ]
181.  Nissen LHC, Pierik M, Derikx LAAP, de Jong E, Kievit W, van den Heuvel TRA, van Rosendael AR, Plasmeijer EI, Dewint P, Verhoeven RHA, Overbeek LIH, Nagtegaal ID, Hoentjen F, van der Meulen-de Jong AE. Risk Factors and Clinical Outcomes in Patients with IBD with Melanoma. Inflamm Bowel Dis. 2017;23:2018-2026.  [PubMed]  [DOI]  [Cited in This Article: ]
182.  Clowry J, Sheridan J, Healy R, Deady S, Keegan D, Byrne K, Cullen G, Mulcahy H, Comber H, Parnell AC, Doherty G, Lally A. Increased non-melanoma skin cancer risk in young patients with inflammatory bowel disease on immunomodulatory therapy: a retrospective single-centre cohort study. J Eur Acad Dermatol Venereol. 2017;31:978-985.  [PubMed]  [DOI]  [Cited in This Article: ]
183.  Khan N, Patel D, Trivedi C, Kavani H, Medvedeva E, Pernes T, Xie D, Lewis J, Yang YX. Repeated Occurrences of Basal Cell Cancer in Patients With Inflammatory Bowel Disease Treated With Immunosuppressive Medications. Am J Gastroenterol. 2020;115:1246-1252.  [PubMed]  [DOI]  [Cited in This Article: ]
184.  Ariyaratnam J, Subramanian V. Association between thiopurine use and nonmelanoma skin cancers in patients with inflammatory bowel disease: a meta-analysis. Am J Gastroenterol. 2014;109:163-169.  [PubMed]  [DOI]  [Cited in This Article: ]
185.  Singh S, Nagpal SJ, Murad MH, Yadav S, Kane SV, Pardi DS, Talwalkar JA, Loftus EV Jr. Inflammatory bowel disease is associated with an increased risk of melanoma: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2014;12:210-218.  [PubMed]  [DOI]  [Cited in This Article: ]
186.  Esse S, Mason KJ, Green AC, Warren RB. Melanoma Risk in Patients Treated With Biologic Therapy for Common Inflammatory Diseases: A Systematic Review and Meta-analysis. JAMA Dermatol. 2020;156:787-794.  [PubMed]  [DOI]  [Cited in This Article: ]
187.  Annese V, Beaugerie L, Egan L, Biancone L, Bolling C, Brandts C, Dierickx D, Dummer R, Fiorino G, Gornet JM, Higgins P, Katsanos KH, Nissen L, Pellino G, Rogler G, Scaldaferri F, Szymanska E, Eliakim R; ECCO. European Evidence-based Consensus: Inflammatory Bowel Disease and Malignancies. J Crohns Colitis. 2015;9:945-965.  [PubMed]  [DOI]  [Cited in This Article: ]
188.  Beaugerie L, Carrat F, Colombel JF, Bouvier AM, Sokol H, Babouri A, Carbonnel F, Laharie D, Faucheron JL, Simon T, de Gramont A, Peyrin-Biroulet L; CESAME Study Group. Risk of new or recurrent cancer under immunosuppressive therapy in patients with IBD and previous cancer. Gut. 2014;63:1416-1423.  [PubMed]  [DOI]  [Cited in This Article: ]
189.  Shelton E, Laharie D, Scott FI, Mamtani R, Lewis JD, Colombel JF, Ananthakrishnan AN. Cancer Recurrence Following Immune-Suppressive Therapies in Patients With Immune-Mediated Diseases: A Systematic Review and Meta-analysis. Gastroenterology. 2016;151:97-109.e4.  [PubMed]  [DOI]  [Cited in This Article: ]
190.  Anderson A, Ferris LK, Click B, Ramos-Rivers C, Koutroubakis IE, Hashash JG, Dunn M, Barrie A, Schwartz M, Regueiro M, Binion DG. Low Rates of Dermatologic Care and Skin Cancer Screening Among Inflammatory Bowel Disease Patients. Dig Dis Sci. 2018;63:2729-2739.  [PubMed]  [DOI]  [Cited in This Article: ]