Vous êtes sur la page 1sur 14

Diagnosis and Management of Autoimmune Pancreatitis

Aravind Sugumar, MD

KEYWORDS Pancreatic cancer Idiopathic duct centric pancreatitis Lymphoplasmacytic sclerosing pancreatitis IgG4

Autoimmune pancreatitis (AIP) is a unique form of corticosteroid responsive chronic pancreatitis. In recent years, a distinct clinical presentation, histology, serology, and subtypes of AIP have been identied. This rare form of chronic pancreatitis was probably rst described by Henry Sarles in the 1960s.1 He described a type of sclerosing pancreatitis associated with hypergammaglobulinemia, which is very similar to the present-day description of AIP. The term autoimmune pancreatitis was not used until 1995, when it was coined by Yoshida and colleagues to describe a type of chronic pancreatitis associated with Sjgren-like syndrome.2 Early descriptions of the disease were mostly in the form of case reports.35 Major milestones in the description of AIP include Hamano and colleagues description of the association between serum IgG4 and AIP, Kamisawa and coworkers explanation that AIP was a systemic disease affecting multiple organs, and Chari and colleagues publication of the Histology, Imaging, Serology, Other organ involvement and Response to therapy (HISORt) criteria for the diagnosis of AIP.6 9 As with any new entity, a plethora of terms and synonyms were used to describe the same disease.10 13 A major reason for this varied use of terms is that AIP is a heterogeneous disease. We now know that previously described entities such as lymphoplasmacytic sclerosing pancreatitis, idiopathic duct centric pancreatitis, IgG4associated pancreatitis, and steroid-sensitive sclerosing pancreatitis are descriptions of the subtypes of AIP and not different diseases.14,15 The use of these alternate terms hindered progress; therefore, it was recently agreed that AIP should be divided into type 1 and type 2 to facilitate international uniformity in description.15,16
SUBTYPES OF AIP

AIP is a heterogeneous disease with two distinct subtypes: type 1 and type 2, which are classied according to histology.15,16 However, the clinical presentation can suggest a strong basis for suspecting one subtype from the other, as there are numerous

The author has nothing to disclose. Division of Gastroenterology and Hepatology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA E-mail address: asugumar@kumc.edu Gastroenterol Clin N Am 41 (2012) 9 22 doi:10.1016/j.gtc.2011.12.008 gastro.theclinics.com 0889-8553/12/$ see front matter 2012 Elsevier Inc. All rights reserved.

10

Sugumar

Table 1 Differences between type 1 and type 2 AIP Type 1 AIP Gender distribution Histology

Type 2 AIP M Yes Infrequent Infrequent Scant\none Present Present Yes No No Yes Absent Strong F

M Yes

Lymphoplasmacytic inltrate Storiform brosis Obliterative phlebitis IgG4 cell inltrate Pancreatic duct destruction GEL lesions

Present Present Present Absent Absent Not in all cases Mostly Yes Yes Present Some

Histologic conrmation need for diagnosis Serum IgG4 elevated Other organ involvement Responds to steroids Disease relapse Association with IBD

Abbreviations: GEL, granulocyte epithelial lesion; IBD, inammatory bowel disease.

differences between the two subtypes, along with a few similarities (Table 1). Most of the current literature on AIP is based on what we know about type 1 AIP.16,17 We are still in the early stages of understanding and describing type 2 AIP. In this article, unless explicitly stated, the generic term AIP pertains to type 1 AIP. Type 1 AIP is best described as the pancreatic manifestation of a systemic broinammatory disease, which at its core has a unique histology and tissue inltration of immunoglobulin G4 (IgG4)-positive inammatory cells.8,9,18 This systemic disease can affect many different organs in such a manner that the umbrella term immunoglobulin G4 associated systemic diseases is used to underscore the unique role played by IgG4.8,9,19,20 All the organs aficted by immunoglobulin G4 associated systemic diseases, including the pancreas, share a common, distinct, and discernible histology. Almost all of the early descriptions of type 1 AIP came from Asia, Japan in particular, and type 1 continues to be the predominant subtype seen there.8,9,19,20 In addition to the established diagnostic criteria that exist for type 1 AIP, the clinical presentation is so distinctive that histologic conrmation is needed in a small proportion of patients.16,17,21 Conversely, type 2 AIP, with its discernible histology, appears to exclusively affect the pancreas. Most of the literature describing this subtype comes from Europe and the United States.11,2225 As of now, although type 2 AIP can be clinically suspected, histologic conrmation is need in all cases to make a certain diagnosis. The differences between the two subtypes are illustrated in Table 1. It is important to note that the two subtypes are virtually indistinguishable on cross-sectional imaging, and both types are exquisitely sensitive to steroid therapy.1517,26
PATHOPHYSIOLOGY

