Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. May 28, 2016; 22(20): 4794-4801
Published online May 28, 2016. doi: 10.3748/wjg.v22.i20.4794
Inflammatory bowel disease and cancer: The role of inflammation, immunosuppression, and cancer treatment
Jordan E Axelrad, Simon Lichtiger, Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, NY 10032, United States
Vijay Yajnik, Department of Medicine, Division of Gastroenterology, The Massachusetts General Hospital, Boston, MA 02445, United States
Author contributions: Axelrad JE, Lichtiger S and Yajnik V wrote the paper.
Conflict-of-interest statement: Authors declare no conflict of interests for this article.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (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/
Correspondence to: Vijay Yajnik, MD, PhD, Department of Medicine, Division of Gastroenterology, The Massachusetts General Hospital, Crohn’s and Colitis Center, 165 Cambridge Street 9th Floor, Boston, MA 02445, United States. vyajnik@mgh.harvard.edu
Telephone: +1-617-7246005 Fax: +1-617-7263080
Received: February 11, 2016
Peer-review started: February 11, 2016
First decision: March 21, 2016
Revised: March 25, 2016
Accepted: April 7, 2016
Article in press: April 7, 2016
Published online: May 28, 2016

Abstract

In patients with inflammatory bowel disease (IBD), chronic inflammation is a major risk factor for the development of gastrointestinal malignancies. The pathogenesis of colitis-associated cancer is distinct from sporadic colorectal carcinoma and the critical molecular mechanisms underlying this process have yet to be elucidated. Patients with IBD have also been shown to be at increased risk of developing extra-intestinal malignancies. Medical therapies that diminish the mucosal inflammatory response represent the foundation of treatment in IBD, and recent evidence supports their introduction earlier in the disease course. However, therapies that alter the immune system, often used for long durations, may also promote carcinogenesis. As the population of patients with IBD grows older, with longer duration of chronic inflammation and longer exposure to immunosuppression, there is an increasing risk of cancer development. Many of these patients will require cancer treatment, including chemotherapy, radiation, hormonal therapy, and surgery. Many patients will require further treatment for their IBD. This review seeks to explore the characteristics and risks of cancer in patients with IBD, and to evaluate the limited data on patients with IBD and cancer, including management of IBD after a diagnosis of cancer, the effects of cancer treatment on IBD, and the effect of IBD and medications for IBD on cancer outcomes.

Key Words: Inflammatory bowel disease, Cancer, Anti-tumor necrosis factor, Immunosuppression, Chemotherapy, Radiation

Core tip: Patients with inflammatory bowel disease (IBD) and cancer represent a challenging population. Gastroenterologists and oncologists caring for patients with IBD and cancer are increasingly confronted with questions regarding the management of IBD after a diagnosis of cancer, and conversely, the management of cancer in patients with IBD. This review seeks to explore the characteristics, risks, and pathogenesis of cancer in patients with IBD, and to evaluate the data on patients with IBD and cancer, including the interaction between IBD and cancer treatment.



INTRODUCTION

Crohn’s disease (CD) and ulcerative colitis (UC) are chronic inflammatory conditions of the gastrointestinal tract. Although the disease pathogenesis is not fully understood, inflammatory bowel disease (IBD) is characterized by chronic inflammation of the gastrointestinal tract in genetically susceptible individuals exposed to environmental risk factors. Together, IBD is estimated to affect more than 0.4% of Europeans and North Americans, a number that is expected to increase over time[1]. It is well recognized that patients with IBD are at an increased risk of developing colorectal cancer (CRC), primarily the result of chronic intestinal inflammation[2-4]. More recently, patients with IBD have also been shown to be at increased risk of developing extra-intestinal malignancies, thought to be a consequence of immunosuppressive therapies and an underlying inflammatory state[5].

As the population of patients with IBD grows and ages, there is an inevitable increase in the risk of cancer development. Moreover, many of these patients may require cancer treatment, including chemotherapy, radiation, and immunotherapy, and many may require further treatment for their IBD. The focus of this review is to evaluate the characteristics, pathogenesis, and risks of cancer in patients with IBD, and to explore the relationship between IBD and cancer treatment.

IBD AND RISK OF CANCER
Cancer secondary to chronic intestinal inflammation

In patients with IBD, chronic intestinal inflammation is the primary risk factor for the development of gastrointestinal malignancy. Cancers as a result of chronic intestinal inflammation include CRC, small bowel adenocarcinoma, intestinal lymphoma, anal cancer, and cholangiocarcinoma (Table 1)[6].

