search
for
 About Bioline  All Journals  Testimonials  Membership  News


Indian Journal of Cancer
Medknow Publications on behalf of Indian Cancer Society
ISSN: 0019-509X EISSN: 1998-4774
Vol. 46, Num. 4, 2009, pp. 288-296

Indian Journal of Cancer, Vol. 46, No. 4, October-December, 2009, pp. 288-296

Review Article

Pancreatic carcinogenesis: The impact of chronic pancreatitis and its clinical relevance

Department of Gastrointestinal Surgical Oncology, Tata Memorial Hospital, Mumbai
Correspondence Address:Department of Gastrointestinal Surgical Oncology, Tata Memorial Hospital, Mumbai
shailushrikhande@hotmail.com

Code Number: cn09070

PMID: 19749458
DOI: 10.4103/0019-509X.55548

Abstract

Pancreatic cancer is a devastating disease with a dismal prognosis and early detection remains a challenge. On the background that inflammation is one of the key steps in the development of cancer, it is natural that chronic pancreatitis is considered as one of the etiological factors for the development of pancreatic cancer. However, the process of pancreatic carcinogenesis is a multifactorial phenomenon rather than a process that evolves solely via inflammation. This review attempts to put into perspective the association between different etiological forms of chronic pancreatitis and pancreatic cancer, and the diverse mechanisms operational in the process of pancreatic carcinogenesis. Furthermore, the clinical relevance of the current understanding of the relationship between chronic pancreatitis and pancreatic cancer, especially with regard to the pancreatic head mass of uncertain etiology, is discussed in this review.

Keywords: Chronic pancreatitis, pancreatic cancer, pancreatic carcinogenesis, inflammation, pancreatic head mass, risk of pancreatic cancer

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy and is the fourth to fifth leading cause of death in the USA, with an incidence rate of 11.2 / 100,000. [1] It is estimated that 32,180 new cases will occur in the United States in 2005. [2] In India, the incidence of pancreatic cancer is low (0.5 - 2.4 per 100,000 men and 0.2 - 1.8 per 100,000 women) in most parts of the country. [3] Despite its apparently high incidence in the West, the pathophysiology of this lethal malignancy had not been well elucidated until recently. Advances in molecular biology have greatly improved the understanding of the pathogenesis of pancreatic cancer. The molecular alterations observed include epigenetic changes such as deregulated expression of a variety of genes, gross chromosomal abnormalities, and gene mutations; all these are crucial in the development and progression of pancreatic cancer. Many patients have mutations of the K- ras proto-oncogene, and inactivation of tumor-suppressor genes. Growth factors also play an important part. However, despite the ongoing molecular research on pancreatic carcinogenesis, little progress has resulted in therapeutic benefits, and hence, the disease prognosis remains poor. Around 15 - 20% of the patients have resectable disease, but only around 20% of these survive to five years, thus reflecting the aggressive nature of the disease. [4],[5]

Chronic pancreatitis (CP), is a chronic inflammatory disease of the exocrine pancreas and affects around eight patients per 100,000 population per year in the USA, with a prevalence of 26.4 cases per 100,000 population. [6] In contrast to pancreatic cancer, CP carries a much better prognosis and multiple treatment options are available to treat the main problems arising out of this disease, such as, pain and exocrine and endocrine deficiency. Ever since the first detailed description of CP by Comfort in 1946, [7] many different forms of CP have been described. It is now known that mutations in the cationic trypsinogen gene are present in patients with hereditary CP, and it has been reported that approximately 30% of the patients with so-called idiopathic CP have mutations in the cystic fibrosis transmembrane conductance regulator gene. [8],[9],[10],[11] Although the morphological changes in CP have been recognized and described for many decades, the pathophysiology of CP is still poorly understood, and it is not known whether the pathophysiological events in different CP etiologies are similar or diverse. In the Western industrialized countries, the dominant etiological factor of CP is alcohol abuse, accounting for approximately 80% of patients. [12] In a small percentage of patients with CP, alcohol abuse can be clearly ruled out as an etiological factor, and if other known etiological factors are absent, these patients are then classified as having idiopathic CP. [12],[13] In contrast with Western countries, in India, especially southern India, an alcohol independent form of CP - tropical pancreatitis - is common. [12] Here there is a chronic calcifying form of CP, in a younger age group, in a region of high incidence, without any history of alcohol consumption. The etiology of tropical CP is largely unknown, and nutritional and environmental factors have been propagated since the 1970s and 1980s, as being the major causative factors in the etiopathogenesis of this form of CP. [14],[15] More recently, SPINK 1 mutations have been implicated as the causative factors of tropical CP. [16],[17]

Irrespective of the underlying etiology, CP is characterized by a continuum of progressive parenchymal destruction, ductal strictures, atrophy of acinar and islet tissue, de-differentiation of acinar cells into tubular complexes, immune cell infiltration and inflammation, alteration of nerves, and progressive fibrosis. [18] Chronic inflammation, the dominant morphological feature of CP, is known to cause neoplastic transformation and it is not surprising that chronic pancreatitis (CP) is considered to be associated with an increased risk of pancreatic cancer. The recognition of the entity of autoimmune pancreatitis has further complicated the picture. The main issues are:

  1. What is the risk of pancreatic cancer in CP?
  2. How does CP accelerate the oncogenic process of pancreatic carcinogenesis?
  3. How does our current understanding help present day practice?
This review attempts to clarify the above-mentioned areas and the background of the relationship between CP and pancreatic cancer.