Because AIP is so responsive to steroid therapy, an autoimmune cause has been inferred.8,2729 The precise pathogenesis of AIP has not been elucidated; however, there are interesting genetic and immunologic associations, in addition to the possibility of molecular mimicry, surrounding AIP.30 33 The most striking observation

Autoimmune Pancreatitis

11

is the association between serum IgG4 and AIP.7 IgG4 constitutes the smallest fraction of total IgG in humans.34 IgG4 is involved in the activation of the classic pathway of complement.35 Marked elevation in serum IgG4 is seen in only a limited number of autoimmune and parasitic diseases.36 39 It is still unclear if IgG4 plays a direct pathogenic role in developing AIP or if it is an epiphenomenon.40 42 Preliminary genetic studies from Japan have suggested that a specic human leukocyte antigen (HLA) type (DRB1*0405-DQB1*0401) is associated with AIP.30 Molecular mimicry by microbial pathogen, which leads to a cross reaction with endogenous antigens, has been postulated as a cause of many autoimmune conditions including AIP. There is limited evidence pointing toward Helicobacter pylori as the punitive organism.32,33 The theory suggests that exposure to Helicobacter pylori could lead to AIP in predisposed individuals.
HISTOLOGY

Both subtypes of autoimmune pancreatitis have common histologic features (periductal lymphoplasmacytic inltrate and an inammatory cellular stroma), which positively differentiate it from other types of chronic pancreatitis.1518 In addition, histology serves as the basis for distinguishing the two clinical phenotypes of AIP.18 The histologic hallmark of type 1 AIP is a set of four features, which together are called lymphoplasmacytic sclerosing pancreatitis (LPSP). LPSP consists of a dense periductal lymphoplasmacytic inltrate, storiform pancreatic brosis, presence of IgG4positive cells ( 10/high-power eld), and obliterative phlebitis.18 In addition, the pancreatic ductal architecture is often preserved and there are prominent lymphoid follicles. When most, if not all, of these features are present, they substantiate the diagnosis of type 1 AIP. In this context, it is important to point out that the presence of IgG4-positive cells is just one of the features of LPSP and that the histologic diagnosis of type 1 AIP should not be made or excluded solely on the presence or absence of this one feature alone. Features of LPSP are also seen in the histologic specimens of the other organs involved in type 1 AIP: salivary glands and lymph nodes.19,43 On histology, type 2 AIP also consists of a predominantly lymphoplasmacytic inltrate but with a few key differences from type 1 AIP. There is a dense periductal inammation with neutrophils leading to duct destruction in most cases. This characteristic lesion is called a granulocyte epithelial lesion. These lesions are pathognomonic of type 2 AIP.18,23,25 There is no inammation around the intrapancreatic artery or vein and there are scant groups of IgG4-positive cells. In both forms of AIP, there is a striking absence of intraductal protein plugs, stones, and pseudocysts, which are seen in other types of chronic pancreatitis.18
CLINICAL CONTEXT

The true incidence of AIP is unknown. The best estimates so far come from studies of patients who were presumed to have pancreatic cancer and underwent resection, but the resection specimen showed AIP. AIP was the diagnosis in approximately 3% to 5% of such patients.24,44 In Japan, the incidence of AIP was reported to be 0.82 per 100,000 people.45 The clinical presentation of AIP can be best divided into the acute and the postacute phase. In the acute phase, the classic presentation of AIP is that of obstructive jaundice with abdominal imaging consistent with pancreatic enlargement.6,1517 Thus, in clinical practice it is imperative to differentiate AIP from pancreatic cancer. Less commonly, AIP presents with mild abdominal pain; elevated pancreatic enzymes, which may also signal acute pancreatitis; or jaundice with proximal bile duct involvement, which must be differentiated from cholangiocarcinoma. In the

12

Sugumar

postacute phase, AIP can present with pancreatic atrophy and steatorrhea resembling chronic pancreatitis. A recent study from the Mayo Clinic concluded that fewer than 33% of patients with AIP have features of acute or chronic pancreatitis at presentation. More importantly, only 7 of 178 (3.9%) patients evaluated for etiology of suspected pancreatitis had AIP.46 Although mild abdominal pain can be seen occasionally in patients with AIP, a feature conspicuous by its absence in AIP is severe unremitting abdominal pain requiring narcotic pain medications.16 The presence of such severe pain should prompt a reevaluation of the diagnosis. Diabetes mellitus (DM) is seen in up to 50% of patients with AIP. Interestingly, this DM resolves in a proportion of patients with corticosteroid therapy.47,48 After initial treatment with corticosteroids, the pancreas can undergo atrophy on cross-sectional imaging, which can be accompanied by signicant steatorrhea (postacute phase presentation). Untreated AIP can also lead to pancreatic atrophy and brosis, which in its late stages can be indistinguishable from other forms of chronic pancreatitis.49
IMAGING

