Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 7, 2021; 27(25): 3825-3836
Published online Jul 7, 2021. doi: 10.3748/wjg.v27.i25.3825
Autoimmune pancreatitis and pancreatic cancer: Epidemiological aspects and immunological considerations
Dimitri Poddighe, Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan
Dimitri Poddighe, Department of Pediatrics, National Research Institute for Maternal and Child Health (NRCMCH), University Medical Center (UMC) Nur-Sultan 010000, Kazakhstan
ORCID number: Dimitri Poddighe (0000-0001-6431-9334).
Author contributions: Poddighe D conceived and wrote this manuscript.
Conflict-of-interest statement: The author declares no conflict of interest.
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: Dimitri Poddighe, MD, MSc, PhD-Eq., Associate Professor, Department of Medicine, School of Medicine, Nazarbayev University, Kerei-Zhanibek Str. 5/1, Nur-Sultan 010000, Kazakhstan. dimitri.poddighe@nu.edu.kz
Received: February 7, 2021
Peer-review started: February 7, 2021
First decision: April 5, 2021
Revised: April 13, 2021
Accepted: May 20, 2021
Article in press: May 20, 2021
Published online: July 7, 2021

Abstract

Ordinary chronic pancreatitis is a well-known risk factor for pancreatic cancer, whereas such an association with autoimmune pancreatitis (AIP) is widely debated. Due to the rarity of the latter disorder, there are few specific clinical and epidemiological studies investigating the relation between AIP and pancreatic cancer, which do not seem to support it. However, these studies are affected by several limitations and, therefore, a link between AIP (and, specifically, type 1 AIP) and pancreatic cancer cannot be ruled out definitively on this basis. Moreover, several immunopathological aspects of type 1 AIP and, in general, immunoglobulin G4-related disease can create an immunological context that may impair the tumoral immunosurveillance and promote the pancreatic carcinogenesis and its progression. In detail, Th2 immunological dominance, type 2 macrophage polarization and basophil infiltration observed in type 1 AIP, may play a permissive role in creating a favorable immunological environment for pancreatic carcinogenesis, in addition to the immunosuppressive therapies that can be used in these patients.

Key Words: Autoimmune pancreatitis, Chronic pancreatitis, Pancreatic cancer, Immunoglobulin G4-related disease, Epidemiology, Immunology, Basophils, Macrophages, Th2 cells, Systemic lupus erythematosus

Core Tip: This mini-review discusses the debated issue of autoimmune pancreatitis (type 1) as a potential risk factor for pancreatic cancer. After summarizing the few available (low-quality) epidemiological evidence that does not clearly support this role, the immunopathological characteristics of type 1 autoimmune pancreatitis (including Th2 immunological dominance, type 2 macrophage polarization and basophil infiltration) are discussed as potential factors that may actually create a tolerogenic immunological environment favorable to pancreatic carcinogenesis and/or tumor progression.



INTRODUCTION
Autoimmune pancreatitis: Definition, pathology, and epidemiology

Chronic pancreatitis (CP) is a persistent inflammatory disease of the exocrine pancreas leading to progressive fibrotic tissue damage. CP prevalence varies between 13.5-98.7 cases per 100000 people with an incidence of around 4-5 new cases per 100000 people every year. Most cases of CP (also defined as “ordinary” or “generic”) are triggered by the repeated and/or persistent activation of intrapancreatic digestive enzymes, due to the variable combination of some environmental factors (e.g., excessive alcohol consumption, regular tobacco use, hypertriglyceridemia, etc.) and/or genetic factors (e.g., Chymotrypsin C mutations)[1].

Autoimmune Pancreatitis (AIP), which represents < 5%-10% of pancreatitis cases and has an estimated prevalence of approximately 1-2/100000 people, is much rarer[1,2]. However, the real incidence of AIP is currently unknown: indeed, it is diagnosed in around 2% of patients who have undergone pancreas surgical resection for presumed pancreatic cancer and, therefore, this condition may be underdiagnosed[3].

The term AIP was originally introduced by Yoshida et al[4] in 1995 to describe a patient diagnosed with pancreatitis who “had hyperglobulinemia, was autoantibody-positive, and responded to steroid therapy”. Currently, AIP diagnosis must be supported by well-defined and specific clinical, radiological, serological and histopathological criteria[3]. Importantly, based on histopathological aspects (according to the Honolulu classification), AIP is classified into two main subtypes that are mainly distinguished by the absence (type 1 AIP) or presence (type 2 AIP) of one peculiar finding, called “granulocytic epithelial lesion”(GEL), consisting of the neutrophilic infiltration of medium/small-sized ducts and often acini[5]. Thus, type 1 AIP is characterized by a substantial lymphoplasmacytic infiltration, where eosinophils may be relatively frequent and neutrophils are rare. Indeed, it was originally described as lymphoplasmacytic sclerosing pancreatitis[5,6]. Moreover, it is much more frequent than type 2 AIP, which accounts for 10%-20% and no more than 5% of all AIP cases in Western and Eastern countries, respectively. Importantly, type 1 AIP is now accepted as a clinical-pathological aspect of immunoglobulin G4 (IgG4)-related disease (IgG4-RD)[6,7].

IgG4-RD is a multisystem immune-mediated fibroinflammatory condition where several organs can be involved and, most frequently, pancreas, bile ducts, salivary glands, lacrimal glands, kidneys, retroperitoneum, and lungs. Pancreas is involved in at least 45% of IgG4-RD patients as type 1 AIP[8]. Therefore, unlike ordinary CP, type 1 AIP is usually the pancreatic manifestation of a systemic immunological disorder, in which several alterations of the innate and acquired immune system contribute to the lympho-plasmocytic inflammatory infiltration of the pancreas[9].

IgG4-RD pathogenesis is complex and involves both the innate and adaptive immune system; as regards the latter one, Th2 (specifically, follicular Th2 cells, Tfh2) and Treg lymphocytes (and related cytokines) have been clearly and mainly implicated in this pathologic process characterized by altered B cell and plasma cell activation, enhancement of IgG4 class switch recombination and, finally, development of ectopic germinal centers and induction of fibrosis[9,10].

Type 2 AIP pathogenesis is more elusive and, indeed, is also called idiopathic duct-centric pancreatitis. It is not characterized by IgG4 increase and other specific extra-pancreatic manifestations, except for an association with inflammatory bowel diseases in 20%-30% cases[6,7].