Risk of Pancreatic Cancer in CP: Epidemiological, Experimental, and Clinical Data

Alcoholic CP

The risk of development of cancer in alcoholic CP increases with increasing duration of the disease. In the landmark study by Lowenfels et al ., [19] it was observed that the risk was 1.8 and 4% at 10 and 20 years of alcoholic CP. The difficulty in evaluating the pathway of progression of Alcoholic CP into pancreatic cancer is complicated by the incidence of other life-threatening sequelae (Cirrhosis / Portal hypertension) that may intervene at an earlier stage, often long before CP causes oncogenic transformation in the affected pancreas.

Tropical and cystic fibrosis associated CP

Considering that the disease is characterized by an early age of onset (as compared to alcoholic CP), the changes induced by prolonged inflammation can therefore be better evaluated. In an observational study by Ramesh et al ., it was noted that 22 out of 266 patients with tropical CP developed pancreatic cancer, with a high risk of cancer in tropical CP of 8.3%. [20,21] Chari et al ., reported that over a 4.5-year period, 24 of 185 patients with TP died and that six (25%) died of pancreatic cancer. The average age of onset was 45 ± 7 years, and the relative risk of pancreatic cancer compared to those without TP was 100 times normal. [22] Similar rates have been noted with cystic fibrosis associated CP. [23],[24]

Hereditary CP

In the Europac study, the largest ever study evaluating the role of hereditary CP in development of pancreatic cancer, 418 patients from 112 families in 14 countries were evaluated and it was noted that the cumulative risk of pancreatic cancer was 8.5% at 40 years, in this form of CP. [25] A follow-up report from the original international study by Lowenfels [26] included 497 patients with hereditary pancreatitis, with 19 cases of pancreatic cancer compared with an expected number of 0.33, yielding a risk ratio of 57 (95%CI : 35 - 90). Both these studies, thus confirm, a high risk of pancreatic cancer in patients with hereditary pancreatitis, irrespective of the underlying mutation.

The above data based predominantly on epidemiological studies, indicates an increased likelihood of the development of pancreatic cancer in patients who have CP.

Role of CP in Oncogenic Transformation: Experimental Data

Extensive and prolonged inflammation results in the release of inflammatory mediators, cytokines, O 2 radicals, and upregulated inflammatory pathways. These events lead to cell death with increased proliferation, with organ regeneration in an oxidative-species-rich environment. These events create a situation where damaged DNA accumulates and this ultimately leads to pancreatic cancer. [27],[28] [Figure - 1] shows the process of oncogenic transformation on the background of inflammation.

Although this hypothesis seems attractive, it must be borne in mind that only 10% of chronic inflammatory conditions ultimately progress to cancer, [29] for example, chronic obstructive pulmonary disease resulting in lung cancer. Also, while it has been proposed that the findings in different etiological forms of CP are essentially similar and a common "surgical stage of CP" is reached despite the different pathways they initially follow, [30] the incidence of development of cancer is not uniform in these different forms of CP, as discussed in the preceding sections. It is thus apparent that other factors, in addition to inflammation, are operational in CP leading to development of pancreatic cancer.

Process of Oncogenic Transformation is Multifactorial

In addition to inflammation in CP, it appears that genetic susceptibility (causes premature trypsinogen activation with development of recurrent acute pancreatitis (RAP)), alcohol abuse, smoking, and viral infections also promote pancreatic parenchymal injury in susceptible individuals and cause RAP. In a multivariate analysis of 497 patients with hereditary CP, Lowenfels [26] has found that smoking independently doubled the risk of pancreatic cancer, which even developed 20 years earlier in smokers compared to nonsmokers and accounted for approximately 25 - 30% of all pancreatic tumors. RAP leads to inflammation, fibrosis, and CP, which ultimately results in cancer.

The PanIN system (noninvasive precursors of invasive pancreatic ductal adenocarcinoma) is a series of pathologic changes that correspond with various genetic alterations, which ultimately lead to the stage of preinvasive cancer. [31] Although, Pancreatic Intraepithelial Neoplasia (PanIN), [32] which refers to a microscopic papillary or flat-invasive, epithelial neoplasm arising in the pancreatic ducts (< 5mm) and is characterized by columnar-cuboidal cells with varying amounts of mucin and degrees of cytologic and architectural atypia, was earlier believed to occur only in pancreatic cancer, there are reports that PanINs can develop without cancer. One such condition in which they do occur is CP. [32] PanINs appear to run an accelerated course in CP compared to sporadic pancreatic cancer. [Figure - 2] shows the multifactorial process of pancreatic cancer development on the background of CP.