Pancreatic imaging is the cornerstone to the diagnosis of AIP. Pancreatic imaging can be subdivided into pancreatic parenchyma imaging and pancreatic ductal imaging. Pancreatic parenchymal imaging in the form of computed tomography (CT) or magnetic resonance imaging (MRI) is often performed as part of the initial testing for obstructive jaundice. A pancreas protocol abdominal CT is rapidly becoming the imaging modality of choice to diagnose AIP. Such a CT scan serves not only to diagnose AIP, but also to help rule in or rule out pancreatic cancer. There are three classic features of AIP on a pancreas protocol abdominal CT: a diffusely enlarged, sausage-shaped pancreas with featureless borders; a low-density rim surrounding the pancreas; and delayed enhancement of the pancreas in the late arterial phase.16,50 53 (Figs. 1 and 2). The presence of a low-density mass, along with an abrupt cutoff of a dilated pancreatic duct and atrophy of the pancreas distal to the obstruction, are all signs of pancreatic cancer, which can be detected on CT/ MRI16,50,52,53 (Fig. 3). AIP can also present as a focal enlargement of the pancreas, in which case it can be very challenging to differentiate it from pancreatic cancer by imaging

Fig. 1. Sausage-shaped pancreas with arrow at the low-density rim.

Autoimmune Pancreatitis

13

Fig. 2. Sausage-shaped pancreas with arrow showing a low-density rim.

alone. Although MRI of the abdomen is comparable to a CT scan, higher cost and decreased availably limit its use.54 56 On MRI, T1-weighted images of the pancreas are often less intense than T2-weighted images when compared to the liver.54 56 Endoscopic ultrasound (EUS) plays a very important adjunctive role in the workup of AIP. Although classic EUS features of AIP have been described (such as a diffusely hypoechoic gland), the greatest benet of EUS is the access it provides to obtain pancreatic tissue by ne-needle aspiration (FNA) or a core biopsy.5759 Although an FNA is sufcient for diagnosing pancreatic cancer, it is not adequate for diagnosing AIP. For histologic conrmation of AIP a core biopsy of the pancreas is needed, and it can also help to establish the basis for the subtypes of AIP.16,59,60 EUS-FNA is paramount in ruling out pancreatic cancer in patients with nonclassic CT imaging

Fig. 3. Enlargement of the pancreatic head with a focal low-density mass (arrow) suggestive of pancreatic cancer.

14

Sugumar

Fig. 4. ERP in AIP. Feature 1, Long narrow stricture greater than one third of the main pancreatic duct. Feature 2, Lack of upstream pancreatic duct dilatation from the stricture. Feature 3, Multiple noncontiguous strictures. Feature 4, Side branch duct arising from a stricture.

features. EUS elastography and positron emission tomography (PET) are exciting newer additions to the imaging armamentarium and hold much promise, but at present are not incorporated into the existing diagnostic criteria.61 63 Pancreatic duct imaging with endoscopic retrograde pancreatography (ERP) can be a useful adjunct in the diagnosis of AIP, especially in patients with nonclassic CT abdominal features for AIP, as well as for pancreatic cancer. A recently concluded study found that ERP features of AIP include the presence of a long narrow stricture (more than one third of the main pancreas duct), the lack of upstream dilation from the stricture, side branches arising from the strictured portion of the duct, and multiple noncontiguous strictures (Figs. 4 and 5). The presence of all these features is more suggestive of AIP than the presence of any one of them.64 Although magnetic resonance cholangiopancreatography (MRCP) is an attractive minimally invasive way of visualizing the pancreatic duct, ERP and MRCP have not been compared to each other.
SEROLOGY

Although numerous autoimmune antibodies have been reported to be elevated in AIP, the latest international consensus diagnostic criteria recommend measuring only serum IgG4 for diagnosing AIP.16 Initial studies showed that elevated IgG4 had a greater than 95% sensitivity and specicity in diagnosing AIP. More recent studies reveal a much lower sensitivity and specicity.7,40,42 Because there is great variation in the way IgG4 is measured in different parts of the world, it is best to interpret it in terms of the elevation above the normal value. When serum IgG4 is at least twofold above the normal, the value is highly suggestive of AIP.16,40 If all the typical features

Autoimmune Pancreatitis

15

Fig. 5. Long narrow stricture greater than one third of the main pancreatic duct seen in AIP.