Type 1 and type 2 AIP cannot be certainly differentiated by radiological imaging and share similar clinical presentations. However, the diagnostic differentiation is very important since the prognosis is quite different. Indeed, type 1 AIP is usually more aggressive, showing a much higher rate of relapse after steroid therapy, and is characterized by synchronous or metachronous extra-pancreatic organs involvement, as previously mentioned[11].

Both AIP types are almost exclusively adulthood diseases with a median age at diagnosis between 40-50 years, even though patients with type 1 AIP are 10-15 years older on average. AIP has been rarely described in the pediatric population, where the few reported cases were consistent with type 2 AIP in most cases[7,12].

PANCREATIC CANCER AND AIP: EPIDEMIOLOGICAL ASPECTS

Pancreatic cancer is represented by adenocarcinoma in > 85% of cases. Nowadays the overall incidence of pancreatic cancer is estimated to be around 11.0 per 100,000 people per year in the United States, where it accounts for 3% of all diagnosed malignancies. These numbers make it be the twelfth and eleventh most common cancer in men and women, respectively. Pancreatic cancer remains a highly fatal malignancy: Worldwide, it is the seventh leading cause of cancer death in both genders, with a 5-year survival rate of 10% or less. Like most malignancies, incidence rates for pancreatic cancer are age-related: In general, it is rare before the age of 50 years. The main risk factors for pancreatic cancer are modifiable and include tobacco smoking, obesity, physical inactivity and high-calorie/fat diets; these lifestyle-related risk factors clearly contributed to the increase of the disease incidence in the last three decades, especially in developed countries, where diagnostic improvements and increased life expectancy have played a role as well[12,13].

CP (and, in detail, ordinary CP) was shown to be a clear risk factor for pancreatic cancer. In 1993 Lowenfels et al[14] published the results of a large multicenter and international cohort study including 2,015 patients with CP, who showed an increased risk of pancreatic cancer, independent from gender, country and type of pancreatitis. It was estimated that approximately 5% of CP patients receive a diagnosis of pancreatic cancer within 2 decades after that diagnosis[12,14].

The potential association between pancreatic cancer and AIP is currently debated. Even though AIP is a rare form of CP, this issue is raised especially in patients affected with type 1 AIP that, compared to type 2 AIP, is more common and characterized by high percentages of disease relapse after steroid therapy[15,16].

Moreover, as summarized and discussed below, all reported cases of AIP-related pancreatic cancer were described in patients affected with type 1 AIP; actually, some concerns were raised in terms of potential association with malignancies in general. Most studies on IgG4-RD and/or type 1 AIP are from Japan: Yamamoto et al[17] and Shiokawa et al[18] first reported an overall increase of malignancy incidence in their patients, even though they actually described no cases of pancreatic cancer.

A larger multi-centric and international study by Hart et al[15] (including 1,064 patients with AIP and, exactly, 978 with type 1 and 86 with type 2) reported 5 diagnoses of pancreatic cancer in type 1 AIP patients (no one affected with type 2 AIP developed this complication). However, as discussed by the authors themselves, even though they found a small number of pancreatic cancers (compared to the total cumulative frequency of malignancies, n = 57), the limited follow-up and lack of a control population may have limited the clinical significance of their analysis. Moreover, this study (and also another one authored by the same research group) were not focused on pancreatic cancer[15,19], as well as the following studies. Hirano et al[20] described their case series of 113 IgG4-RD patients (type 1 AIP: n = 95) and reported 2 patients who developed pancreatic cancer; in general, their conclusion was that the cumulative incidence of any kind of malignancies in IgG4-RD patients was similar to that observed in the general population. Shimizu et al[21] also made the same conclusion in their 84 type 1 AIP patients; in detail, among 9 patients diagnosed with cancer, only one developed it at the pancreas. Buijs et al[22] analyzed a Dutch case series of 107 patients affected with AIP (type 1: 90%, type 2: 10%), and reported no apparent difference in malignancy incidence compared to an ethnic-, age-, and sex-matched reference population (in detail, no patients developed pancreatic cancer).

However, although several clinical studies tried to address this issue, the prevalence and risk assessment of pancreatic cancer in the pathological setting of AIP have been greatly hampered by the relatively small number of participants, variable and/or relatively short follow-up period, retrospective study design and the lack of appropriate control groups in most of these studies[23].

Therefore, some authors proposed that patients affected with AIP should be regularly monitored to reveal any potential cancerous changes and/or malignancy onset, anyway[24]. For instance, unlike the aforementioned studies, Huggett et al[16] reported a statistically significant odd ratio increase (odds ratio = 2.2) for all malignancy risk in type 1 AIP patients: 13 malignancy cases (out of 115 type 1 AIP and/or IgG4-RD and, 106 patients with pancreatic disease) were diagnosed, and all those tumors arose in systemic IgG4-RD patients, who represented only 56% of the whole cohort. However, only one case was affected with pancreatic adenocarcinoma.

Additional and interesting research articles, which were more focused on pancreatic complications and carcinogenesis in AIP patients, were published in the last few years. A clinical research by Gupta et al[25] investigated the pancreatic carcinogenesis in patients affected with AIP. These authors reviewed a case series of 84 AIP patients, and they compared the prevalence of pancreatic intraepithelial neoplasia (PanIN) in 28 cases of AIP and 30 cases of CP not otherwise specified. Overall, 82% AIP patients showed PanIN with variable histologic grade, which resulted to be more frequent than in non-autoimmune CP (63%), though such a difference was not statistically significant. Therefore, the prevalence of PanIN in AIP resulted to be comparable to ordinary CP; moreover, in the same article the authors also described their clinical experience with 84 AIP patients, and the only 2 cases of pancreatic cancer were diagnosed in the context of type 1 AIP, which was diagnosed 6 and 10 years before the detection of the malignancy, respectively. Therefore, this study raised some specific concerns as regards the risk of pancreatic malignancy in type 1 AIP patients[25]. Similarly, Ikeura et al[26] diagnosed 3 cases of pancreatic cancer (4.8%) in their 63 type 1 AIP patients, which was similar to what observed in the comparison group, consisting of 41 patients affected with ordinary (alcoholic and hereditary) CP, characterized by only one patient (2.1%) with a diagnosis of pancreatic malignancy.