Molecular Markers for Differentiating CP from Pancreatic Cancer

Genetic studies of oncogene mutations are useful in expanding our knowledge on the relationship between CP and pancreatic adenocarcinoma. In a prospective study of 76 patients with CP (59 alcoholic, 15 idiopathic, and 2 hereditary), van Laethem [33] examined K-ras mutations in collected ductal brushings during therapeutic ERCP, and after a long-term follow-up, every 6 - 12 months, K-ras (codon 12) mutations - GAT and GTT in a majority of the cases - were found in 25% of the patients, quite similar to those found in adenocarcinoma. There was no relationship between the presence of mutant K-ras and the etiology of CP (alcoholic: 20% of mutant K-ras; idiopathic: 30%; hereditary: 50%), the duration of the disease before the analysis, the presence of acute attacks, the existence of pancreas divisum, or the presence of a pancreatic stent.

p53, p16, and Smad 4 mutations have also been evaluated as markers differentiating CP from pancreatic cancer. In fact, Talar-Wojnarowska et al . [34] in a review of the available literature on the incidence of specific sequences of oncogenes and tumor suppressor genes, including K-ras, Her-2/neu, p16, p53, and DPC4, found that the incidence of mutations of K-ras, p16, and p53 were much more common in pancreatic cancer as compared to CP. However, the current findings are not specific and sensitive enough for their routine clinical recommendation. [35]

DPC4 staining of ducts seen in CP is lost in about half of the patients with pancreatic cancer. [36] On the other hand, Ki-67, a marker of proliferative activity of neoplastic cells is much higher in pancreatic cancer as compared to CP. [36] Another immunohistological marker that may be useful in differentiating the two entities, is MUC1, which is not expressed in CP, but may be found in pancreatic cancer. [36]

New evidence is emerging on the role of micro RNAs (miRNA) in differentiating CP from pancreatic cancer. Bloomston et al . [37] found 15 overexpressed and eight underexpressed miRNAs in cancer. These included miRNA 221, a hitherto unreported miRNA in pancreatic cancer that has been found in thyroid cancer and is associated with angiogenesis. [37]. Based on proteomic analysis of tissue specimens from pancreatic cancer and CP, it has been found that expression analysis of UHRF1, ATP7A, and aldehyde oxidase 1, in combination, could potentially provide a useful additional diagnostic tool for fine-needle aspirated or cytological specimens obtained during endoscopic investigations. [38]

Pancreatic Head Mass: Chronic Pancreatitis or Cancer?

Complications of CP may affect the gland or the surrounding structures. Among others, CP can result in the development of an inflammatory head mass, which can become a considerable source of diagnostic confusion. It has been reported that an inflammatory mass larger than 4 cm may occur in as many as 75% of the patients requiring surgery for CP. [39] Such an inflammatory mass often raises suspicions of pancreatic cancer in the mind of a treating surgeon.

Clinical Features

Overlap in imaging studies, clinical findings, and treatment options frequently leads to confusion in diagnosis and management. [40] Differentiating between CP and pancreatic cancer can therefore prove to be extremely difficult due to this overlap in the signs and symptoms of each disease. Epigastric pain, weight loss, steatorrhoea,-associated diabetes, and obstructive jaundice are observed in both cancer and inflammation; however, they may occur in varying frequencies. [40] While CP usually has the classical picture of longstanding epigastric pain radiating to the back, history of alcohol consumption in the western countries, previous acute exacerbations, and history of chronic analgesic intake, the disease may often appear without an apparent previous history [41] and raise the doubt of pancreatic cancer in the clinicians mind. On the other hand, painless progressive jaundice, classical of a malignant lesion, is uncommonly associated with CP.

However, a high index of suspicion and attention to clinical detail may aid in reaching a precise diagnosis. Malignant transformation should be suspected when "new symptoms" develop in an established longstanding case of CP, such as, sudden and severe weight loss in a controlled diabetic or the development of jaundice.

Despite a high index of suspicion, pancreatic cancer is missed in 2 - 4% of patients undergoing pancreatic duct drainage for presumed CP. [42] Conversely, many series have reported a 5 - 10% inadvertent resection rate for "pancreatic cancer" ultimately proved to be CP. [43],[44],[45]

It is thus clear that additional inputs are necessary in some patients to arrive at a diagnosis, even after a detailed history and the best of clinical examinations.

Laboratory Investigations

CA 19-9

This tumor marker was first described in 1982 by Koprowski and his associates. [46] It is the most widely used conventional marker for pancreatic cancer and is a useful tool in the diagnosis and follow-up of patients after resection. However, its role in the early detection of pancreatic cancer is limited. The sensitivity of CA 19-9 is > 80% in pancreatic cancer, but the specificity is lower. [47] It has been suggested that a subsequent rise of CA 19-9 after postoperative normalization may precede clinical detection of recurrent disease. [48] Elevation of CA 19-9 levels corresponds to advanced disease and poorer outcomes in pancreatic cancer and even in gastric cancer. In a study evaluating the expression of CA 19-9 in normal pancreas, chronic pancreatitis, and pancreatic cancer, elevated levels were observed in 87% of the patients with pancreatic carcinoma compared to only 13% in the group of benign disease. [49],[50]

Currently, an elevation of serum CA 19-9 over previous baseline levels should be viewed with suspicion from the point of view of a malignant transformation. To date, there is no other specific laboratory investigation that might aid in differentiating pancreatic cancer from an inflammatory mass of CP.