of AIP are seen on abdominal CT imaging, then any elevation in serum IgG4 is supportive of AIP (Fig. 6). In the absence of typical imaging features, the threshold for serum IgG4 to help diagnose AIP has been set to twofold of the upper limit of normal.40 It is also important to note that some patients with type 1 AIP are seronegative for IgG4.16 Therefore, it should not be assumed that patients who are seronegative have type 2 AIP. Also, up to 7% to 10% of patients with pancreatic cancer can have elevated IgG4 levels.40,65 As helpful as serum IgG4 may be, elevated IgG4 level should never be considered the sole criterion for diagnosis of AIP, especially in the absence of abdominal imaging evidence. Likewise, during the treatment phase of AIP, serum IgG4 level should not be used as a marker to gauge response to therapy. IgG4 levels often uctuate during corticosteroid therapy and do not correlate with disease activity.
OTHER ORGAN INVOLVEMENT

Type 1 AIP is the pancreatic manifestation of a multisystem disease. Thus, the well-described pattern of the multiple other organs involved is an important clue to the diagnosis.14,16,66 69 The presence of other organ involvement can lead to characteristic symptoms, such as dry eyes and dry mouth (Sjgren-like syndrome), jaundice (bile duct involvement), and swelling in the groin (regional lymphadenopathy). Other organ involvement (OOI) that can also be seen on abdominal imaging includes retroperitoneal brosis, and renal involvement. Although not recommended as part of the routine workup of AIP, PET can be useful in picking up the other organs involved. When OOI is present, it not only strengthens the diagnosis of AIP, but it also allows for histologic conrmation of AIP, as tissue can be readily obtained if needed (eg, lymph node biopsy). The commonly described involvement of other organs includes

16

Sugumar

Fig. 6. Algorithm for distinguishing AIP from pancreatic cancer.

the proximal bile duct followed by salivary gland, retroperitoneum, orbits, ampulla of Vater, lymph nodes, and kidneys.14,66 70 Less commonly, gallbladder and gastric involvement have also been described.68,71 Symptoms related to OOI often improve with treatment and can serve as useful indicators of response to treatment.17,72
DIAGNOSIS

In clinical practice, diagnosing AIP can be quiet challenging. As the disease mimics pancreatic cancer, the price of misdiagnosis is often high. The Japan Pancreas Society (JPS) in 2002 devised the rst diagnostic criteria for AIP. These criteria were later modied in 2006 (JPS 2006). Early on, the emphasis was to not miss pancreatic cancer, rather than to diagnose AIP. Since that time, a plethora of diagnostic criteria

Autoimmune Pancreatitis

17

have been proposed, including the HISORt (2006), the Korean criteria (2007), the Asian Consensus criteria (2008), and the most recent International Consensus Diagnostic Criteria (2010).6,16,7376 All of these criteria, in some way, combine pancreatic imaging and histology with serology or other organ involvement or response to corticosteroid therapy, or all of these. One possible algorithm to help diagnose AIP is illustrated in Fig. 6. More detailed algorithms have been published elsewhere.16,50,53 When confronted with a clinical situation in which AIP is suspected, the gastroenterologist must remember two cardinal facts: rst, AIP is a rare disease, and second, pancreatic cancer is far more common than AIP. Recent national and international consensus criteria agree that abdominal imaging in the form of CT/MRI should form the cornerstone to the diagnosis of AIP.16,17 If imaging features typical of AIP are present, along with some collateral evidence in the form OOI or elevated serum IgG4 (any level), the patient can be started on corticosteroid therapy for AIP. In the absence of typical imaging features of AIP, a thorough workup to rule out pancreatic cancer needs to be done (EUS-FNA, carbohydrate antigen 19-9 [CA 19-9], etc). In this setting, despite ruling out pancreatic cancer, the bar for the collateral evidence for AIP has to be set high (ie, histology from OOI, twofold elevation of serum IgG4, or presence of ERP features of AIP). If the above highly suggestive pieces of collateral evidence for AIP are not present, then for these patients, a pancreatic core biopsy, a steroid trial, or surgical resection of the pancreas is required to make a diagnosis. These interventions, and especially the steroid trial (see later), should be limited to select situations and performed only by gastroenterologists with expertise in treating AIP.
TREATMENT