Table 1 Cancer secondary to chronic intestinal inflammation.
Cancer typeStandardized incidence ratio
Colorectal cancer[3]5.7 (95%CI: 4.6-7.0)
Small bowel adenocarcinoma[20]27.1 (95%CI: 14.9-49.2)
Intestinal lymphoma[36]17.51 (95%CI: 6.43-38.11)
Anal cancer[60]Data not available
Cholangiocarcinoma[23]916.63 (95%CI: 297.88-2140.99) in UC

The risk and pathogenesis of inflammation-associated cancer has chiefly been described in colitis-associated CRC. In a meta analysis, quantitative estimates of CRC risk in UC have been reported to be 2% after 10 years, 8% after 20 years, and 18% after 30 years of disease[3]. Moreover, studies of CRC in UC have noted a high concordance between CRC risk with the location and extent of disease, with a standardized incidence ratio (SIR) of 1.7 for proctitis, 2.8 for left-sided colitis, and 14.8 for pancolitis[7]. All of these studies support the strong association between inflammation and cancer development.

Patients with IBD develop colon cancer in a manner similar to well described sporadic molecular mechanisms including mutations in the adenomatous polyposis coli (APC) gene, aneuploidy, DNA methylation, microsatellite instability (MSI), activation of the oncogene k-ras, activation of COX-2, and mutation in tumor suppressor genes DCC/DPC4, and eventual loss of p53 function[8]. However, underlying colonic inflammation changes the timing and sequence of these genomic changes, yielding a process of carcinogenesis that is faster and multifocal[4]. Contrary to sporadic cancers in which the dysplastic precursor is the adenomatous polyp, dysplasia in patients with IBD can be localized, diffuse, or multifocal[4,9].

Studies mapping genomic instability secondary to DNA aneuploidy in patients with IBD indicate that these cell populations became more widely distributed, occupying larger areas of colonic mucosa[9]. Over time, further subpopulations with increasingly unstable genomics arise and expand, representing a whole field change, marking the entire colon at risk for further carcinogenesis[9,10].

In terms of specific molecular mechanisms that differ between colitis-associated cancer and sporadic cancer, early mutation in p53 is thought to play a fundamental role. Changes in p53 have been found in up to 85% of colitis-associated cancers[11]. Furthermore, alterations in p53 have been observed in biopsies from inflamed mucosa in more than 50% of patients with UC who did not have cancer, indicating a significant role of inflammation in these mutations[12]. In addition, loss of APC, an early event in the development of sporadic CRC, is less frequent and tends to occur later in colitis-associated cancer[13]. DNA methylation also differs with increased hypermethylation of several genes, including hMLH1 and p16, occurring earlier and contributing to microsatellite instability[14].

The immune response and oxidative stress play a critical role in the initiation and progression of carcinogenesis, contributing to the aforementioned molecular mechanisms leading to cancer. The inflammatory microenvironment of IBD, consisting of a variety of immune cells, epithelial cells, stromal cells, cytokines, and chemokines, has many similarities to the microenvironment of cancers, suggesting similar inflammatory mediators and mechanisms that promote both IBD and cancer development[15]. These mediators, produced by inflammatory cells, include tumor necrosis factor alpha (TNF-α), ILs-1, 6, 12, 13, 17, 22, and 23[15]. The interaction between the signaling of these cytokines and immune response play a major role in inflammation and colitis-associated cancer.

The increased expression of several inflammation-associated genes in IBD, such as cyclooxygenase-2 (COX-2) and nitric oxide synthase-2 (NOS-2), have also been noted in colonic neoplasia[12]. It is thought that reactive oxygen and nitrogen species produced by inflammatory cells expressing these genes not only directly damage colonic epithelium, but also contribute to the genetic alterations driving carcinogenesis[9].

In addition, alterations in the microbiota contribute to colitis-associated cancer. In mouse models of colitis-associated cancer susceptible to inflammation or cancer, cancer did not develop when the mice were germ-free or treated with antibiotics[16,17]. Studies of the microbiota in patients with CRC have demonstrated varying populations of bacteria that differ from cancer-free controls, suggesting that the complex interaction between the host genome, colonic epithelial-cell receptors, and the luminal microbiota create an environment conducive to carcinogenesis. Stool samples derived from CRC patients had higher levels of Fusobacterium, Enterococcus, Escherichia, Shigella, Klebsiella, Streptococcus, and Peptostreptococcus, Firmicutes, Bacteroidetes, and a depletion of bacteria belonging to Lachnospiraceae family compared to controls[18,19]. Although we are just beginning to understand the association between specific gastrointestinal microbes and cancer, much remains unknown regarding the causes and effects of these relationships and how manipulating the microbiome may have therapeutic potential.

In addition to CRC, small-bowel adenocarcinoma, specifically ileal carcinoma, has been shown to be significantly associated with the severity and duration of CD, and it is 20 to 30 fold more common in patients with CD compared to the general population[20]. Moreover, it is often found in areas with previous or synchronous ileal dysplasia, suggesting that it may evolve in a similar manner to the molecular and immune mechanisms of CRC described above[21]. In addition, cholangiocarcinoma, when associated with UC-primary sclerosing cholangitis (PSC), yields a risk nearly 160 fold greater than controls, suggesting the inflammatory state of IBD-PSC may contribute to biliary carcinogenesis[22,23].