Starting from a different perspective, Ngwa et al[27] described 548 patients diagnosed with pancreatic cancer and 99 different patients affected with type 1 AIP. In this study, whose main aim was to compare the IgG4 serum profiles between those two groups of patients (rather than investigating an epidemiological and/or causal link), the authors suggested no relationship between AIP and pancreatic cancer; however, a minority of histological specimens (only 30 pancreatic tissue specimens from patients with pancreatic cancer and 6 patients with “high-risk features” AIP) was reviewed. Indeed, a recent retrospective study by Xiang et al[28], including 74 patients with type 1 AIP, revived the debate by reporting that 5 of them (6.7%) were concomitantly diagnosed with a pancreatic tumor (pancreatic ductal adenocarcinoma, n = 3; solitary extramedullary plasmacytoma, n = 1; cholangiocarcinoma, n = 1). Moreover, an interesting observation was reported by Hedayat et al[29], who re-examined 21 pancreas specimens of previously diagnoses of intraductal papillary-mucinous neoplasm, which is a cystic and usually benign neoplasm, but with the potential for progression to pancreatic cancer. Interestingly, 4 of them (19%) showed infiltrates of IgG4-positive plasma cells, consistent with a “peri-tumoral” type 1 AIP reaction[29,30].

Conversely, two very recent retrospective studies were not supportive in this sense. Tang et al[31] described 17 neoplastic cases in a large cohort of 587 Chinese patients diagnosed with IgG4-RD; among those, 11 were also affected with AIP, but all developed extra-pancreatic malignancies only. The study by Ishikawa et al[32], including 123 type 1 AIP patients, identified only 2 patients diagnosed with pancreatic cancer (1.6%) and concluded that “AIP is unlikely to be a precancerous condition of the pancreas”, but at the same time they stated that “because of the small number of cases, the characteristic findings of pancreatic cancers that develop in AIP patients are not clear”.

Therefore, as summarized by these two final statements, this short and schematic overview of clinical studies provides conflicting evidence and conclusions regarding the association between AIP (and, in detail, type 1) and pancreatic cancer. Table 1 summarizes all the aforementioned clinical studies investigating the association between AIP and pancreatic cancer, by using a chronological order and focusing on the main study features and findings.

Table 1 Schematic overview of the clinical studies investigating the association between autoimmune pancreatitis and pancreatic cancer.
Ref.
Study population (disease)
Total patients (n)
AIP type 1 (n)       
Cancer (overall) (n)
Pancreatic cancer (n)
Median follow-up (yr)              
Additional specifications
Yamamoto et al[17] (Japan, 2012)IgG4-RD1061020NA-
Shiokawa et al[18] (Japan, 2013)AIP1081041803.3These 18 malignancies were diagnosed in 15 patients.
Hart et al[15] (International, 2013)AIP1064978575NANo patients with AIP type 2 developed any malignancies.
Gupta et al[25] (United States, 2013)CP5811NA7 (PanIN)N/ARetrospective analysis of patients with CP. There was no statistically significant difference in the frequency of PanIN between ordinary CP and AIP: in the latter group, no difference between type 1 and type 2. In general, the only case of PanIN3 was detected in one AIP type 1 specimen.
Gupta et al[25] (United States, 2013)AIP84NANA24.1These 2 cases of pancreatic cancer were diagnosed in type 1 AIP patients.
Hart et al[19] (United States, 2014)AIP1161162313.6-
Hirano et al[20] (Japan, 2014)IgG4-RD1139514262 IgG4-RD patients diagnosed with malignancy (out of 14) were not affected with AIP.
Huggett et al[16] (United Kingdom, 2014)IgG4-RD1151061332.7Of these 3 cases of pancreatic malignancies, 2 were cholangiocarcinoma cases and 1 was a pancreatic adenocarcinoma.
Ikeura et al[26] (Japan, 2014)AIP6363NA35.2-
Shimizu et al[21] (Japan, 2015)AIP8484914.5-
Buijs et al[22] (The Netherlands, 2015)AIP10796806.25-
Ngwa et al[27] (United States, 2015)AIP9999NA0NAThe aim of this study was to evaluate the clinical significance of elevated sgG4 levels in patients with AIP and pancreatic cancer and potential prognostic implications of those in patients with pancreatic cancer.
Xiang et al[28] (China, 2019)AIP7474NA4NA3 patients were diagnosed with pancreatic ductal adenocarcinoma and 1 with cholangiocarcinoma. There was also 1 case of solitary extramedullary plasmacytoma.
Tang et al[31] (China, 2020)IgG4-RD587NA1705.1-
Ishikawa et al[32] (Japan, 2020)AIP123123NA24.6-

Even though one may conclude that most of the available studies do not support this association, there are some clinical and pathological observations coming from studies more focused on pancreatic cancer[25,26,28] which should keep high the attention on this issue before making final conclusions; however, this will require larger, prospective, and longer (in terms of follow-up period) studies. Unfortunately, the clinical research on this topic is undoubtedly hampered by the low prevalence and incidence of AIP, in general and in the landscape of CP, in addition to the challenges of pancreatic diagnostics.

Nonetheless, it is established that chronic inflammatory processes represent a risk factor for pancreatic cancer and, as such, type 1 AIP, especially if relapsing and/or persistent, should be considered in the same way in principle. Moreover, concomitant immunosuppressive therapies (including steroids, rituximab, azathioprine., etc.) may represent additional predisposing factors to the cancer development[11]. Finally, some specific immunological aspects and considerations (also related to IgG4-RD pathogenesis) might support these concerns, as discussed in the next sections.

TYPE 1 AIP: GENERAL IMMUNOPATHOGENESIS

IgG4-RD is the clinical expression of a systemic immunological dysregulation leading to chronic inflammation and lymphocyte infiltration in several organs (including pancreas), which might have some implications in terms of pancreatic and extra-pancreatic carcinogenesis[8]. However, as discussed, the available clinical evidence does not support this hypothesis, but the small number of studies, which are also characterized by several and important limitations, likewise do not allow to rule out it either.

As said, type 1 AIP is the prevalent form of AIP and all cases of AIP-related pancreatic malignancies were described in this pathological setting. Therefore, the following immunological considerations on AIP immunologic pathogenesis and its potential role in pancreatic carcinogenesis, will refer to type 1 AIP and/or IgG4-RD specifically.