There are no prospective data from clinical trials to guide management of AIP; therefore, treatment regimens are based on retrospective analysis and expert opinion.47,49 Both subtypes of AIP are exquisitely sensitive to corticosteroid therapy. This fact has been so well described that response to corticosteroid therapy can be both diagnostic and therapeutic. It is important to distinguish the two distinct roles corticosteroids play in AIP. When typical imaging features and collateral evidence for AIP are absent and pancreatic cancer has been reliably ruled out, a steroid trial of oral prednisone 40 mg (or 0.6 mg/kg) once a day for 2 weeks can be started. When performed by gastroenterologists experienced in treating AIP, a steroid trial can be used to diagnose AIP. Response to steroids is based on objective data such as radiologic evidence showing a dramatic decrease in the pancreatic mass or OOI, resolution of the obstructive jaundice without biliary stenting (or being able to remove preexisting biliary stent without the recurrence of the jaundice), and normalization of liver function test. If there is no such improvement or if the CA 19-9 level is rising, then the diagnosis of AIP should be reconsidered and further efforts to rule out pancreatic cancer should be pursued.16,17,50 Subjective criteria, such as the patient reporting more energy or feeling better, or serum IgG4 levels normalizing, should not be considered a response to steroids. Once the diagnosis of AIP has been established, the best initial treatment is oral prednisone 40 mg (or 0.6 mg/kg) for 4 weeks, including the 2-week steroid trial, if performed. Beginning at week 4, with continued objective response to therapy, the dose should be tapered by 5 mg per week. Thus, the total duration of initial corticosteroid therapy is 12 weeks (4 weeks of therapy plus 8 weeks of taper). Up to 40% of patients (mostly type 1 AIP) will have disease relapse after the rst course of corticosteroid therapy.72,77,78 Proximal bile duct involvement (away from common bile duct) can be a predictor of disease relapse. Disease relapse can be observed by

18

Sugumar

radiologic (reappearance of the pancreatic mass), clinical (obstructive jaundice, recurrence of OOI, weight loss), or serologic (repeat elevation in serum IgG4) means. Most experts agree that an isolated serologic relapse can observed and does not need maintenance therapy. Clinical and radiologic relapses, on the other hand, require treatment.72 Most patients with disease relapse will need a full course of oral prednisone similar to the initial 12-week treatment along with concomitant oral azathioprine (at 22.5 mg/kg).26,79 These patients are advised to continue maintenance azathioprine (at 22.5 mg/kg) for at least 2 years. If there are no further relapses, maintenance therapy can be stopped. While the patient is on maintenance therapy, it is imperative to evaluate the complete blood counts and liver function test frequently. Early data for the use of Rituximab in patients with AIP who have relapsed on azathioprine or who cannot tolerate corticosteroids are encouraging.80
SUMMARY

AIP is a heterogeneous disease with two distinct subtypes, now called type 1 and type 2. The proportions of these subtypes vary in their distribution worldwide. Pancreatic cancer is the leading differential diagnosis for AIP, although AIP can mimic any other major pancreatobiliary disease. Cross-sectional abdominal imaging CT/MRI should form the cornerstone to the diagnosis of AIP. Serum IgG4 provides collateral evidence for the diagnosis of AIP and should not be the sole basis for the diagnosis. False-positive elevation in serum IgG4 can be seen in up to 10% of patients with pancreatic cancer. A steroid trial should be performed only in select situations after ruling out pancreatic cancer and by gastroenterologists experienced in treating AIP. Disease recurrence can be seen in up to 40% of patients after initial steroid therapy.
REFERENCES

1. Sarles H, Sarles JC, Muratore R, et al. Chronic inammatory sclerosis of the pancreasan autonomous pancreatic disease? Am J Dig Dis 1961;6:688 98. 2. Yoshida K, Toki F, Takeuchi T, et al. Chronic pancreatitis caused by an autoimmune abnormality. Proposal of the concept of autoimmune pancreatitis. Dig Dis Sci 1995; 40(7):1561 8. 3. Chutaputti A, Burrell MI, Boyer JL. Pseudotumor of the pancreas associated with retroperitoneal brosis: a dramatic response to corticosteroid therapy. Am J Gastroenterol 1995;90(7):1155 8. 4. Sood S, Fossard DP, Shorrock K. Chronic sclerosing pancreatitis in Sjogrens syndrome: a case report. Pancreas 1995;10(4):419 21. 5. Horiuchi A, Kaneko T, Yamamura N, et al. Autoimmune chronic pancreatitis simulating pancreatic lymphoma. Am J Gastroenterol 1996;91(12):26079. 6. Chari ST, Smyrk TC, Levy MJ, et al. Diagnosis of autoimmune pancreatitis: the Mayo Clinic experience. Clin Gastroenterol Hepatol 2006;4(8):1010 6. 7. Hamano H, Kawa S, Horiuchi A, et al. High serum IgG4 concentrations in patients with sclerosing pancreatitis. N Engl J Med 2001;344(10):732 8. 8. Kamisawa T, Egawa N, Nakajima H. Autoimmune pancreatitis is a systemic autoimmune disease. Am J Gastroenterol 2003;98(12):28112. 9. Kamisawa T, Funata N, Hayashi Y. Lymphoplasmacytic sclerosing pancreatitis is a pancreatic lesion of IgG4-related systemic disease. Am J Surg Pathol 2004;28(8): 1114.