Cancer secondary to immunosuppression

Given that chronic inflammation underlies the disease state of IBD, medications that mitigate inflammation by suppression of the immune system represent the cornerstone of treatment. In addition to treating IBD, it is postulated that these medications, such as immunomodulators [thiopurines (azathioprine or mercaptopurine) or methotrexate] and biologic agents (TNF-α antagonists), may reduce the incidence of inflammation-associated cancer. However, given that immunomodulators and biologic agents act on the immune system, they may also promote carcinogenesis.

Thiopurines and methotrexate promote the development of cancer by a variety of mechanisms including direct alteration in DNA, activation of oncogenes, reduction in physiologic immunosurveillance of malignant cells, and impaired immune control of oncogenic viruses[24-26]. Less is known about the carcinogenic potential of biologic therapies that block TNF-α and existing molecular data is inconsistent. TNF-α has been shown to exhibit anti-tumor effects by initiating cellular apoptosis of malignant cells, but it is secreted by most tumors to facilitate cellular survival and enhance neoplastic proliferation as a pro-tumor inflammatory cytokine[27-29].

Several studies have indicated a risk of therapy-associated malignancies in IBD patients. Population-based cohort and meta-analyses have demonstrated that current use of thiopurines for IBD is associated with a 1.3 to 1.7 overall relative risk of cancer, which is reversible after withdrawal[30,31]. Current exposure to TNF-α antagonists has not been shown to be associated with an overall excess risk of cancer, but data is very limited[32]. Specific cancers thought secondary to long-standing immunosuppression in the setting of IBD include lymphomas, acute myeloid leukemia, myelodysplastic syndromes, skin cancers, and urinary tract cancers (Table 2).

Table 2 Cancer secondary to immunosuppression.
Increased risk under anti-metabolitesIncreased risk under anti-TNFαIncreased risk under anti-metabolite with anti-TNFα
Non-Hodgkin lymphoma[33-35]Melanoma[42]Hepatosplenic T-cell lymphoma[38]
Acute myeloid leukemia and Myelodysplastic syndromes[61]
Non-melanoma skin cancers (basal and squamous cell carcinomas)[39-41]
Urinary tract cancers[62]

For lymphoma, multiple studies have demonstrated incidence ratios of non-Hodgkin lymphoma following thiopurine exposure ranging from 1.6 to 37.5, with no excess risk attributed to IBD itself[33-35]. The exception to this is primary intestinal lymphoma, where duration and severity of CD play a primary role[36]. In the setting of thiopurines, most lymphoma is Epstein-Barr virus (EBV)-associated and thought to result from the loss of immune control of EBV-infected B lymphocytes[37]. Furthermore, there have been several cases of fatal early postmononucleosis lymphoma in young men who are previously seronegative for EBV[33]. In addition, Hepatosplenic T-cell Lymphoma, though very rare, is primarily associated with thiopurine exposure in combination with TNF-α antagonists in both adolescent and young males[38]. However, recent data suggests that there is no excess risk of lymphoma in patients with IBD exposed to TNF-α antagonists[32].

In a study from the Cancers Et Surrisque Associé aux Maladies inflammatoires intestinales En France (CESAME) cohort, the risk of myeloid disorders was not increased among patients with IBD or ongoing thiopurine treatment (SIR = 1.54, 95%CI: 0.05-8.54), but patients with past exposures to thiopurines had an increased risk of myeloid disorders (SIR = 6.98; 95%CI: 1.44-20.36)[31].

For skin cancers, there is substantial evidence that thiopurines increase the risk of basal cell and squamous cell carcinomas, collectively known as nonmelanoma skin cancers (NMSC)[39-41]. In another study from the CESAME group, an increased risk of NMSC was observed in the patients with IBD and associated with ongoing thiopurine exposure (HR = 5.9; 95%CI: 2.1-16.4) and past thiopurine exposure (HR = 3.9; 95%CI: 1.3-12.1)[41]. However, in a large retrospective cohort of patients with IBD, there was no excess risk of nonmelanoma skin cancer attributable to TNF-α antagonists[40]. In addition, studies have demonstrated an increased risk of melanoma in patients with IBD, with no increased risk associated with thiopurine exposure[40-42]. Conversely, patients exposed to TNF-α antagonists have been found to be 1.5 to 2 times more likely to develop melanoma to patients with IBD who were not exposed to TNF-α antagonists[32]. As such, thiopurines increase the risk of NMSC whereas TNF-α antagonists increase the risk of melanoma.

Secondary or recurrent cancer in patients with a history of cancer

Given the above-mentioned risks of immunomodulator and biologic-associated malignancy, patients with a history of cancer were excluded from clinical trials of TNF-α antagonists. Additionally, there is substantial data within the transplant literature indicating that immunosuppression, such as thiopurines and calcineurin inhibitors, increases the risk of new and recurrent malignancies in patients with a history of cancer[43,44]. As such, oncologists and gastroenterologists generally suspend immunosuppression for IBD after a diagnosis of cancer, both while undergoing cancer treatment and during remission from cancer. This approach may worsen IBD and even complicate appropriate cancer management. Although there is little data on patients with IBD and a history of cancer, there is emerging data regarding the management of IBD after a diagnosis of cancer.