Even though the pathogenesis of IgG4-RD is complex and relies on both innate and acquired immune system mechanisms, the final result is an immunological environment characterized by a Th2 dominant immune response, which is indeed associated with increased level of serum IgG4 (systemically) and IgG4 switched lymphocytes and plasma cells (in the pancreas and other affected organs). Therefore, IgG4-RD and type 1 AIP are characterized by a cytokine profile whereby interleukin (IL)-4, IL-5, IL-13, IL-10 and transforming growth factor (TGF)-β are overexpressed[33].

Recent studies evidenced the central role that is played by the T follicular helper cells (Tfh cells), which can be found in the extra-nodal ectopic germinal centers and are involved in the generation of long-lasting humoral responses by B cells and plasma cells[9]. Increased levels of Tfh cells and, in detail, Th2-polarized cells (Tfh2 cells), have been described in IgG4-RD patients, in whom those cells correlate with serum IgG4 and IL-4 levels[34]. Moreover, Tfh2 cells, but not Tfh1 or Tfh17 cells, were demonstrated to induce the differentiation of naïve B cells into plasma blasts (with enhanced production of IgG4) and, importantly, the activation of Tfh2 cells resulted to correlate with the disease activity[35].

However, in addition to the probable intrinsic dysregulation of the adaptive immune system, the origin of these Tfh2 expansion and activation (and, more in general, Th2 predominance) must be sought in the innate immune system as well[9]. Through the use of an experimental murine model of AIP (namely, MRL/Mp mice treated with polyinosinic-polycytidylic acid), Arai et al[36] described the pancreatic accumulation of plasmocytoid dendritic cells (pDCs) producing IFN-α. They also showed that pDCs from human patients with type 1 AIP had an increased production of IFN-α, which was able to promote the B cells switch toward IgG4 production in a T-cell independent manner, probably by the enhanced production of BAFF (B cell-activating factor belonging to the tumor necrosis factor family). Actually, BAFF production upon Toll-like-receptor (TLR) activation was described in other innate immune cells, including basophils, through the stimulation of TLR2 and TLR4[37]. This finding is particularly interesting because the presence of basophils has been recently described in the pancreatic tissue of most patients with type 1 AIP[38].

Basophils have been demonstrated to be more than simple effector cells in several pathological contexts (including asthma and other allergic diseases) and, in detail, were proposed as one of the main sources of early IL-4, being able to drive and/or support the Th2 polarization of activated CD4+ T cells[39-41]. Overall, these observations may support the fact that basophils are activated in a IgE- (and, thus, B-cell) independent manner and, indeed, TLR-activated basophils can contribute to drive the Th2 response in type 1 AIP[38].

Interestingly, in a recent review article, Watanabe et al[9] highlighted several immunological similarities between IgG4-RD and Systemic Lupus Erythematosus (SLE), which is also characterized by an increased type I IFN production through pDCs, a Th2 immunological dominance and a substantial dysregulation of humoral immunity. Moreover, some human studies and murine experimental models suggested a pathogenic role for basophils in SLE, indeed[42].

However, in the landscape of all the innate immune cells implicated in the pathogenesis of type 1 AIP, the role of pDCs appears to be prominent. In addition to IFN-α, these cells resulted to be the main source of another important cytokine that has been implicated in type 1 AIP immunopathogenesis, namely IL-33. Importantly, IL-33 was also recognized to be an important inflammatory mediator in ordinary CP, whereby actually the main cell source is represented by the pancreatic acinar cells[9,43]. IL-33 is able to activate Th2 cells and also group 2 innate lymphoid cells, which can further stimulate the production of IL-4, IL-5, and IL-13[44,45].

Finally, to complete the landscape of the innate immunity involvement in type 1 AIP, it is worth to mention M2-polarized macrophages, which were described in the pancreatic tissues of type 1 AIP patients affected with IgG4-RD[46]. Compared to classically activated or M1 macrophages (which are clearly pro-inflammatory, are polarized by lipopolysaccharide and/or Th1 cytokines, and mainly produce IL-1β, IL-6, IL-12, IL-23 and tumor necrosis factor-α), alternatively activated or M2 macrophages are polarized by Th2 cytokines and produce anti-inflammatory cytokines, such as IL-10 and TGF-β. Therefore, M2 macrophages have been implicated in angiogenesis and tissue repair, and are considered to exert an anti-inflammatory (and, for some aspects, tolerogenic effect) also in IgG4-RD and type 1 AIP[47,48].

Indeed, IL-10 and TGF-β are the main cytokines secreted by the antigen-induced or adaptive Tregs. In detail, IL-10 regulates the functions of many different immune cells, including macrophages themselves, dendritic cells, and both T and B lymphocytes. In detail, IL-10 is a potent inhibitor of antigen presentation by reducing the expression of major histocompatibility complex molecules and costimulatory molecules CD80 and CD86. Moreover, it inhibits the differentiation of dendritic cells (DCs) themselves from monocyte precursors and their further maturation. Finally, upon antigen presentation, IL-10 inhibits macrophage production of pro-inflammatory cytokines (IL-1, IL-6, IL-12, tumor necrosis factor-α), even though it does not seem to directly affect the Th1/Th2 balance[49,50].

These immune-pathogenic aspects of type 1 AIP and IgG4-RD may create a favorable immunological background in the affected pancreas for carcinogenesis promotion, as discussed in the next section.

PANCREATIC CANCER AND TYPE 1 AIP: IMMUNOLOGICAL CONSIDERATIONS

As already mentioned, the cumulative risk of pancreatic cancer in subjects with ordinary CP was reported to be 1.8% after 10 years and 4.0% after 20 years, and it is estimated to be more than 10 times higher in these patients than in healthy subjects[14,51].

Whereas carcinogenesis in the context of ordinary CP is then well established epidemiologically, this methodological approach has not clearly supported the association between AIP (in detail, type 1 AIP) and pancreatic cancer, but the numerous study limitations should prevent from ruling it out definitively, as it was previously discussed.

To start with, a few initial molecular findings should be considered. Some specific molecular alterations of pancreatic carcinogenesis may be functional to this discussion, such as K-ras mutations, which resulted to be present in > 90% of pancreatic cancers and, importantly, may occur at an early stage of this neoplastic process, namely at the PanIN stage[52]. For instance, Kamisawa et al[53] found that codon-12 mutation of K-ras was significantly more frequent in the pancreato-biliary regions of patients with AIP than in those affected with chronic alcoholic pancreatitis.