Autoimmune Pancreatitis

19

10. Kawaguchi K, Koike M, Tsuruta K, et al. Lymphoplasmacytic sclerosing pancreatitis with cholangitis: a variant of primary sclerosing cholangitis extensively involving pancreas. Hum Pathol 1991;22(4):38795. 11. Ectors N, Maillet B, Aerts R, et al. Non-alcoholic duct destructive chronic pancreatitis. Gut 1997;41(2):263 8. 12. Kazumori H, Ashizawa N, Moriyama N, et al. Primary sclerosing pancreatitis and cholangitis. Int J Pancreatol 1998;24(2):1237. 13. Petter LM, Martin JK Jr, Menke DM. Localized lymphoplasmacellular pancreatitis forming a pancreatic inammatory pseudotumor. Mayo Clin Proc 1998;73(5):44750. 14. Sugumar A, Chari S. Autoimmune pancreatitis: an update. Expert Rev Gastroenterol Hepatol 2009;3(2):197204. 15. Sugumar A, Kloppel G, Chari ST. Autoimmune pancreatitis: pathologic subtypes and their implications for its diagnosis. Am J Gastroenterol 2009;104(9):2308 10 [quiz 11]. 16. Shimosegawa T, Chari ST, Frulloni L, et al. International consensus diagnostic criteria for autoimmune pancreatitis: guidelines of the International Association of Pancreatology. Pancreas 40(3):352 8. 17. Chari ST, Kloeppel G, Zhang L, et al. Histopathologic and clinical subtypes of autoimmune pancreatitis: the Honolulu consensus document. Pancreas 2010;39(5): 549 54. 18. Zhang L, Chari S, Smyrk TC, et al. Autoimmune pancreatitis (AIP) type 1 and type 2: an international consensus study on histopathologic diagnostic criteria. Pancreas 2011;40(8):11729. 19. Kamisawa T. IgG4-positive plasma cells specically inltrate various organs in autoimmune pancreatitis. Pancreas 2004;29(2):167 8. 20. Zen Y, Harada K, Sasaki M, et al. IgG4-related sclerosing cholangitis with and without hepatic inammatory pseudotumor, and sclerosing pancreatitis-associated sclerosing cholangitis: do they belong to a spectrum of sclerosing pancreatitis? Am J Surg Pathol 2004;28(9):1193203. 21. Okazaki K, Uchida K, Fukui T. Recent advances in autoimmune pancreatitis: concept, diagnosis, and pathogenesis. J Gastroenterol 2008;43(6):409 18. 22. Deshpande V, Chicano S, Finkelberg D, et al. Autoimmune pancreatitis: a systemic immune complex mediated disease. Am J Surg Pathol 2006;30(12):1537 45. 23. Kloppel G, Luttges J, Lohr M, et al. Autoimmune pancreatitis: pathological, clinical, and immunological features. Pancreas 2003;27(1):14 9. 24. Yadav D, Notahara K, Smyrk TC, et al. Idiopathic tumefactive chronic pancreatitis: clinical prole, histology, and natural history after resection. Clin Gastroenterol Hepatol 2003;1(2):129 35. 25. Zamboni G, Luttges J, Capelli P, et al. Histopathological features of diagnostic and clinical relevance in autoimmune pancreatitis: a study on 53 resection specimens and 9 biopsy specimens. Virchows Arch 2004;445(6):552 63. 26. Sugumar A, Chari ST. Autoimmune pancreatitis. J Gastroenterol Hepatol 2011;26(9): 1368 73. 27. Kim KP, Kim MH, Song MH, et al. Autoimmune chronic pancreatitis. Am J Gastroenterol 2004;99(8):160516. 28. Kojima E, Kimura K, Noda Y, et al. Autoimmune pancreatitis and multiple bile duct strictures treated effectively with steroid. J Gastroenterol 2003;38(6):6037. 29. Ito T, Nakano I, Koyanagi S, et al. Autoimmune pancreatitis as a new clinical entity. Three cases of autoimmune pancreatitis with effective steroid therapy. Dig Dis Sci 1997;42(7):1458 68.