In 17047 patients in the CESAME prospective observational cohort, exposure to immunosuppression was independently associated with the development of cancer with an adjusted HR of 1.9 (95%CI: 1.2-3.0)[31]. However, it did not increase the risk of new or recurrent cancer in patients with a history of cancer[31]. Given the limited number of patients with IBD and a history of cancer with subsequent exposure to immunosuppression in the cohort, this conclusion only applied to thiopurine exposure and no conclusions were drawn on anti-TNF-α therapies[31].

A similar study from the New York Crohn’s and Colitis Organization (NYCCO) representing a consortium of 8 academic medical centers found that nearly 30% of patients with IBD and a history of cancer developed new or recurrent cancer[45]. However, exposure to TNF-α antagonists, antimetabolites, or the combination was not associated with an increased risk of new or recurrent cancer within 5 years following a diagnosis of cancer (Log-rank P = 0.14)[45]. Furthermore, after adjusting for the risk of recurrence of prior cancer, there was still no difference in risk of new or recurrent cancer between exposure groups (anti-TNF-α HR = 0.32, 95%CI: 0.09-1.09; anti-TNF-α with an antimetabolite HR = 0.64, 95%CI: 0.26-1.59; antimetabolite HR = 1.08, 95%CI: 0.54-2.15)[45].

In addition, data from NYCCO showed that duration of anti-TNF-α after a diagnosis of cancer was not associated with the risk or type of new or recurrent cancer[45]. Studies within the rheumatoid arthritis literature corroborate these findings with data demonstrating no difference in the development of new or recurrent cancer in patients with a history of cancer who were subsequently exposed to anti-TNF-α agents compared with those receiving disease-modifying anti-rheumatic drugs alone[46,47]. However, given small sample sizes, these studies often grouped different types of cancers together. In the NYCCO study for example, all solid malignancies, such as breast, prostate, and lung, were grouped together. This statistical approach may not reflect the natural biologic activity of carcinogenesis and the direct effects of immunosuppression on cancer development, limiting the ability to draw conclusions on specific cancers.

CANCER TREATMENT AND IBD

While data on the risk of new or recurrent cancer under immunosuppression in patients with IBD and a history of cancer is limited, though increasing, considerably less is known regarding the effects of cancer treatment on IBD, and the effect of IBD and medications for IBD on important cancer outcomes.

Effect of cancer treatment on IBD

In a study from the Massachusetts General Hospital, 84 patients with IBD and extra-intestinal cancer were assessed for the effect of cancer treatment on the course of IBD[48]. The authors found that 66.7% of patients with active IBD at their cancer diagnosis experienced remission from IBD thought secondary to cytotoxic chemotherapy. Conversely, 17.4% of patients in remission from IBD at their cancer diagnosis experienced a flare during or within 5 years after their cancer treatment[48]. In the remission group, the authors found the risk of flare to be greatest among patients who received hormonal therapies (combination cytotoxic chemotherapy with adjuvant hormone therapy HR = 12.25, 95%CI: 1.51-99.06; hormone monotherapy HR = 11.56, 95%CI: 1.39-96.43). This suggests that hormonal therapies for cancer, such as breast and prostate, may increase the risk of IBD reactivation or counter the protective effects of cytotoxic chemotherapy[48]. A majority of patients with active IBD at their cancer diagnosis appeared to benefit from cancer treatment in the form of IBD remission, which was much more likely if the cancer treatment included cytotoxic chemotherapeutics and less likely if patients were treated with hormonal monotherapy[48].

In this cohort, there was no appreciable modification in IBD medications after a diagnosis of cancer. TNF-α antagonists were continued in three patients and the proportion of patients maintained on immunomodulators decreased slightly from 22% to 14% after a cancer diagnosis[48]. These data, however, were not compared to a control group of patients without chemotherapy or without cancer to assess whether patients with IBD and extra-intestinal cancer experienced a course of IBD different from patients without chemotherapy or cancer.

However, other studies have demonstrated a major modification in IBD medications after a diagnosis of cancer. In a study from a French clinical prospective database, a diagnosis of extra-intestinal cancer had a marked impact on the management of IBD, but was not associated with significant modifications in activity of IBD[49]. A diagnosis of extra-intestinal cancer led to some changes in therapeutic strategy, with a lesser use of thiopurines (19% vs 25%, P < 0.001) and an increased use of intestinal surgery (4% vs 2.5%, P = 0.05)[49].