More recently, Kinugawa et al[54] also described some methylation abnormalities of tumor suppressor genes in AIP patients. Promoter region hyper-methylation with consequent gene silencing was reported for several genes in pancreatic cancer. In detail, among the 6 genes that these authors have investigated (because those were previously reported as methylated in pancreatic cancer), they found a statistically significant difference in terms of the TFPI2 (tissue factor pathway inhibitor 2) methylation ratio between specimens taken from AIP patients and those from pancreas resected for non-tumoral diseases. TFPI2 was recognized as a tumor suppressor gene, and a previous study described methylation abnormalities for this gene in 73% of pancreatic carcinomas, whereas those were completely absent in normal pancreas specimens[55].

However, further theoretical support and biological plausibility supporting a role for type 1 AIP as potential risk factors for pancreatic cancer, may derive from some immunological considerations.

Indeed, the role of the immune system and, in detail, the impairment of the immunological surveillance in the development and progression of cancer, is well supported. Both the innate and the adaptive immune system play an active role in this regard. Generally speaking, cytotoxic CD8+ T cells, Th1 cells, mature DCs, classically activated pro-inflammatory macrophages M1 and natural killer cells are variably described as the main effectors of this anti-cancer immunological surveillance[56,57]. Therefore, several aspects of the immunological environment created by and during type 1 AIP (and, systemically, by IgG4-RD) may be favorable to cancer development in the pancreas.

First, the Th2 signature dominating these diseases, does not support cytotoxic CD8+ T cell responses[56]. Tassi et al[58] showed that patients affected with pancreatic cancer have a Th2 skewed immune response, which can impair the specific CD4+ T cells response against a tumor-associated antigen, namely CEA; moreover, this polarization was suggested to negatively affect the T CD8+ cell compartment as well.

Second, in type 1 AIP macrophages tend to switch to a M2 phenotype, characterized by the production of IL-10 and TGF-β, which can create a tolerogenic immunological environment in the pancreas. By the way, Th2 cells themselves produce IL-10, in addition to their hallmark cytokines, such as IL-4, IL-5 and IL-13[47]. As explained, these M2 macrophages are also characterized by poor antigen presenting ability rather than supporting lymphocyte activation, proliferation and cytotoxicity. Moreover, they are committed to promote tissue remodeling and, importantly, neo-angiogenesis. Finally, M2 macrophages also express inhibitory ligands, such as PD-L1, which is known to induce peripheral T cell tolerance. All these properties suggested an important role of M2 macrophage in reducing the anti-tumor immunity for several malignancies, including pancreatic cancer. Indeed, they represent the prevalent macrophage phenotype among so-called tumor-associated macrophages and have been thus implicated in the processes of tumoral growth and cell migration/metastasis[59-62]. Accordingly, the degree and type of M2 macrophages infiltration into the tumoral stroma resulted to be an independent prognostic factor in patients with pancreatic cancer[63].

Third, in addition to M2 macrophages, basophils were recently suggested to play a role in the pathophysiology of type 1 AIP, upon activation via TLR signaling[38,64]. As previously explained, these cells can strongly shift the immunological balance toward a more tumor-tolerogenic Th2-polarized tissue microenvironment. Therefore, in addition to some initial evidence that basophils may be directly implicated in type 1 AIP pathogenesis, this way basophils might indirectly promote the pancreatic tumorigenesis as well. Recently, De Monte et al[65] investigated the presence of basophils in in tumor-draining lymph nodes of patients affected with pancreatic cancer, which resulted to correlate with both Th2 inflammation and reduced patients’ survival. In this study, the role of basophils in tumor development/progression was also supported through some observations in basophil-deficient murine experimental models. Therefore, basophils may favor the tumorigenesis and progression of pancreatic cancer by promoting both Th2 and M2 polarization[66].

Interestingly, these novel findings might open to additional important associations between pancreatic cancer and autoimmune diseases other than type 1 AIP and IgG4-RD. As mentioned, Watanabe et al[9] provided an interesting immunopathological parallelism between IgG4-RD and SLE. Therefore, it is very interesting that a very recent meta-analysis by Seo et al[67] evidenced that SLE was associated with increased risk for pancreatic cancer. These authors speculated several mechanisms that may potentially explain this association (e.g., chronic inflammation, excess autoantibody effect, metabolic alterations); however, based on the present discussion, basophils activation and, more in general, Th2-driven inflammation described in active SLE might be added to these speculations on the hypothetical pathophysiologic permissive mechanisms[68,69].

CONCLUSION

Currently, the association between (type 1) AIP and pancreatic cancer does not find any clear epidemiological support, even though it cannot be ruled out definitively due to the small number of participants in the available clinical studies and the numerous study limitations. Some immunological aspects characterizing type 1 AIP, such as Th2 dominance, M2 macrophage polarization and basophil implication, may lead to an immunological environment favorable to pancreatic carcinogenesis and/or tumor progression.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Kazakhstan

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: Kishida D S-Editor: Gao CC L-Editor: Filipodia P-Editor: Ma YJ