20

Sugumar

30. Kawa S, Ota M, Yoshizawa K, et al. HLA DRB10405DQB10401 haplotype is associated with autoimmune pancreatitis in the Japanese population. Gastroenterology 2002;122(5):1264 9. 31. Park do H, Kim MH, Oh HB, et al. Substitution of aspartic acid at position 57 of the DQbeta1 affects relapse of autoimmune pancreatitis. Gastroenterology 2008;134(2): 440 6. 32. Kountouras J, Zavos C, Chatzopoulos D. A concept on the role of Helicobacter pylori infection in autoimmune pancreatitis. J Cell Mol Med 2005;9(1):196 207. 33. Kountouras J, Zavos C, Gavalas E, et al. Challenge in the pathogenesis of autoimmune pancreatitis: potential role of Helicobacter pylori infection via molecular mimicry. Gastroenterology 2007;133(1):368 9. 34. Vlug A, Nieuwenhuys EJ, van Eijk RV, et al. Nephelometric measurements of human IgG subclasses and their reference ranges. Ann Biol Clin [Paris] 1994;52(7 8):5617. 35. van der Zee JS, van Swieten P, Aalberse RC. Inhibition of complement activation by IgG4 antibodies. Clin Exp Immunol 1986;64(2):41522. 36. Aalberse RC, Van Milligen F, Tan KY, et al. Allergen-specic IgG4 in atopic disease. Allergy 1993;48(8):559 69. 37. Hussain R, Poindexter RW, Ottesen EA. Control of allergic reactivity in human lariasis. Predominant localization of blocking antibody to the IgG4 subclass. J Immunol 1992;148(9):27317. 38. Shirakata Y, Shiraishi S, Sayama K, et al. Subclass characteristics of IgG autoantibodies in bullous pemphigoid and pemphigus. J Dermatol 1990;17(11):661 6. 39. Ljungstrom I, Hammarstrom L, Kociecka W, et al. The sequential appearance of IgG subclasses and IgE during the course of Trichinella spiralis infection. Clin Exp Immunol 1988;74(2):230 5. 40. Ghazale A, Chari ST, Smyrk TC, et al. Value of serum IgG4 in the diagnosis of autoimmune pancreatitis and in distinguishing it from pancreatic cancer. Am J Gastroenterol 2007;102(8):1646 53. 41. Ghazale A, Chari ST, Zhang L, et al. Immunoglobulin G4-associated cholangitis: clinical prole and response to therapy. Gastroenterology 2008;134(3):706 15. 42. Raina A, Yadav D, Krasinskas AM, et al. Evaluation and management of autoimmune pancreatitis: experience at a large US center. Am J Gastroenterol 2009;104(9):2295 306. 43. Zhang L, Notohara K, Levy MJ, et al. IgG4-positive plasma cell inltration in the diagnosis of autoimmune pancreatitis. Mod Pathol 2007;20(1):23 8. 44. Smith CD, Behrns KE, van Heerden JA, et al. Radical pancreatoduodenectomy for misdiagnosed pancreatic mass. Br J Surg 1994;81(4):5859. 45. Nishimori I, Tamakoshi A, Otsuki M. Prevalence of autoimmune pancreatitis in Japan from a nationwide survey in 2002. J Gastroenterol 2007;42(Suppl 18):6 8. 46. Sah RP, Pannala R, Chari ST, et al. Prevalence, diagnosis, and prole of autoimmune pancreatitis presenting with features of acute or chronic pancreatitis. Clin Gastroenterol Hepatol 2010;8(1):91 6. 47. Kamisawa T, Shimosegawa T, Okazaki K, et al. Standard steroid treatment for autoimmune pancreatitis. Gut 2009;58(11):1504 7. 48. Nishimori I, Tamakoshi A, Kawa S, et al. Inuence of steroid therapy on the course of diabetes mellitus in patients with autoimmune pancreatitis: ndings from a nationwide survey in Japan. Pancreas 2006;32(3):244 8. 49. Kamisawa T, Yoshiike M, Egawa N, et al. Treating patients with autoimmune pancreatitis: results from a long-term follow-up study. Pancreatology 2005;5(23):234 8; discussion 8 40.