Imaging

CT scan

The sensitivity of a CT scan is around 83% and the specificity 93% for ductal carcinomas [51] . In CP, the imaging features are ductal dilation, parenchymal calcification, and a chain-of-lakes appearance (mainly in tropical CP), often with ductal calculi, in addition to enlargement of the pancreatic head. Furthermore, as the disease progresses, the gland may look small and atrophic due to the ongoing fibrosis. On the other hand, pancreatic head cancer shows a slightly hypovascular lesion with irregular borders. [52],[53] Also, pancreatic calcification is uncommon. However, as seen in CP, pancreatic duct obstruction also occurs in pancreatic cancer with resultant ductal dilatation, and this can lead to a diagnostic dilemma.

Magnetic resonance imaging (MRI)

MRI with cholangiopancreatography (MRCP) has now been accepted as a technique that is able to completely replace diagnostic ERCP. When initially described in the mid 1990s, further studies were necessary for a better evaluation of the potential advantages and disadvantages of this technique. [54] The advantage of this technique is that the bilio- pancreatic ductal anatomy is better delineated, without any contrast injection being required. Although by no means 100% accurate, ultrafast MRI (UMRI) is now reported to be equal or even superior to all other staging methods. It has replaced other methods in many centers, because it provides an "all-in-one" investigation, avoiding endoscopy, vascular cannulation for angiography, allergic reactions, and radiation. However, it is not yet perfect and hence the final verdict on resectability of a pancreatic head mass still depends on surgical exploration in some cases. [55]

Positron emission tomography (PET)

(18)F-FDG PET scan imaging has been shown to be a useful tool in the diagnosis of pancreatic cancer and even to detect metastasis in patients post chemoradiotherapy. [56] It is also known to be useful in differentiating benign from malignant lesions based on an increased standardized uptake value (SUV) measured at two different time points. [57] An early experience with this technique in differentiating pancreatic cancer from chronic pancreatitis was demonstrated by Lan et al . [57] Thereafter, Bang et al . [58] have provided evidence to support the role of PET-CT as being an effective modality in detecting pancreatic cancer. However, further studies comparing the ability of PET imaging to differentiate cancer from pancreatitis are awaited before its place in the diagnostic Figure of an undifferentiated mass of pancreas can be confirmed.

Endoscopic Ultrasonography (EUS)

EUS has gained much attention, as it eliminates the problems of gas and obesity seen with the conventional ultrasound and can detect small pathologies, especially in the periampullary region, in experienced hands. EUS-guided fine needle aspiration of the pancreas (FNA) has a sensitivity of 80% and specificity of 82% in detecting tumors, [59] and this is an attractive concept in situations where a pathological diagnosis is necessary prior to initiating some form of treatment. [60] Horwhat et al . [60] in the randomized cross-over trial comparing EUS-guided FNA to CT-guided FNA were able to demonstrate a high sensitivity of 84% for EUS, as opposed to 62% for CT-guided FNA. Their study, however, was underpowered, leading to a non-significant p value. Although, what the study did confirm was that no malignancies were missed in the EUS-guided FNA group.

However, the sensitivity of the EUS-guided FNA falls to around 54% when used for diagnosing pancreatic cancer in chronic pancreatitis. [61],[62] Thus, while EUS is highly sensitive in detecting abnormal masses in cases of CP, the positive predictive value of the diagnosis of pancreatic cancer is low. [63] The reasons ascribed to this are the difficulty in targeting the exact lesion, coupled with the presence of fibrosis and inflammation in the surrounding pancreas. [64]

A recent work by Moller et al . [65] seems to indicate that combining the histological examination of core biopsies with cytological analysis (both obtained by EUS-guidance), can actually result in a higher sensitivity, with a lower number of biopsies required.

Do Combinations of Tests Improve Diagnostic Yield?

It has been reported [48] that a combination of CA 19-9 and ultrasound improved the sensitivity of each test by 10 - 15%. Currently, a careful, detailed history and clinical examination along with a high resolution contrast-enhanced CT scan and serum CA 19-9 levels would seem to be the best combination available to reach a likely preoperative diagnosis in a pancreatic head mass of uncertain etiology.