References
1.  Beyer G, Habtezion A, Werner J, Lerch MM, Mayerle J. Chronic pancreatitis. Lancet. 2020;396:499-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 203]  [Article Influence: 50.8]  [Reference Citation Analysis (1)]
2.  Uchida K, Masamune A, Shimosegawa T, Okazaki K. Prevalence of IgG4-Related Disease in Japan Based on Nationwide Survey in 2009. Int J Rheumatol. 2012;2012:358371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 103]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
3.  Madhani K, Farrell JJ. Autoimmune Pancreatitis: An Update on Diagnosis and Management. Gastroenterol Clin North Am. 2016;45:29-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
4.  Yoshida K, Toki F, Takeuchi T, Watanabe S, Shiratori K, Hayashi N. Chronic pancreatitis caused by an autoimmune abnormality. Proposal of the concept of autoimmune pancreatitis. Dig Dis Sci. 1995;40:1561-1568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1044]  [Cited by in F6Publishing: 891]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
5.  Chari ST, Kloeppel G, Zhang L, Notohara K, Lerch MM, Shimosegawa T;  Autoimmune Pancreatitis International Cooperative Study Group (APICS). Histopathologic and clinical subtypes of autoimmune pancreatitis: the Honolulu consensus document. Pancreas. 2010;39:549-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 173]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
6.  Blaho M, Dítě P, Kunovský L, Kianička B. Autoimmune pancreatitis - An ongoing challenge. Adv Med Sci. 2020;65:403-408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
7.  de Pretis N, Frulloni L. Autoimmune pancreatitis type 2. Curr Opin Gastroenterol. 2020;36:417-420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
8.  Vashi B, Khosroshahi A. IgG4-Related Disease with Emphasis on Its Gastrointestinal Manifestation. Gastroenterol Clin North Am. 2019;48:291-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
9.  Watanabe T, Minaga K, Kamata K, Kudo M, Strober W. Mechanistic Insights into Autoimmune Pancreatitis and IgG4-Related Disease. Trends Immunol. 2018;39:874-889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 47]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
10.  Tsuboi H, Honda F, Takahashi H, Ono Y, Abe S, Kondo Y, Matsumoto I, Sumida T. Pathogenesis of IgG4-related disease. Comparison with Sjögren's syndrome. Mod Rheumatol. 2020;30:7-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
11.  Rebours V, Lévy P. Pancreatic and biliary tract involvement in IgG4-related disease. Presse Med. 2020;49:104015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
12.  Lee HM, Deheragoda M, Harrison P, Devlin J, Sellars M, Hadzic N, Dhawan A, Grammatikopoulos T. Autoimmune pancreatitis in children: A single centre experience in diagnosis, management and long term follow up. Pancreatology. 2019;19:169-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
13.  Khalaf N, El-Serag HB, Abrams HR, Thrift AP. Burden of Pancreatic Cancer: From Epidemiology to Practice. Clin Gastroenterol Hepatol. 2021;19:876-884.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 139]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
14.  Lowenfels AB, Maisonneuve P, Cavallini G, Ammann RW, Lankisch PG, Andersen JR, Dimagno EP, Andrén-Sandberg A, Domellöf L. Pancreatitis and the risk of pancreatic cancer. International Pancreatitis Study Group. N Engl J Med. 1993;328:1433-1437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1255]  [Cited by in F6Publishing: 1106]  [Article Influence: 35.7]  [Reference Citation Analysis (0)]
15.  Hart PA, Kamisawa T, Brugge WR, Chung JB, Culver EL, Czakó L, Frulloni L, Go VL, Gress TM, Kim MH, Kawa S, Lee KT, Lerch MM, Liao WC, Löhr M, Okazaki K, Ryu JK, Schleinitz N, Shimizu K, Shimosegawa T, Soetikno R, Webster G, Yadav D, Zen Y, Chari ST. Long-term outcomes of autoimmune pancreatitis: a multicentre, international analysis. Gut. 2013;62:1771-1776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 382]  [Cited by in F6Publishing: 337]  [Article Influence: 30.6]  [Reference Citation Analysis (0)]
16.  Huggett MT, Culver EL, Kumar M, Hurst JM, Rodriguez-Justo M, Chapman MH, Johnson GJ, Pereira SP, Chapman RW, Webster GJM, Barnes E. Type 1 autoimmune pancreatitis and IgG4-related sclerosing cholangitis is associated with extrapancreatic organ failure, malignancy, and mortality in a prospective UK cohort. Am J Gastroenterol. 2014;109:1675-1683.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 157]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
17.  Yamamoto M, Takahashi H, Tabeya T, Suzuki C, Naishiro Y, Ishigami K, Yajima H, Shimizu Y, Obara M, Yamamoto H, Himi T, Imai K, Shinomura Y. Risk of malignancies in IgG4-related disease. Mod Rheumatol. 2012;22:414-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 100]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
18.  Shiokawa M, Kodama Y, Yoshimura K, Kawanami C, Mimura J, Yamashita Y, Asada M, Kikuyama M, Okabe Y, Inokuma T, Ohana M, Kokuryu H, Takeda K, Tsuji Y, Minami R, Sakuma Y, Kuriyama K, Ota Y, Tanabe W, Maruno T, Kurita A, Sawai Y, Uza N, Watanabe T, Haga H, Chiba T. Risk of cancer in patients with autoimmune pancreatitis. Am J Gastroenterol. 2013;108:610-617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 158]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
19.  Hart PA, Law RJ, Dierkhising RA, Smyrk TC, Takahashi N, Chari ST. Risk of cancer in autoimmune pancreatitis: a case-control study and review of the literature. Pancreas. 2014;43:417-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 62]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
20.  Hirano K, Tada M, Sasahira N, Isayama H, Mizuno S, Takagi K, Watanabe T, Saito T, Kawahata S, Uchino R, Hamada T, Miyabayashi K, Mohri D, Sasaki T, Kogure H, Yamamoto N, Nakai Y, Yoshida H, Ito Y, Akiyama D, Toda N, Arizumi T, Yagioka H, Takahara N, Matsubara S, Yashima Y, Koike K. Incidence of malignancies in patients with IgG4-related disease. Intern Med. 2014;53:171-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 109]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
21.  Shimizu S, Naitoh I, Nakazawa T, Hayashi K, Miyabe K, Kondo H, Nishi Y, Yoshida M, Umemura S, Hori Y, Kato A, Okumura F, Sano H, Hirata Y, Takada H, Ohara H, Joh T. Correlation between long-term outcome and steroid therapy in type 1 autoimmune pancreatitis: relapse, malignancy and side effect of steroid. Scand J Gastroenterol. 2015;50:1411-1418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
22.  Buijs J, Cahen DL, van Heerde MJ, Rauws EA, de Buy Wenniger LJ, Hansen BE, Biermann K, Verheij J, Vleggaar FP, Brink MA, Beuers UH, van Buuren HR, Bruno MJ. The Long-Term Impact of Autoimmune Pancreatitis on Pancreatic Function, Quality of Life, and Life Expectancy. Pancreas. 2015;44:1065-1071.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