Effect of IBD on cancer

Little is known regarding specific cancer outcomes in patients with IBD. Oncologists have generally been reluctant to administer pelvic irradiation in the setting of IBD, as the tolerance of pelvic irradiation in these patients is largely unknown. There exists only one study in the literature from Green et al[50] which retrospectively examined 47 patients with IBD and rectal cancer treated over a 34-year period (1960-1994) from the Mount Sinai Hospital, New York. The authors found a five-year overall survival rate of 42% and disease-free survival of 43%, which were comparable to results published for non-IBD-associated rectal cancer at that time, however, patients with high-grade tumors had statistically lower rates[50]. Complications, such as gastrointestinal morbidity or small bowel obstruction, were comparable to those reported in several large randomized trials of adjuvant chemoradiation therapy in rectal cancer arising in the general population[50].

In terms of chemotherapy and associated cancer outcomes, a small study on 8 patients with IBD and gastrointestinal malignancy showed that the most common gastrointestinal adverse event was diarrhea, with 38% of patients experiencing greater than 7 stools per day over baseline and/or fecal incontinence, all of which occurred in patients with CD[51]. Several studies have examined the effect of IBD medications on cancer outcomes. Multiple studies have demonstrated a role of anti-TNF-α in improving cachexia and increasing chemotherapy tolerance in patients with non-small cell lung cancer, renal cell carcinoma, and pancreatic cancer[52-54]. Moreover, in patients treated with TNF-α antagonists, the occurrence of cancer during treatment was not associated with a worse prognosis, and may even have a protective effect by reducing aggressive metastatic breast cancers at a cellular level[55,56].

Immunotherapies for cancer and immune-related colitis

Immunotherapy for cancer has shown promise in cases refractory to conventional treatment. However, unguided immune stimulation in cancer patients presents its own challenges. There are several reports of anti-cytotoxic T-lymphocyte-associated protein-4 antibodies used for melanoma, such as ipilimumab, and programmed cell death-1 receptor inhibitors used for melanoma and non-small cell lung cancer, such as and nivolumab, producing an immune-related colitis that is remarkably similar to IBD[57,58]. These medications, particularly when used in combination, result in clinical symptoms, endoscopic manifestations, and pathologic cellular infiltrates that emulate IBD. Fortunately, the majority of these cases respond to conventional treatments for IBD such as systemic corticosteroids, budesonide, and infliximab[57,58].

In a recent study, 50% of patients with advanced melanoma and baseline autoimmune disease, such as rheumatoid arthritis, IBD, and psoriasis, experienced either autoimmune exacerbations or immune-related adverse reactions when treated with ipilimumab[59]. These reactions were generally manageable with standard treatment including corticosteroids and infliximab[59]. As the field of immunotherapy for cancer evolves, we may see an increase in immune mediated colitis, which highlights the important role for T-cell checkpoint inhibitors in exacerbating IBD or causing an IBD-like colitis.

CONCLUSION

Patients with IBD are at an increased risk of cancer secondary to long-standing intestinal inflammation and secondary to immunosuppressive therapies. As the population of patients with IBD ages, there is an increasing risk of cancer development. Many of these patients will require cancer treatment and many will require further treatment for their IBD.

Much research is being devoted to exploring the role of chronic intestinal inflammation from IBD in carcinogenesis, and the role of immunosuppressive medications used to treat IBD in the promotion and prevention of cancer. Despite these efforts, much remains unknown regarding the interaction between IBD, medications for IBD, and cancer treatment, and the risk of cancer recurrence in patients with IBD and a history of cancer.

Understanding the effects of chemotherapy, hormonal therapies, radiation, and surgery for cancer on IBD may help identify patients at the highest risk for disease exacerbation during and after specific cancer treatments, especially in those who may require re-initiation of immunosuppressive therapies for IBD. In addition, while retrospective data has demonstrated some evidence for the safety of immunosuppression in patients with IBD and a history of cancer, prospective data are needed to validate these findings. Furthermore, data is lacking regarding specific cancers, treatments, and risk of recurrence under varying immunosuppressive medications for IBD. More data will permit the development of evidence-based, quantitative risk-benefit models including cancer and IBD-related variables to assist clinicians in managing this complex patient population.

Footnotes

P- Reviewer: Felix K, Gonzalez-Perez RR, Kanat O S- Editor: Qi Y L- Editor: A E- Editor: Wang CH