Intra-operative Diagnosis

In many instances, surgeons rely on an exploration in the case of pancreatic head masses of uncertain etiology, when the clinical picture and investigations do not exclude a malignancy. In these cases, trying to reach an intra-operative diagnosis may be complicated by findings that are equivocal for an ampullary or pancreatic head lesion or CP, such as, obstruction of the main pancreatic duct, a surrounding desmoplastic reaction, and so on. However, the main pancreatic ductal dilation with multiple strictures, calculi, pseudocysts, and parenchymal calcification are some of the features that would favor a diagnosis of chronic pancreatitis in a head mass rather than pancreatic cancer. While a fine needle aspiration (usually a transduodenal route) in combination with an intra-operative ultrasound may help reach a diagnosis, [66],[67] the surgeon may often have to resort to a pancreaticoduodenectomy. In the variant of autoimmune pancreatitis (which responds best to steroid treatment), the whole gland is usually affected and a distinctive head mass can often not be appreciated. [68]

Approach to Treatment

Whenever there is a possibility of cancer, pancreatic resection should be performed, as it is both diagnostic and therapeutic. In a study evaluating "blind" Whipple resections for periampullary and pancreatic lesions, without a prior diagnosis of malignancy, clinical judgment was observed to be correct in 90% of these patients. [69] This study reported that in patients with potentially resectable lesions (based on CT scan examination), biopsy information did not affect the choice of therapy, since a negative biopsy still committed the patient to surgery and the study was concluded by questioning the practice of numerous biopsy attempts in patients with periampullary lesions. [Figure - 3] provides a broad outline for management of a pancreatic head mass.

Summary

There is increasing evidence that pancreatic carcinogenesis is a multi-step phenomenon. Molecular diagnostic methods such as detection of K-ras mutations in pure pancreatic or duodenal juice, in addition to fine needle aspirations, can achieve earlier diagnosis of pancreatic cancer in selected subgroups, for example, in patients with CP. [70] Redirection of experimental work toward pancreatic inflammation and mechanisms of carcinogenesis may identify other novel anti-inflammatory agents or other methods to screen and even prevent pancreatic cancer. Potential markers (e.g., Annexin A4, Cyclophilin A, Cathepsin D, S100A8, etc.), unique to pancreatic cancer compared to the normal pancreas and CP, have recently been identified by using a proteomic approach of two-dimensional (2D) gel electrophoresis and mass spectrometry. They may eventually serve as diagnostic markers or therapeutic targets [71] in pancreatic tumors. miRNAs have also been described, which may help in differentiating the two entities.

With regard to the treatment where a dilemma exists between pancreatitis and cancer, the most important variable would be the surgeon himself and his team. The more experienced the pancreatic surgeon, the more likely it is for a major pancreatic resection to be performed with minimal morbidity and mortality, and an aggressive approach can be pursued in experienced centers so that some cancers get curative resection, and if the diagnosis ultimately turns out to be CP, an adequate relief from symptoms is maximally achieved. As far as autoimmune pancreatitis is concerned, a high index of suspicion needs to be maintained, to improve the rates of diagnosis, and unnecessary surgery has to be avoided.