23.  Ikeura T, Miyoshi H, Shimatani M, Uchida K, Takaoka M, Okazaki K. Long-term outcomes of autoimmune pancreatitis. World J Gastroenterol. 2016;22:7760-7766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 17]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
24.  Bojková M, Dítě P, Dvořáčková J, Novotný I, Floreánová K, Kianička B, Uvírová M, Martínek A. Immunoglobulin G4, autoimmune pancreatitis and pancreatic cancer. Dig Dis. 2015;33:86-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
25.  Gupta R, Khosroshahi A, Shinagare S, Fernandez C, Ferrone C, Lauwers GY, Stone JH, Deshpande V. Does autoimmune pancreatitis increase the risk of pancreatic carcinoma? Pancreas. 2013;42:506-510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 63]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
26.  Ikeura T, Miyoshi H, Uchida K, Fukui T, Shimatani M, Fukui Y, Sumimoto K, Matsushita M, Takaoka M, Okazaki K. Relationship between autoimmune pancreatitis and pancreatic cancer: a single-center experience. Pancreatology. 2014;14:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 58]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
27.  Ngwa T, Law R, Hart P, Smyrk TC, Chari ST. Serum IgG4 elevation in pancreatic cancer: diagnostic and prognostic significance and association with autoimmune pancreatitis. Pancreas. 2015;44:557-560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 29]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
28.  Xiang P, Zhang X, Wang C, Lang Y, Xu L, Huang L, Shen J, Feng ST. Pancreatic tumor in type 1 autoimmune pancreatitis: a diagnostic challenge. BMC Cancer. 2019;19:814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
29.  Hedayat AA, Lisovsky M, Suriawinata AA, Longnecker DS. Association of IgG4 response and autoimmune pancreatitis with intraductal papillary-mucinous neoplasms. Pancreatology. 2017;17:263-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
30.  Morales-Oyarvide V, Fong ZV, Fernández-Del Castillo C, Warshaw AL. Intraductal Papillary Mucinous Neoplasms of the Pancreas: Strategic Considerations. Visc Med. 2017;33:466-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
31.  Tang H, Yang H, Zhang P, Wu D, Zhang S, Zhao J, Peng L, Chen H, Fei Y, Zhang X, Zhao Y, Zeng X, Zhang F, Zhang W. Malignancy and IgG4-related disease: the incidence, related factors and prognosis from a prospective cohort study in China. Sci Rep. 2020;10:4910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
32.  Ishikawa T, Kawashima H, Ohno E, Iida T, Suzuki H, Uetsuki K, Yamada K, Yashika J, Yoshikawa M, Gibo N, Aoki T, Kataoka K, Mori H, Fujishiro M. Risks and characteristics of pancreatic cancer and pancreatic relapse in autoimmune pancreatitis patients. J Gastroenterol Hepatol. 2020;35:2281-2288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
33.  Islam AD, Selmi C, Datta-Mitra A, Sonu R, Chen M, Gershwin ME, Raychaudhuri SP. The changing faces of IgG4-related disease: Clinical manifestations and pathogenesis. Autoimmun Rev. 2015;14:914-922.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
34.  Akiyama M, Suzuki K, Yamaoka K, Yasuoka H, Takeshita M, Kaneko Y, Kondo H, Kassai Y, Miyazaki T, Morita R, Yoshimura A, Takeuchi T. Number of Circulating Follicular Helper 2 T Cells Correlates With IgG4 and Interleukin-4 Levels and Plasmablast Numbers in IgG4-Related Disease. Arthritis Rheumatol. 2015;67:2476-2481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 148]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
35.  Akiyama M, Yasuoka H, Yamaoka K, Suzuki K, Kaneko Y, Kondo H, Kassai Y, Koga K, Miyazaki T, Morita R, Yoshimura A, Takeuchi T. Enhanced IgG4 production by follicular helper 2 T cells and the involvement of follicular helper 1 T cells in the pathogenesis of IgG4-related disease. Arthritis Res Ther. 2016;18:167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 124]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
36.  Arai Y, Yamashita K, Kuriyama K, Shiokawa M, Kodama Y, Sakurai T, Mizugishi K, Uchida K, Kadowaki N, Takaori-Kondo A, Kudo M, Okazaki K, Strober W, Chiba T, Watanabe T. Plasmacytoid Dendritic Cell Activation and IFN-α Production Are Prominent Features of Murine Autoimmune Pancreatitis and Human IgG4-Related Autoimmune Pancreatitis. J Immunol. 2015;195:3033-3044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
37.  Watanabe T, Yamashita K, Sakurai T, Kudo M, Shiokawa M, Uza N, Kodama Y, Uchida K, Okazaki K, Chiba T. Toll-like receptor activation in basophils contributes to the development of IgG4-related disease. J Gastroenterol. 2013;48:247-253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
38.  Yanagawa M, Uchida K, Ando Y, Tomiyama T, Yamaguchi T, Ikeura T, Fukui T, Nishio A, Uemura Y, Miyara T, Okamoto H, Satoi S, Okazaki K. Basophils activated via TLR signaling may contribute to pathophysiology of type 1 autoimmune pancreatitis. J Gastroenterol. 2018;53:449-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
39.  Sokol CL, Medzhitov R. Emerging functions of basophils in protective and allergic immune responses. Mucosal Immunol. 2010;3:129-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 48]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
40.  Poddighe D, Mathias CB, Freyschmidt EJ, Kombe D, Caplan B, Marseglia GL, Oettgen HC. Basophils are rapidly mobilized following initial aeroallergen encounter in naïve mice and provide a priming source of IL-4 in adaptive immune responses. J Biol Regul Homeost Agents. 2014;28:91-103.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Poddighe D, Mathias CB, Brambilla I, Marseglia GL, Oettgen HC. Importance of basophils in eosinophilic asthma: the murine counterpart. J Biol Regul Homeost Agents. 2018;32:335-339.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Dossybayeva K, Abdukhakimova D, Poddighe D. Basophils and Systemic Lupus Erythematosus in Murine Models and Human Patients. Biology (Basel). 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
43.  Watanabe T, Yamashita K, Arai Y, Minaga K, Kamata K, Nagai T, Komeda Y, Takenaka M, Hagiwara S, Ida H, Sakurai T, Nishida N, Strober W, Kudo M. Chronic Fibro-Inflammatory Responses in Autoimmune Pancreatitis Depend on IFN-α and IL-33 Produced by Plasmacytoid Dendritic Cells. J Immunol. 2017;198:3886-3896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
44.  Cayrol C, Girard JP. IL-33: an alarmin cytokine with crucial roles in innate immunity, inflammation and allergy. Curr Opin Immunol. 2014;31:31-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 431]  [Cited by in F6Publishing: 481]  [Article Influence: 48.1]  [Reference Citation Analysis (0)]
45.  Vivier E, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie ANJ, Mebius RE, Powrie F, Spits H. Innate Lymphoid Cells: 10 Years On. Cell. 2018;174:1054-1066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 986]  [Cited by in F6Publishing: 1264]  [Article Influence: 252.8]  [Reference Citation Analysis (0)]