Autoimmune Pancreatitis

21

50. Chari ST, Takahashi N, Levy MJ, et al. A diagnostic strategy to distinguish autoimmune pancreatitis from pancreatic cancer. Clin Gastroenterol Hepatol 2009;7(10): 1097103. 51. Irie H, Honda H, Baba S, et al. Autoimmune pancreatitis: CT and MR characteristics. AJR Am J Roentgenol 1998;170(5):13237. 52. Kamisawa T, Imai M, Yui Chen P, et al. Strategy for differentiating autoimmune pancreatitis from pancreatic cancer. Pancreas 2008;37(3):e627. 53. Sugumar A, Chari ST. Distinguishing pancreatic cancer from autoimmune pancreatitis: a comparison of two strategies. Clin Gastroenterol Hepatol 2009;7(11 Suppl): S59 62. 54. Mikami K, Itoh H. MR imaging of multifocal autoimmune pancreatitis in the pancreatic head and tail: a case report. Magn Reson Med Sci 2002;1(1):54 8. 55. Ito K, Koike S, Matsunaga N. MR imaging of pancreatic diseases. Eur J Radiol 2001;38(2):78 93. 56. Kamisawa T, Chen PY, Tu Y, et al. MRCP and MRI ndings in 9 patients with autoimmune pancreatitis. World J Gastroenterol 2006;12(18):2919 22. 57. Kubota K, Kato S, Akiyama T, et al. A proposal for differentiation between early- and advanced-stage autoimmune pancreatitis by endoscopic ultrasonography. Dig Endosc 2009;21(3):1629. 58. Hoki N, Mizuno N, Sawaki A, et al. Diagnosis of autoimmune pancreatitis using endoscopic ultrasonography. J Gastroenterol 2009;44(2):154 9. 59. Mizuno N, Bhatia V, Hosoda W, et al. Histological diagnosis of autoimmune pancreatitis using EUS-guided trucut biopsy: a comparison study with EUS-FNA. J Gastroenterol 2009;44(7):74250. 60. Levy MJ, Smyrk TC, Takahashi N, et al. Idiopathic duct-centric pancreatitis: disease description and endoscopic ultrasonography-guided trucut biopsy diagnosis. Pancreatology 2011;11(1):76 80. 61. Dietrich CF, Hirche TO, Ott M, et al. Real-time tissue elastography in the diagnosis of autoimmune pancreatitis. Endoscopy 2009;41(8):718 20. 62. Itokawa F, Itoi T, Sofuni A, et al. EUS elastography combined with the strain ratio of tissue elasticity for diagnosis of solid pancreatic masses. J Gastroenterol 2011;46(6): 84353. 63. Kamisawa T, Takum K, Anjiki H, et al. FDG-PET/CT ndings of autoimmune pancreatitis. Hepatogastroenterology 2010;57(99 100):44750. 64. Sugumar A, Levy MJ, Kamisawa T, et al. Endoscopic retrograde pancreatography criteria to diagnose autoimmune pancreatitis: an international multicentre study. Gut 60(5):666 70. 65. Raina A, Krasinskas AM, Greer JB, et al. Serum immunoglobulin G fraction 4 levels in pancreatic cancer: elevations not associated with autoimmune pancreatitis. Arch Pathol Lab Med 2008;132(1):48 53. 66. Fukukura Y, Fujiyoshi F, Nakamura F, et al. Autoimmune pancreatitis associated with idiopathic retroperitoneal brosis. AJR Am J Roentgenol 2003;181(4):9935. 67. Eerens I, Vanbeckevoort D, Vansteenbergen W, et al. Autoimmune pancreatitis associated with primary sclerosing cholangitis: MR imaging ndings. Eur Radiol 2001;11(8):1401 4. 68. Kamisawa T, Nakajima H, Egawa N, et al. Autoimmune pancreatitis can be conrmed with gastroscopy. Dig Dis Sci 2004;49(1):155 6. 69. Kamisawa T, Okamoto A. Autoimmune pancreatitis: proposal of IgG4-related sclerosing disease. J Gastroenterol 2006;41(7):61325. 70. Kamisawa T, Tu Y, Egawa N, et al. A new diagnostic endoscopic tool for autoimmune pancreatitis. Gastrointest Endosc 2008;68(2):358 61.

22

Sugumar

71. Leise MD, Smyrk TC, Takahashi N, et al. IgG4-associated cholecystitis: another clue in the diagnosis of autoimmune pancreatitis. Dig Dis Sci 2011;56(5):1290 4. 72. Chari S, Murray JA. Autoimmune pancreatitis Part II: The relapse. Gastroenterology 2008. 73. Society MotCCfAPotJP. Diagnostic criteria for autoimmune pancreatitis by the Japan Pancreas Society. J Jpn Pancreas [Suizou] 2002;17:587. 74. Okazaki K, Kawa S, Kamisawa T, et al. Clinical diagnostic criteria of autoimmune pancreatitis: revised proposal. J Gastroenterol 2006;41(7):626 31. 75. Choi EK, Kim MH, Lee TY, et al. The sensitivity and specicity of serum immunoglobulin G and immunoglobulin G4 levels in the diagnosis of autoimmune chronic pancreatitis: Korean experience. Pancreas 2007;35(2):156 61. 76. Otsuki M, Chung JB, Okazaki K, et al. Asian diagnostic criteria for autoimmune pancreatitis: consensus of the Japan-Korea Symposium on Autoimmune Pancreatitis. J Gastroenterol 2008;43(6):403 8. 77. Gardner TB, Chari ST. Autoimmune pancreatitis. Gastroenterol Clin North Am 2008; 37(2):439 60, vii. 78. Ghazale A, Chari ST. Optimising corticosteroid treatment for autoimmune pancreatitis. Gut 2007;56(12):1650 2. 79. Sandanayake NS, Church NI, Chapman MH, et al. Presentation and management of post-treatment relapse in autoimmune pancreatitis/immunoglobulin G4-associated cholangitis. Clin Gastroenterol Hepatol 2009;7(10):1089 96. 80. Topazian M, Witzig TE, Smyrk TC, et al. Rituximab therapy for refractory biliary strictures in immunoglobulin G4-associated cholangitis. Clin Gastroenterol Hepatol 2008;6(3):364 6.

Vous aimerez peut-être aussi