References

1.Surveillance, Epidemiology, and End Results (SEER) Program. SEERFNx01Stat database. Incidence - SEER 9 Regs Public-Use, Nov 2003 Sub (1973-2001). Available at: www.seer.cancer.gov. Released April 2004, based on the November 2002 submission. National Cancer Institute, DCCPS, Surveillance Research Program, Cancer Statistics Branch.  Back to cited text no. 1    
2.Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, et al . Cancer statistics, 2005. CA Cancer J Clin 2005;55:10-30.  Back to cited text no. 2    
3.Dhir V, Mohandas KM. Epidemiology of digestive tract cancers in India IV. Gall bladder and pancreas. Indian J Gastroenterol 1999;18:24-8.  Back to cited text no. 3    
4.Smeenk HG, Tran TC, Erdmann J, van Eijck CH, Jeekel J. Survival after surgical management of pancreatic adenocarcinoma: does curative and radical surgery truly exist? Langenbecks Arch Surg 2005;390:94-103. Epub 2004 May 14.  Back to cited text no. 4    
5.Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet 2004;363:1049-57.  Back to cited text no. 5    
6.Steer ML, Waxman I, Freedman S. Chronic Pancreatitis. New Eng J Med 1995;332:1482-90.  Back to cited text no. 6    
7.Comfort A, Gambill E, Bagenstoss AH. Chronic relapsing pancreatitis: A study of 29 cases without associated diseases of the biliary or gastrointestinal tract. Gastroenterology 1968;54:760-65.  Back to cited text no. 7    
8.Cohn JA, Friedman KJ, Noone PG, Knowles MR, Silverman LM, Jowell PS. Relation between Mutations of the Cystic Fibrosis Gene and Idiopathic Pancreatitis. N Engl J Med 1998;339:653-8.  Back to cited text no. 8    
9.Cohn JA, Jowell PS. Are mutations in the cystic fibrosis gene important in chronic pancreatitis? Surg Clin North Am 1999;79:723-31.  Back to cited text no. 9    
10.Choudari CP, Lehman GA, Sherman S. Pancreatitis and cystic fibrosis gene mutations. Gastroenterol Clin North Am 1999;28:543-9.  Back to cited text no. 10    
11.Taylor CJ. Chronic pancreatitis and mutations of the cystic fibrosis gene. Gut 1999;44:8-9.  Back to cited text no. 11    
12.Pitchumoni CS. Chronic pancreatitis: a historical and clinical sketch of the pancreas and pancreatitis. Gastroenterologist 1998;6:24-33.  Back to cited text no. 12    
13.Ammann R, Sulser H. "Senile" chronic pancreatitis; a new nosologic entity? Studies in 38 cases. Indications of a vascular origin and relationship to the primarily painless chronic pancreatitis. Schweiz Med Wochenschr 1976;106:429-37.  Back to cited text no. 13    
14.Nwokolo C, Oli J. Pathogenesis of juvenile tropical pancreatitis syndrome. Lancet 1980;1:456-9.  Back to cited text no. 14    
15.Braganza JM, Schofield D, Snehalatha C, Mohan V. Micronutrient antioxidant status in tropical compared with temperate-zone chronic pancreatitis. Scand J Gastroenterol 1993;28:1098-104.   Back to cited text no. 15    
16.Chandak GR, Idris MM, Reddy DN, Bhaskar S, Sriram PV, Singh L. Mutations in the pancreatic secretory trypsin inhibitor gene (PSTI/SPINK1) rather than the cationic trypsinogen gene (PRSS1) are significantly associated with tropical calcific pancreatitis. J Med Genet 2002;39:347-51.  Back to cited text no. 16    
17.Rossi L, Pfützer RH, Parvin S, Ali L, Sattar S, Kahn AK, et al. SPINK1/PSTI mutations are associated with tropical pancreatitis in Bangladesh. A preliminary report. Pancreatology 2001;1:242-5.  Back to cited text no. 17    
18.Buchler MW, Friess H, Uhl W, Malfertheiner P eds. Chronic Pancreatitis: Novel concepts in biology and therapy. Blackwell Publishing 2002. p. 27-9.  Back to cited text no. 18    
19.Lowenfels AB, Maisonneuve P, Cavallini G, Ammann RW, Lankisch PG, Andersen JR, et al . Pancreatitis and the risk of pancreatic cancer. N Eng J Med 1993;328:1433-7.  Back to cited text no. 19    
20.Augustine P, Ramesh H. Is tropical pancreatitis premalignant? Am J Gastroenterol 1992;87:1005-8.  Back to cited text no. 20    
21.Chari ST, Mohan V, Pitchumoni CS, Viswanathan M, Madanagopalan N, Lowenfels AB. Risk of pancreatic carcinoma in tropical calcifying pancreatitis: an epidemiologic study. Pancreas 1994;9:62-6.  Back to cited text no. 21    
22.Chari ST, Mohan V, Pitchumoni CS, Viswanathan M, Madanagopalan N, Lowenfels AB. Risk of pancratic carcinoma in tropical calcifying pancreatitits: an epidemicologic study. Pancreas 1994;9:62-6.  Back to cited text no. 22    
23.Sheldon CD, Hodson ME, Carpenter LM, Swerdlow AJ. A cohort study of cystic fibrosis and malignancy. Br J Cancer 1993;68:1025-8.  Back to cited text no. 23    
24.Neglia JP, FitzSimmons SC, Maisonneuve P, Schöni MH, Schöni-Affolter F, Corey M, et al . The risk of cancer among patients with cystic fibrosis. Cystic Fibrosis and Cancer Study Group. N Engl J Med 1995;332:494-9.  Back to cited text no. 24    
25.Howes N, Lerch MM, Greenhalf W, Stocken DD, Ellis I, Simon P, et al. European registry of hereditary pancreatitis and pancreatic cancer (EUROPAC). Clinical and genetic characteristics of hereditary pancreatitis in Europe. Clin Gastroenterol Hepatol 2004;2:252-61.  Back to cited text no. 25    