References
1.  Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, Benchimol EI, Panaccione R, Ghosh S, Barkema HW. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142:46-54.e42; quiz e30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3134]  [Cited by in F6Publishing: 3261]  [Article Influence: 271.8]  [Reference Citation Analysis (1)]
2.  Rutter MD, Saunders BP, Wilkinson KH, Rumbles S, Schofield G, Kamm MA, Williams CB, Price AB, Talbot IC, Forbes A. Thirty-year analysis of a colonoscopic surveillance program for neoplasia in ulcerative colitis. Gastroenterology. 2006;130:1030-1038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 492]  [Cited by in F6Publishing: 503]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
3.  Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut. 2001;48:526-535.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Ullman TA, Itzkowitz SH. Intestinal inflammation and cancer. Gastroenterology. 2011;140:1807-1816.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 738]  [Cited by in F6Publishing: 779]  [Article Influence: 59.9]  [Reference Citation Analysis (0)]
5.  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: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 192]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
6.  Beaugerie L, Itzkowitz SH. Cancers complicating inflammatory bowel disease. N Engl J Med. 2015;372:1441-1452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 351]  [Cited by in F6Publishing: 361]  [Article Influence: 40.1]  [Reference Citation Analysis (0)]
7.  Ekbom A, Helmick C, Zack M, Adami HO. Ulcerative colitis and colorectal cancer. A population-based study. N Engl J Med. 1990;323:1228-1233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1294]  [Cited by in F6Publishing: 1161]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
8.  Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA, Kinzler KW. Cancer genome landscapes. Science. 2013;339:1546-1558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5242]  [Cited by in F6Publishing: 5196]  [Article Influence: 472.4]  [Reference Citation Analysis (0)]
9.  Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol. 2004;287:G7-G17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 870]  [Cited by in F6Publishing: 857]  [Article Influence: 42.9]  [Reference Citation Analysis (0)]
10.  Rubin CE, Haggitt RC, Burmer GC, Brentnall TA, Stevens AC, Levine DS, Dean PJ, Kimmey M, Perera DR, Rabinovitch PS. DNA aneuploidy in colonic biopsies predicts future development of dysplasia in ulcerative colitis. Gastroenterology. 1992;103:1611-1620.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Yin J, Harpaz N, Tong Y, Huang Y, Laurin J, Greenwald BD, Hontanosas M, Newkirk C, Meltzer SJ. p53 point mutations in dysplastic and cancerous ulcerative colitis lesions. Gastroenterology. 1993;104:1633-1639.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Hussain SP, Amstad P, Raja K, Ambs S, Nagashima M, Bennett WP, Shields PG, Ham AJ, Swenberg JA, Marrogi AJ. Increased p53 mutation load in noncancerous colon tissue from ulcerative colitis: a cancer-prone chronic inflammatory disease. Cancer Res. 2000;60:3333-3337.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Aust DE, Terdiman JP, Willenbucher RF, Chang CG, Molinaro-Clark A, Baretton GB, Loehrs U, Waldman FM. The APC/beta-catenin pathway in ulcerative colitis-related colorectal carcinomas: a mutational analysis. Cancer. 2002;94:1421-1427.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Fleisher AS, Esteller M, Harpaz N, Leytin A, Rashid A, Xu Y, Liang J, Stine OC, Yin J, Zou TT. Microsatellite instability in inflammatory bowel disease-associated neoplastic lesions is associated with hypermethylation and diminished expression of the DNA mismatch repair gene, hMLH1. Cancer Res. 2000;60:4864-4868.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Francescone R, Hou V, Grivennikov SI. Cytokines, IBD, and colitis-associated cancer. Inflamm Bowel Dis. 2015;21:409-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 200]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
16.  Abreu MT, Peek RM. Gastrointestinal malignancy and the microbiome. Gastroenterology. 2014;146:1534-1546.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 210]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
17.  Irrazábal T, Belcheva A, Girardin SE, Martin A, Philpott DJ. The multifaceted role of the intestinal microbiota in colon cancer. Mol Cell. 2014;54:309-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 187]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
18.  Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, Barnes R, Watson P, Allen-Vercoe E, Moore RA. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22:299-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1164]  [Cited by in F6Publishing: 1324]  [Article Influence: 101.8]  [Reference Citation Analysis (0)]
19.  Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, Jia W, Cai S, Zhao L. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012;6:320-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 738]  [Cited by in F6Publishing: 834]  [Article Influence: 64.2]  [Reference Citation Analysis (0)]
20.  Jess T, Gamborg M, Matzen P, Munkholm P, Sørensen TI. Increased risk of intestinal cancer in Crohn’s disease: a meta-analysis of population-based cohort studies. Am J Gastroenterol. 2005;100:2724-2729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 393]  [Cited by in F6Publishing: 374]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
21.  Svrcek M, Piton G, Cosnes J, Beaugerie L, Vermeire S, Geboes K, Lemoine A, Cervera P, El-Murr N, Dumont S. Small bowel adenocarcinomas complicating Crohn’s disease are associated with dysplasia: a pathological and molecular study. Inflamm Bowel Dis. 2014;20:1584-1592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 59]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