46.  Ma X, Wu D, Zhou S, Wan F, Liu H, Xu X, Zhao Y, Tang M. The pancreatic cancer secreted REG4 promotes macrophage polarization to M2 through EGFR/AKT/CREB pathway. Oncol Rep. 2016;35:189-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 43]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
47.  Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT, Sahebkar A. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233:6425-6440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1445]  [Cited by in F6Publishing: 2348]  [Article Influence: 391.3]  [Reference Citation Analysis (0)]
48.  Furukawa S, Moriyama M, Tanaka A, Maehara T, Tsuboi H, Iizuka M, Hayashida JN, Ohta M, Saeki T, Notohara K, Sumida T, Nakamura S. Preferential M2 macrophages contribute to fibrosis in IgG4-related dacryoadenitis and sialoadenitis, so-called Mikulicz's disease. Clin Immunol. 2015;156:9-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 97]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
49.  Mingomataj EÇ, Bakiri AH. Regulator Versus Effector Paradigm: Interleukin-10 as Indicator of the Switching Response. Clin Rev Allergy Immunol. 2016;50:97-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
50.  Mosser DM, Zhang X. Interleukin-10: new perspectives on an old cytokine. Immunol Rev. 2008;226:205-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 697]  [Cited by in F6Publishing: 748]  [Article Influence: 49.9]  [Reference Citation Analysis (0)]
51.  Raimondi S, Lowenfels AB, Morselli-Labate AM, Maisonneuve P, Pezzilli R. Pancreatic cancer in chronic pancreatitis; aetiology, incidence, and early detection. Best Pract Res Clin Gastroenterol. 2010;24:349-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in F6Publishing: 399]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
52.  Maitra A, Fukushima N, Takaori K, Hruban RH. Precursors to invasive pancreatic cancer. Adv Anat Pathol. 2005;12:81-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 198]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
53.  Kamisawa T, Tsuruta K, Okamoto A, Horiguchi S, Hayashi Y, Yun X, Yamaguchi T, Sasaki T. Frequent and significant K-ras mutation in the pancreas, the bile duct, and the gallbladder in autoimmune pancreatitis. Pancreas. 2009;38:890-895.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 58]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
54.  Kinugawa Y, Uehara T, Sano K, Matsuda K, Maruyama Y, Kobayashi Y, Nakajima T, Hamano H, Kawa S, Higuchi K, Hosaka N, Shiozawa S, Ishigame H, Ota H. Methylation of Tumor Suppressor Genes in Autoimmune Pancreatitis. Pancreas. 2017;46:614-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
55.  Sato N, Parker AR, Fukushima N, Miyagi Y, Iacobuzio-Donahue CA, Eshleman JR, Goggins M. Epigenetic inactivation of TFPI-2 as a common mechanism associated with growth and invasion of pancreatic ductal adenocarcinoma. Oncogene. 2005;24:850-858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 113]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
56.  Sideras K, Braat H, Kwekkeboom J, van Eijck CH, Peppelenbosch MP, Sleijfer S, Bruno M. Role of the immune system in pancreatic cancer progression and immune modulating treatment strategies. Cancer Treat Rev. 2014;40:513-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 118]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
57.  Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6912]  [Cited by in F6Publishing: 7713]  [Article Influence: 482.1]  [Reference Citation Analysis (0)]
58.  Tassi E, Gavazzi F, Albarello L, Senyukov V, Longhi R, Dellabona P, Doglioni C, Braga M, Di Carlo V, Protti MP. Carcinoembryonic antigen-specific but not antiviral CD4+ T cell immunity is impaired in pancreatic carcinoma patients. J Immunol. 2008;181:6595-6603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 83]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
59.  Siveen KS, Kuttan G. Role of macrophages in tumour progression. Immunol Lett. 2009;123:97-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 265]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
60.  Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, Albacker LA, Koulmanda M, Freeman GJ, Sayegh MH, Sharpe AH. Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med. 2006;203:883-895.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 839]  [Cited by in F6Publishing: 932]  [Article Influence: 51.8]  [Reference Citation Analysis (0)]
61.  Tian L, Ma J, Ma L, Zheng B, Liu L, Song D, Wang Y, Zhang Z, Gao Q, Song K, Wang X. PD-1/PD-L1 expression profiles within intrahepatic cholangiocarcinoma predict clinical outcome. World J Surg Oncol. 2020;18:303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
62.  Lu SW, Pan HC, Hsu YH, Chang KC, Wu LW, Chen WY, Chang MS. IL-20 antagonist suppresses PD-L1 expression and prolongs survival in pancreatic cancer models. Nat Commun. 2020;11:4611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
63.  Hu H, Hang JJ, Han T, Zhuo M, Jiao F, Wang LW. The M2 phenotype of tumor-associated macrophages in the stroma confers a poor prognosis in pancreatic cancer. Tumour Biol. 2016;37:8657-8664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 129]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
64.  Poddighe D, Brambilla I, Marseglia GL. Basophils activated via TLR signaling may contribute to pathophysiology of type I autoimmune pancreatitis". J Gastroenterol. 2018;53:791-792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
65.  De Monte L, Wörmann S, Brunetto E, Heltai S, Magliacane G, Reni M, Paganoni AM, Recalde H, Mondino A, Falconi M, Aleotti F, Balzano G, Algül H, Doglioni C, Protti MP. Basophil Recruitment into Tumor-Draining Lymph Nodes Correlates with Th2 Inflammation and Reduced Survival in Pancreatic Cancer Patients. Cancer Res. 2016;76:1792-1803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 88]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
66.  Marone G, Schroeder JT, Mattei F, Loffredo S, Gambardella AR, Poto R, de Paulis A, Schiavoni G, Varricchi G. Is There a Role for Basophils in Cancer? Front Immunol. 2020;11:2103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
67.  Seo MS, Yeo J, Hwang IC, Shim JY. Risk of pancreatic cancer in patients with systemic lupus erythematosus: a meta-analysis. Clin Rheumatol. 2019;38:3109-3116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
68.  Pan Q, Gong L, Xiao H, Feng Y, Li L, Deng Z, Ye L, Zheng J, Dickerson CA, An N, Yang C, Liu HF. Basophil Activation-Dependent Autoantibody and Interleukin-17 Production Exacerbate Systemic Lupus Erythematosus. Front Immunol. 2017;8:348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 40]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
69.  Poddighe D, Marseglia GL. Commentary: Basophil Activation-Dependent Autoantibody and Interleukin-17 Production Exacerbate Systemic Lupus Erythematosus. Front Immunol. 2017;8:787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]