26.Lowenfels AB, Maisonneuve P, Whitcomb DC, Lerch MM, DiMagno EP. Cigarette smoking as a risk factor for pancreatic cancer in patients with hereditary pancreatitis. JAMA 2001;286:169-70.  Back to cited text no. 26    
27.Farrow B, Evers BM. Inflammation and the development of pancreatic cancer. Surg Oncol 2002;10:153-69.  Back to cited text no. 27    
28.Whitcomb DC. Inflammation and cancer. Chronic pancreatitis and pancreatic cancer. Am J Physiol Gastrointest Liver Physiol 2004;287:G315-9.   Back to cited text no. 28    
29.Vakkila J, Lotze MT. Inflammation and necrosis promote tumor growth. Nat Rev Immunol 2004;4:641-8.  Back to cited text no. 29    
30.Shrikhande SV, Martignoni ME, Shrikhande M, Kappeler A, Ramesh H, Zimmermann A, et al . Comparison of histological features and inflammatory cell reaction in alcoholic, idiopathic and tropical chronic pancreatitis. Br J Surg 2003;90:1565-72.  Back to cited text no. 30    
31.Hruban RH, Adsay NV, Albores-Saavedra J, Compton C, Garrett ES, Goodman SN, et al . Pancreatic intraepithelial neoplasia: a new nomenclature and classification system for pancreatic duct lesions. Am J Surg Pathol 2001;25:579-86.  Back to cited text no. 31    
32.Hruban RH, Takaori K, Klimstra DS, Adsay NV, Albores-Saavedra J, Biankin AV, et al . An illustrated Consensus on the Classification of Pancreatic Intraepithelial Neoplasia and Intraductal Papillary Mucinous Neoplasms. Am J Surg Pathol 2004;28:977-87.  Back to cited text no. 32    
33.Van Laethem JL, Bourgeois V, Parma J, Delhaye M, Cochaux P, Velu T, et al . Relative contribution of Ki-ras gene analysis and brush cytology during ERCP for the diagnosis of biliary and pancreatic diseases. Gastrointest Endosc 1998;47:479-85.  Back to cited text no. 33    
34.Talar-Wojnarowska R, Malecka-Panas E. Molecular pathogenesis of pancreatic adenocarcinoma: Potential clinical implications. Med Sci Monit 2006;12:RA186-93.  Back to cited text no. 34    
35.Shrikhande SV, Friess H, Kleeff J, Büchler MW. Genetic basis of diseases of the pancreas. In: GI Surgery Annual by TK Chattopadhyay (ed)., New Delhi 2004.  Back to cited text no. 35    
36.Klöppel G, Adsay NV. Chronic pancreatitis and the differential diagnosis versus pancreatic cancer. Arch Pathol Lab Med 2009;133:382-7.  Back to cited text no. 36    
37.Bloomston M, Frankel WL, Petrocca F, Volinia S, Alder H, Hagan JP, et al . MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA 2007;297:1901-8.  Back to cited text no. 37    
38.Crnogorac-Jurcevic T, Gangeswaran R, Bhakta V, Capurso G, Lattimore S, Akada M, et al . Proteomic analysis of chronic pancreatitis and pancreatic adenocarcinoma. Gastroenterology 2005;129:1454-63.   Back to cited text no. 38    
39.Beger HG, Schlosser W, Friess HM, Büchler MW. Duodenum-preserving head resection in chronic pancreatitis changes the natural course of the disease: a single-center 26-year experience. Ann Surg 1999;230:512-9; discussion 519-23.  Back to cited text no. 39    
40.Evans JD, Morton DG, Neoptolemos JP. Chronic pancreatitis and pancreatic carcinoma. Postgrad Med J 1997;73:543-8.  Back to cited text no. 40    
41.Lankisch PG, Mahlke R, Graf D, Becher R, Riesner K. Painless acute pancreatitis mimicking pancreatic carcinoma. Pancreas 1995;10:413-4.  Back to cited text no. 41    
42.Prinz RA. Surgical options in chronic pancreatitis. Int J Pancreatol 1993;14:97-105.  Back to cited text no. 42    
43.Jones BA, Langer B, Taylor BR, Girotti M. Periampllary tumors. Which ones should be resected? Am J Surg 1985;149:46-52.  Back to cited text no. 43    
44.Taylor B. Carcinoma of the head of the pancreas versus chronic pancreatitis: Diagnostic dilemma with significant consequences. World J Surg 2003;27:1249-57.  Back to cited text no. 44    
45.Warren KW, Choe DS, Plaza J, Relihan M. Results of radical resection for periampullary cancer. Ann Surg 1975;181:534-40.  Back to cited text no. 45    
46.Herlyn M, Sears HF, Steplewski Z, Koprowski H. Monoclonal antibody detection of a circulating tumor-associated antigen. I. Presence of antigen in sera of patients with colorectal, gastric, and pancreatic carcinoma. J Clin Immunol 1982;2:135-40.  Back to cited text no. 46    
47.Ni XG, Bai XF, Mao YL, Shao YF, Wu JX, Shan Y, et al . The clinical value of serum CEA, CA19-9, and CA242 in the diagnosis and prognosis of pancreatic cancer. Eur J Surg Oncol 2005;31:164-9.  Back to cited text no. 47    
48.Grem J. The prognostic importance of tumor markers in adenocarcinomas of the gastrointestinal tract. Curr Opin Oncol 1997;9:380-7.  Back to cited text no. 48    
49.Safi F, Roscher R, Beger HG. The clinical relevance of the tumor marker CA 19-9 in the diagnosing and monitoring of pancreatic carcinoma. Bull Cancer 1990;77:83-91.  Back to cited text no. 49    
50.Safi F, Roscher R, Bittner R, Schenkluhn B, Dopfer HP, Beger HG. High sensitivity and specificity of CA 19-9 for pancreatic carcinoma in comparison to chronic pancreatitis. Serological and immunohistochemical findings. Pancreas 1987;2:398-403.  Back to cited text no. 50    