22.  Singh S, Talwalkar JA. Primary sclerosing cholangitis: diagnosis, prognosis, and management. Clin Gastroenterol Hepatol. 2013;11:898-907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 81]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
23.  Manninen P, Karvonen AL, Laukkarinen J, Aitola P, Huhtala H, Collin P. Colorectal cancer and cholangiocarcinoma in patients with primary sclerosing cholangitis and inflammatory bowel disease. Scand J Gastroenterol. 2015;50:423-428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
24.  Münz C, Moormann A. Immune escape by Epstein-Barr virus associated malignancies. Semin Cancer Biol. 2008;18:381-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 59]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
25.  O’Donovan P, Perrett CM, Zhang X, Montaner B, Xu YZ, Harwood CA, McGregor JM, Walker SL, Hanaoka F, Karran P. Azathioprine and UVA light generate mutagenic oxidative DNA damage. Science. 2005;309:1871-1874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 455]  [Cited by in F6Publishing: 469]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
26.  Zitvogel L, Tesniere A, Kroemer G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol. 2006;6:715-727.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 866]  [Cited by in F6Publishing: 878]  [Article Influence: 48.8]  [Reference Citation Analysis (0)]
27.  Balkwill F. Tumour necrosis factor and cancer. Nat Rev Cancer. 2009;9:361-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1213]  [Cited by in F6Publishing: 1266]  [Article Influence: 84.4]  [Reference Citation Analysis (0)]
28.  Beaugerie L. Inflammatory bowel disease therapies and cancer risk: where are we and where are we going? Gut. 2012;61:476-483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
29.  Danial NN, Korsmeyer SJ. Cell death: critical control points. Cell. 2004;116:205-219.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  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: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 160]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
31.  Beaugerie L, Carrat F, Colombel JF, Bouvier AM, Sokol H, Babouri A, Carbonnel F, Laharie D, Faucheron JL, Simon T. 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: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 102]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
32.  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: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 188]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
33.  Beaugerie L, Brousse N, Bouvier AM, Colombel JF, Lémann M, Cosnes J, Hébuterne X, Cortot A, Bouhnik Y, Gendre JP. Lymphoproliferative disorders in patients receiving thiopurines for inflammatory bowel disease: a prospective observational cohort study. Lancet. 2009;374:1617-1625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 774]  [Cited by in F6Publishing: 748]  [Article Influence: 49.9]  [Reference Citation Analysis (0)]
34.  Korelitz BI, Mirsky FJ, Fleisher MR, Warman JI, Wisch N, Gleim GW. Malignant neoplasms subsequent to treatment of inflammatory bowel disease with 6-mercaptopurine. Am J Gastroenterol. 1999;94:3248-3253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 108]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
35.  Lewis JD, Bilker WB, Brensinger C, Deren JJ, Vaughn DJ, Strom BL. Inflammatory bowel disease is not associated with an increased risk of lymphoma. Gastroenterology. 2001;121:1080-1087.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Sokol H, Beaugerie L, Maynadié M, Laharie D, Dupas JL, Flourié B, Lerebours E, Peyrin-Biroulet L, Allez M, Simon T. Excess primary intestinal lymphoproliferative disorders in patients with inflammatory bowel disease. Inflamm Bowel Dis. 2012;18:2063-2071.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
37.  Pietersma F, Piriou E, van Baarle D. Immune surveillance of EBV-infected B cells and the development of non-Hodgkin lymphomas in immunocompromised patients. Leuk Lymphoma. 2008;49:1028-1041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 52]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
38.  Kotlyar DS, Osterman MT, Diamond RH, Porter D, Blonski WC, Wasik M, Sampat S, Mendizabal M, Lin MV, Lichtenstein GR. A systematic review of factors that contribute to hepatosplenic T-cell lymphoma in patients with inflammatory bowel disease. Clin Gastroenterol Hepatol. 2011;9:36-41.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 297]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
39.  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: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 224]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
40.  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: ]  [Cited by in Crossref: 341]  [Cited by in F6Publishing: 360]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
41.  Peyrin-Biroulet L, Khosrotehrani K, Carrat F, Bouvier AM, Chevaux JB, Simon T, Carbonnel F, Colombel JF, Dupas JL, Godeberge P. Increased risk for nonmelanoma skin cancers in patients who receive thiopurines for inflammatory bowel disease. Gastroenterology. 2011;141:1621-1628.e1-5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 333]  [Cited by in F6Publishing: 330]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
42.  Singh S, Nagpal SJ, Murad MH, Yadav S, Kane SV, Pardi DS, Talwalkar JA, Loftus EV. 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: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 130]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
43.  Gutierrez-Dalmau A, Campistol JM. Immunosuppressive therapy and malignancy in organ transplant recipients: a systematic review. Drugs. 2007;67:1167-1198.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Penn I. Kidney transplantation in patients previously treated for renal carcinomas. Transpl Int. 1993;6:350.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Axelrad J, Bernheim O, Colombel JF, Malerba S, Ananthakrishnan A, Yajnik V, Hoffman G, Agrawal M, Lukin D, Desai A. Risk of New or Recurrent Cancer in Patients With Inflammatory Bowel Disease and Previous Cancer Exposed to Immunosuppressive and Anti-Tumor Necrosis Factor Agents. Clin Gastroenterol Hepatol. 2016;14:58-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 74]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
46.  Dixon WG, Watson KD, Lunt M, Mercer LK, Hyrich KL, Symmons DP. Influence of anti-tumor necrosis factor therapy on cancer incidence in patients with rheumatoid arthritis who have had a prior malignancy: results from the British Society for Rheumatology Biologics Register. Arthritis Care Res (Hoboken). 2010;62:755-763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 132]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
47.  Strangfeld A, Hierse F, Rau R, Burmester GR, Krummel-Lorenz B, Demary W, Listing J, Zink A. Risk of incident or recurrent malignancies among patients with rheumatoid arthritis exposed to biologic therapy in the German biologics register RABBIT. Arthritis Res Ther. 2010;12:R5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 181]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
48.  Axelrad JE, Fowler SA, Friedman S, Ananthakrishnan AN, Yajnik V. Effects of cancer treatment on inflammatory bowel disease remission and reactivation. Clin Gastroenterol Hepatol. 2012;10:1021-1027.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
49.  Rajca S, Seksik P, Bourrier A, Sokol H, Nion-Larmurier I, Beaugerie L, Cosnes J. Impact of the diagnosis and treatment of cancer on the course of inflammatory bowel disease. J Crohns Colitis. 2014;8:819-824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
50.  Green S, Stock RG, Greenstein AJ. Rectal cancer and inflammatory bowel disease: natural history and implications for radiation therapy. Int J Radiat Oncol Biol Phys. 1999;44:835-840.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Naito A, Mizushima T, Takeyama H, Sakai D, Uemura M, Kudo T, Nishimura J, Shinzaki S, Hata T, Sato T. Feasibility of Chemotherapy in Patients with Inflammatory Bowel Disease-Related Gastrointestinal Cancer. Hepatogastroenterology. 2014;61:942-946.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Harrison ML, Obermueller E, Maisey NR, Hoare S, Edmonds K, Li NF, Chao D, Hall K, Lee C, Timotheadou E. Tumor necrosis factor alpha as a new target for renal cell carcinoma: two sequential phase II trials of infliximab at standard and high dose. J Clin Oncol. 2007;25:4542-4549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 183]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
53.  Jatoi A, Ritter HL, Dueck A, Nguyen PL, Nikcevich DA, Luyun RF, Mattar BI, Loprinzi CL. A placebo-controlled, double-blind trial of infliximab for cancer-associated weight loss in elderly and/or poor performance non-small cell lung cancer patients (N01C9). Lung Cancer. 2010;68:234-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 131]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
54.  Wiedenmann B, Malfertheiner P, Friess H, Ritch P, Arseneau J, Mantovani G, Caprioni F, Van Cutsem E, Richel D, DeWitte M. A multicenter, phase II study of infliximab plus gemcitabine in pancreatic cancer cachexia. J Support Oncol. 2008;6:18-25.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Hamaguchi T, Wakabayashi H, Matsumine A, Sudo A, Uchida A. TNF inhibitor suppresses bone metastasis in a breast cancer cell line. Biochem Biophys Res Commun. 2011;407:525-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
56.  Raaschou P, Simard JF, Neovius M, Askling J. Does cancer that occurs during or after anti-tumor necrosis factor therapy have a worse prognosis? A national assessment of overall and site-specific cancer survival in rheumatoid arthritis patients treated with biologic agents. Arthritis Rheum. 2011;63:1812-1822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
57.  Weber J. Ipilimumab: controversies in its development, utility and autoimmune adverse events. Cancer Immunol Immunother. 2009;58:823-830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 177]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
58.  Weber JS, Dummer R, de Pril V, Lebbé C, Hodi FS. Patterns of onset and resolution of immune-related adverse events of special interest with ipilimumab: detailed safety analysis from a phase 3 trial in patients with advanced melanoma. Cancer. 2013;119:1675-1682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 327]  [Cited by in F6Publishing: 333]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
59.  Johnson DB, Sullivan RJ, Ott PA, Carlino MS, Khushalani NI, Ye F, Guminski A, Puzanov I, Lawrence DP, Buchbinder EI. Ipilimumab Therapy in Patients With Advanced Melanoma and Preexisting Autoimmune Disorders. JAMA Oncol. 2016;2:234-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 419]  [Cited by in F6Publishing: 462]  [Article Influence: 57.8]  [Reference Citation Analysis (0)]
60.  Slesser AA, Bhangu A, Bower M, Goldin R, Tekkis PP. A systematic review of anal squamous cell carcinoma in inflammatory bowel disease. Surg Oncol. 2013;22:230-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
61.  Lopez A, Mounier M, Bouvier AM, Carrat F, Maynadié M, Beaugerie L, Peyrin-Biroulet L. Increased risk of acute myeloid leukemias and myelodysplastic syndromes in patients who received thiopurine treatment for inflammatory bowel disease. Clin Gastroenterol Hepatol. 2014;12:1324-1329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 72]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
62.  Bourrier A, Carrat F, Colombel JF, Bouvier AM, Abitbol V, Marteau P, Cosnes J, Simon T, Peyrin-Biroulet L, Beaugerie L. 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: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 92]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]