51.Niederau C, Grendell JH. Diagnosis of pancreatic carcinoma. Imaging techniques and tumor markers. Pancreas 1992;7:66-86.  Back to cited text no. 51    
52.Andersen HB, Effersøe H, Tjalve E, Burcharth F. CT for assessment of pancreatic and periampullary cancer. Acta Radiol 1993;34:569-72.  Back to cited text no. 52    
53.Megibow AJ, Zhou XH, Rotterdam H, Francis IR, Zerhouni EA, Balfe DM, et al. Pancreatic adenocarcinoma: CT versus MR imaging in the evaluation of resectability - report of the Radiology Diagnostic Oncology Group. Radiology 1995;195:327-32.   Back to cited text no. 53    
54.Lomanto D, Pavone P, Laghi A, Panebianco V, Mazzocchi P, Fiocca F, et al . Magnetic resonance-cholangiopancreatography in the diagnosis of biliopancreatic diseases. Am J Surg 1997;174:33-8.  Back to cited text no. 54    
55.Trede M, Rumstadt B, Wendl K, Gaa J, Tesdal K, Lehmann KJ, et al . Ultrafast magnetic resonance imaging improves the staging of pancreatic tumors. Ann Surg 1997;226:393-405; discussion 405-7.  Back to cited text no. 55    
56.Jadvar H, Fischman AJ. Evaluation of pancreatic carcinoma with FDG-PET. Abdom Imaging 2001;26:254-9.  Back to cited text no. 56    
57.Lan XL, Zhang YX, Wu ZJ, Jia Q, Wei H, Gao ZR. The value of dual time point (18)F-FDG PET imaging for the differentiation between malignant and benign lesions. Clin Radiol 2008;63:756-64.  Back to cited text no. 57    
58.Bang S, Chung HW, Park SW, Chung JB, Yun M, Lee JD, et al. The clinical usefulness of 18-fluorodeoxyglucose positron emission tomography in the differential diagnosis, staging, and response evaluation after concurrent chemoradiotherapy for pancreatic cancer. J Clin Gastroenterol 2006;40:923-9.  Back to cited text no. 58    
59.Afify AM, al-Khafaji BM, Kim B, Scheiman JM. Endoscopic ultrasound-guided fine needle aspiration of the pancreas. Diagnostic utility and accuracy. Acta Cytol 2003;47:341-8.  Back to cited text no. 59    
60.Horwhat JD, Paulson EK, McGrath K, Branch MS, Baillie J, Tyler D, et al . A randomized comparison of EUS-guided FNA versus CT or US -guided FNA for the evaluation of pancreatic mass lesions. Gastrointest Endosc 2006;63:966-75.  Back to cited text no. 60    
61.Fritscher-Ravens A, Brand L, Knöfel WT, Bobrowski C, Topalidis T, Thonke F, et al . Comparison of endoscopic ultrasound-guided fine needle aspiration for focal pancreatic lesions in patients with normal parenchyma and chronic pancreatitis. Am J Gastroenterol 2002;97:2768-75.  Back to cited text no. 61    
62.Yusuf TE, Bhutani MS. Differentiating pancreatic cancer from pseudotumorous chronic pancreatitis. Curr Gastroenterol Rep 2002;4:135-9.  Back to cited text no. 62    
63.Barthet M, Portal I, Boujaoude J, Bernard JP, Sahel J. Endoscopic ultrasonographic diagnosis of pancreatic cancer complicating chronic pancreatitis. Endoscopy 1996;28:487-91.  Back to cited text no. 63    
64.Graham RA, Bankoff M, Hediger R, Shaker HZ, Reinhold RB. Fine-needle aspiration biopsy of pancreatic ductal adenocarcinoma: loss of diagnostic accuracy with small tumors. J Surg Oncol 1994;55:92-4.  Back to cited text no. 64    
65.Moller K, Papanikolaou IS, Toermer T, Delicha EM, Sarbia M, Schenck U, et al . EUS-guided FNA of solid pancreatic masses: High yield of 2 passes with combined histologic-cytologic analysis. Gastrointest Endosc 2009; Epub  Back to cited text no. 65    
66.Klöppel G, Lingenthal G, Klapdor R, Kern HF, Rückert K, von Bülow M. Morphologic criteria of the growth behavior of pancreatic cancers. Experimental and clinico-pathologic studies. Dtsch Med Wochenschr 1984;109:702-8.  Back to cited text no. 66    
67.Keighley MR, Moore J, Thompson H. The place of fine needle aspiration cytology for the intraoperative diagnosis of pancreatic malignancy. Ann R Coll Surg Engl 1984;66:405-8.  Back to cited text no. 67    
68.Kamisawa T, Yoshiike M, Egawa N, Nakajima H, Tsuruta K, Okamoto A. Treating patients with autoimmune pancreatitis: Results from a long-term follow-up study. Pancreatology 2005;5:234-8; discussion 238-40. Epub 2005 Apr 22.  Back to cited text no. 68    
69.Camp ER, Vogel SB. Blind Whipple resections for periampullary and pancreatic lesions. Am Surg 2004;70:6-10; discussion 11-12.  Back to cited text no. 69    
70.Sakorafas GH, Tsiotou AG. Multi-step pancreatic carcinogenesis and its clinical implications. Eur J Surg Oncol 1999;25:562-5.   Back to cited text no. 70    
71.Shen J, Person MD, Zhu J, Abbruzzese JL, Li D. Protein expression profiles in pancreatic adenocarcinoma compared with normal pancreatic tissue and tissue affected by pancreatitis as detected by two-dimensional gel electrophoresis and mass spectrometry. Cancer Res 2004;64:9018-26.  Back to cited text no. 71    

Copyright 2009 - Indian Journal of Cancer


The following images related to this document are available:

Photo images

[cn09070f2.jpg] [cn09070f1.jpg] [cn09070f3.jpg]
Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil