The future of surgery for pancreatic cancer

The future of surgery for pancreatic cancer

Annals of Oncology 10 Suppl. 4: S285-S290, 1999. © 1999 Kluwer Academic Publishers. Printed in the Netherlands. Review. The future of surgery for pan...

763KB Sizes 0 Downloads 94 Views

Annals of Oncology 10 Suppl. 4: S285-S290, 1999. © 1999 Kluwer Academic Publishers. Printed in the Netherlands.

Review. The future of surgery for pancreatic cancer J. Slavin, P. Ghaneh, L. Jones, R. Sutton, M. Hartley & J.P. Neoptolemos Department of Surgery, University of Liverpool, Royal Liverpool University Hospital, Liverpool, L69 3GA, UK

Carcinoma of the pancreas has a grim prognosis even following surgical resection. Only a relatively small proportion of patients have a resectable tumour at presentation. At the present time it is uncertain whether the use of radical forms of surgery, or adjuvant therapy improve survival. It is however unlikely that either of these approaches will greatly increase the number of long term survivors. Earlier diagnosis particularly in individuals who are at greater risk of developing carcinoma of the pancreas is one way in which results might be improved. Unfortunately current imaging

Key words: adjuvant treatment, gene therapy, molecular diagnosis, pancreatectomy, pancreatic cancer, prognosis, surgery.

mortality approaching 30% in non-specialist units over the period 1978-1987 [6]. A number of series from specialist centres, however, have shown that it is possible to resect with mortality rates below 6% although morbidity remains high at around 40% [7-12]. A major factor in these improved results has been the development of specialist units with high volume throughput. Comparative studies from the United states and latterly Europe show that when the institutional case volume passes a threshold (approximately 12 pancreatic resections per year) pancreatic surgery can be performed with a relatively low mortality (Table 1) and that units performing fewer than this critical threshold are likely to have significantly increased mortality rates [13-16]. Good results can also be obtained in those of advanced age provided they are selected appropriately [17, 18].

Historical perspective Carcinoma of the pancreas is a relatively common, and highly lethal condition. The median survival of patients with unresected disease is 4-6 months and only about 10% of patients live beyond a year. Resection when this is possible, prolongs survival with a good quality of life but cure is rarely, if ever, achieved [1]. A l e x a n d e r C o d v i l l a undertook the first pancreaticoduodenectomy in 1898 and by 1908, 11 resections had been performed by different surgeons although none of these patients survived beyond the immediate post-operative period [2]. Kausch reported the first successful pancreatectomy in 1912; this operation was performed in two stages, an initial biliary bypass followed by a later resection [3]. In 1934 Whipple described a twostage procedure [4]. Five years later he performed a onestage procedure, which included a pancreatojejunal anastomosis; this operation was reported in 1946 [5]. Whipple's procedure, however, was still associated with such a high mortality that many suggested that it had no place in surgical practice. Even relatively recently an epidemiological study in the West Midlands of 13560 patients with pancreatic cancer revealed a post operative

Diagnosis Most patients present with obstructive jaundice and have an endoscopic retrograde cholangiopancreatogram (ERCP) as part of their diagnostic workup, at which time it is relatively easy to establish internal biliary drainage. Most surgeons are

Table 1: Post-operative mortality and survivalratesafter Kausch-Whipple (KW) and pylorus preserving pancreatoduodenectomy (PPPD) excluding series with patients who have received adjuvant treatment Year

Number of Resections

Post-operative Mortality

5 year survival

Tredeetal[ll]

1989

118

0%

24%

Gall et al [7]

1991

138

6%

16%

Geer et al [95]

1993

146

3.4%

24%

Nitecki et al [96]

1995

186

Russell et al [97]

1996

61

Sperti et al [8]

1996

113

15%

12%

UKPACA[16]

1996

421

5.9%

_

Mosca et al [98]

1997

221

8.2% (KW) 7.0% (PPPD)

Author

3%

7%

_

8%

9.6%

Median survival

11 months _ _ 13 months _

Downloaded from http://annonc.oxfordjournals.org/ at Dalhousie University on May 17, 2015

techniques are inadequate for the diagnosis of early disease. New molecular diagnostics techniques that can identify example mutations in oncogenes such as K-ras or deletions of tumour suppressor genes such as P53 or P16 are being developed. These tumour specific abnormalities are also a target for gene therapy. Surgery alone cannot cure any patient with pancreatic cancer but may in the future in conjunction with these new approaches.

Summary

286

Presymptomatic diagnosis Particular patient groups can now be identified that are at increased risk of pancreatic cancer; these include patients with sporadic chronic pancreatitis and individuals from families with hereditary pancreatitis or familial pancreatic cancer [26, 27]. With current diagnostic modalities by the time a tumor of the pancreas is detected micrometases have occurred. To improve the results of surgery methods of detecting tumours at a far earlier stage are required. The use of molecular markers for the diagnosis and staging of pancreatic disease has shown great promise. K-ras mutations are present in approximately 100% of cases with pancreatic ductal adenocarcinomas. [28, 29]. They have been detected in early lesions (<20mm in size) [30], and also in areas of ductal hyperplasia of the pancreas, suggesting that these mutations occour early in the development of pancreatic ductal adenocarcinoma [31]. K-ras mutations have been detected in free DNA circulating in the peripheral blood of patients with pancreatic ductal adenocarcinomas with a sensitivity of 60-90% and a specificity of 100%; the high detection rate is however only seen in advanced disease [32]. K-ras mutations have also been detected in the stool of patients with pancreatic ductal adenocarcinomas [31]. K-ras mutations of pancreatic duct brushings obtained at ERCP gave a sensitivity of 81% and specificity of 75% for the diagnosis of pancreatic ductal adenocarcinoma [33]. More recent studies using pure pancreatic juice have found that 40% of patients with chronic pancreatitis harbour a K-ras mutation and these patients do not go on to develop pancreatic ductal adenocarcinoma on longer term prospective follow up [34]. Additionally, K-ras mutations

have been detected in micro-dissected foci of pancreatic hyperplasia in patients with no pancreatic disease, suggesting that there may be a low background rate of K-ras mutations in normal pancreas glands [35]. In order to improve diagnostic accuracy, other markers will be used in tandem with K-ras. Mutations or deletions that affect p53, pl6 and SMAD4 are also commonly found in pancreatic ductal adenocarcinomas but not in tissue from patients with chronic pancreatitis [36-38]. Further work is underway to try and detect these mutations/deletions in pancreatic juice. Prognosis using molecular markers Overexpression of growth factors EGF, TGFa, TGFP 1-3, aFGF, bFGF, and growth factor receptors c-erbB-2, c-erbB3, TGFpRI-in is common in pancreatic cancers. High levels of mutations are found in genes which control the cell cycle such as p53, pi6, p21, DPC4, cyclin Dl, abnormal expression of apoptotic genes such as bcl-2, bcl-XL and bax are also found. In general, no significant correlation between overexpression of growth factors and their receptors and patient survival has been found, although they are associated with poorly differentiated tumours of an advanced stage. Inactivation and loss of expression of tumour suppressor genes pi6, p53 and p21 has not been found to be of any prognostic significance [39]. In one study overexpression of cyclin Dl was associated with significantly shorter patient survival but this was not confirmed by another larger study [39, 40]. TGFpi expression has been shown to be associated with significantly longer survival in patients with pancreatic cancer [41]. Bax expression has also been shown to be a significant indicator of prolonged survival in one study but was not confirmed in our own and there is no significant relationship between bcl-2 expression and survival [42,43]. Two studies (including our own) have confirmed bcl-XL expression to be significantly associated with poor outcome and survival in patients with pancreatic cancer [43, 44]. Certain types of K-ras mutation may differentiate between poor and good long-term survival following resection (unpublished data). These results confirm the potential use of molecular markers to predict the outcome of patients with pancreatic cancer. Surgical palliation With the development of endoscopic techniques of biliary bypass and improved preoperative staging the role of open surgical palliation has decreased. Three randomised clinical trials have compared open surgical bypass and endoscopic drainage in patients with distal bile duct obstruction [45-47]. The two methods have a similar initial success rate and overall survival of patients does not seem to differ. The advantages of the endoscopic route are decreased morbidity and possibly mortality in the period immediately following the drainage procedure and a decreased hospital stay with cost savings following stenting [48]. Stent blockage that requires re-ERCP and stenting occurs in a proportion of patients, particularly in those living longer than six months when the alternative is to use the self-expanding metal stents [49]. Randomised controlled studies from Europe [50, 51] and the USA [52] showed that self-expanding metal stents

Downloaded from http://annonc.oxfordjournals.org/ at Dalhousie University on May 17, 2015

convinced that in jaundiced patients a period of biliary drainage prior to a major resection of the pancreatic head is beneficial, and reduces postoperative complications. A randomised controlled trial of preoperative endoscopic biliary drainage versus early surgery failed to demonstarte any reduction in mortality (14% vs 14%) or morbidity (41% vs 37%) following surgery but the study was seriously underpowered [19]. The success of endoscopic biliary drainage also means that it is important to avoid an unnecessary laparotomy in patients with advanced disease through the use of preoperative staging protocols. Laparoscopy will identify macroscopic intraperitoneal spread and may reduce the number of unnecessary laparotomies [20]. The use of ultrasound at the time of laparoscopy [21] orendoscopy may improve assessment of resectability and provide improved imaging of the gland and of associated vasculature [22]. Spiral CT however remains the gold standard for the assessment of tumour, resectability [23], although there are encouraging reports of MRI [24]. A common diagnostic quandary is the need to differentiate chronic pancreatitis from malignancy. Biopsy carries the risk of intra-peritoneal seeding with intra-abdominal spread [25] and because of this is employed only if a lesion is felt to be irresectable or there is metastatic disease. Resection en bloc with later histological analysis is justified on the basis of a low postoperative mortality. Percutaneous biopsy is required for histological diagnosis in advanced pancreatic cancer if treatment with chemotherapy is contemplated.

287 remained patent for longer than plastic stents. Metal stents must be avoided if there is a possibility of tumour excision, s resection then becomes hazardous or impossible. Patients with gastric outlet obstruction have previously required an open surgical bypass; insertion of a metal stent into the duodenum obviates the need for surgery [53]. Laparoscopic cholecystojejunostomy offers another treatment strategy, but cholangiography is essential to confirm that the cystic duct is patent and that the point of insertion into the common bile duct is well away from the tumour [54]. Gastric bypass also may be performed laparoscopically [55]. Standard Kausch-Whipple pancreatectomy

Following the finding by Cubilla et al in 1978 [70] that up to one third of patients had metastases in lymph nodes not usually removed with the standard operation the concept of a radical extended pancreatectomy was introduced by Former [71]. Japanese pancreatic surgeons in the 1980s also developed aggressive strategies to treat pancreatic cancer and developed surgical approaches including extended lymph node and connective tissue clearance with autonomic nerve dissection around the coeliac and superior mesenteric arteries [72]. The results following extended resection from a number of series are shown in Table 2. The radicality of the operation varies from centre to centre and thus it is often difficult to compare results. Most of the series are retrospective, lacking even the dubious benefit of a withincentre comparison of a historical control set - an exception being the study of Ishikawa et al (1988). Moreover, group migration using retrospective data accounts for an apparently better survival in one group compared to another, yet the overall survival is the same. In the case of extended resection there are increased numbers of lymph nodes for histological analysis and the tendency therefore for cases to migrate to a higher stage. This produces an apparent improvement in the survival of early cases which is illussory since the overall survival is the same. Recent large retrospective studies from Japan have shown no benefit following extended lymphadenectomy in patients with more advanced tumours [73-75]. As can be observed from the comparitive results in Tables 1 and 2, that extended resections are unlikely to result in longer patient survival compared to a standard resection. Only two randomised-controlled studies have examined the role of extended lymphadenectomy in this condition. The first is a single centre study, with only a few patients accrued [76]. A randomised controlled study comparing extended lymphadenectomy added to the conventional Kausch-Whipple proceedure involving five centres in Italy and one in the USA showed no significant survival benefit nor in post-operative morbidity or mortality. Analysis of the subgroup of patients who were found to be lymph node positive showed a small significant survival benefit with extended lymphadenectomy. Unfortunately this post-hoc analysis has little scientific meaning [77]. A definitive multi-centre randomised study is required to adequately address the role of extended lymphadenectomy in pancreatic carcinoma [78]. Adjuvant therapy Even after resection survival is short for patients with pancreatic cancer. Any extra gain must not be outweighed excessive to morbidity. Radiotherapy is generally felt to give the best local control in advanced disease but there has never been a randomised controlled study [78]. The use of a 5 flourouracil (5-FU) as a chemosensitising agent with radiotherapy significantly improves survival of patients with advanced disease compared to radiotherapy alone [79]. Polychemotherapy regimens in advanced pancreatic cancer do not have any survival benefit over 5-FU alone but are associated with increased morbidity [80]. The use of systemic 5-FU given as a weekly bolus after radiochemotherapy however was shown to improve survival in advanced pancreatic cancer compared

Downloaded from http://annonc.oxfordjournals.org/ at Dalhousie University on May 17, 2015

There has been confusion about what constitutes a standard Kausch-Whipple operation. A consensus conference attended by a number of International experts was held at Castelfranco Veneto in Italy to resolve this issue in May 1998 [56]. In the standard operation the resected specimen consists of the distal stomach, the duodenum and proximal jejunum, attached bile duct and gallbladder, as well as the head of the pancreas. Lymph nodes (according to the Japanese Pancreatic Society classification) which are removed en bloc during the standard Kausch-Whipple procedure include the anterior pancreatoduodenal nodes, (13a, 13b); lymph nodes on the right side of the hepatoduodenal ligament (12b 1,12b2,12c); the nodes to the right side of the superior mesenteric artery, from the origin of the superior mesenteric artery at the aorta to the inferior pancreaticoduodenal artery (14a,14b); and the anterior pancreaticoduodenal nodes (17a, 17b). The outcome from the Standard Kausch-Whipple pancreatodenectomy is shown in Table 1. Portal vein involvement does not necessarily preclude resection as part of it may be resected [57]. Similarly portal vein involvement does not necessarily indicate a more aggressive tumour as survival following portal vein resection is similar to other tumours of similar stage [58]. Involvement of the mesenteric artery is usually considered to indicate irresectability but combined resection of the coeliac artery with a distal pancreatectomy has been shown to increase the resectability rate with an acceptable post-operative mortality [59]. The exact definition of a resectable tumour is thus to some extent surgeon dependent. Pylorus preserving pancreatoduodenectomy (PPPD) first described by Watson in 1944 [60] does not compromise longer term survival, despite initial fears that this might be the case [61-63]. PPPD may shorten operative time and reduce blood loss [62]. Although preservation of the pyloric sphincter should allow for more controlled emptying of the upper gastrointestinal tract and reduce biliary reflux, whilst preservation of the duodenal pacemaker should reduce delayed gastric emptying and improve late gastro-intestinal functions, clinical studies are inconclusive [62,64]. Total pancreatectomy was advocated at one time in an attempt to reduce recurrence rates and postoperative morbidity, particularly leakage from the pancreatic anastomosis [65-67]. Unfortunately following total pancreatectomy patients become diabetic and a number of studies have failed to confirm the procedure's oncological benefits, reduced morbidity or mortality. It is now used infrequently [68, 69].

Extended lymphadenectomy

288 Table 2: Results of radical surgery and extended lvmphadenectomv for pancreatic adenocarcinoma Series

Period (19..)

Number of resections

Resection Type

Operative mortality

Median Survival

1 year survival

3 year survival

5 year survival

Former [99]

72-78 79-88

35 33

Regional Regional

9 (26%) 3(9%)

15 months

-

-

Manabe et al [100]

66-87 66-87

42 32

S E

4(10%) 2(6%)

-

27% 65%

0% 38%

0% 33%

Hiraokaetal[101]

66-75 66-75

19 9

S E

3 (16%) 1(11%)

-

27% 44%

0% 12%

0% 0%

Ishikawaetal [102]

71-81 81-89

45 72

E R

6(13%) 3(4%)

-

39% 59%

10% 36%

8% 27%

Henne-Bruns et al [76]

88-91 88-91

11 5

S E

-

25% 50%

0% 50%

-

Hanyu et al [103]

68-95 68-95

67 249

S E

8 (12%) 10 (4%)

-

41% 42%

10% 10%

8% 8%

Naganuma et al [104]

76-96 76-96

11 71

S R

0(%) 1 (1.4%)

-

27% 48%

9% 29%

0% 13%

Pedrazzoli et al [77]

91-94 91-94

40 41

S E

2 2

18.4 months 19.6 months

-

-

-

37%

to radiotherapy alone [81]. Subsequently, a randomised controlled trial of adjuvant chemo-irradiation with follow-on chemotherapy (weekly injections of 5-FU for two years) in patients undergoing potentially curative resection of the pancreas was performed. The control group received no treatment. The trial was eventually terminated because of poor recruitment. At this time results showed a doubling in the median and 2 year survival in the treatment group but there were only 43 patients in the whole study. A further group of 30 patients were subsequently recruited to the same treatment protocol; although survival was similar to that seen in the original treatment group the problems of an unconvincing underpowered study remained [82]. A number of studies using similar protocols of chemotherapy/radiation have since been undertaken in an attempt to confirm these findings. The European Organization for Research and Treatment of Cancer (EORTC) randomised study of postoperative radiotherapy showed a non-significant benefit in the treated group, perhaps because the trial was underpowered [83]. A subsequent EORTC trial is comparing radiotherapy with 5FU post-resection and then randomising to follow on chemotherapy with 5-FU or gemcitabine. Only one trial has examined the role of adjuvant chemotherapy alone in pancreatic cancer. Treatment with a multi-chemotherapy regimen consisting of 5-FU, adriamycin and mitomycin-C resulted in a significant doubling of median survival in comparison to a control group but no increase in 5-year survival. The morbidity associated with this regimen was considerable and few patients actually completed the full course of chemotherapy [84]. The European Study Group for Pancreatic Cancer (ESPAC) initiated a multi-centre adjuvant trial in 1994 (ESPAC-1) randomising patients to one of four groups: control radiochemotherapy, chemotherapy (5-FU) for six months, and radiochemotherapy and chemotherapy (5-FU) [85]. This study has recruited over 500 patients and should definitely answer questions as to the role of conventional adjuvant treatment in pancreatic cancer. It will be some years before the final results from ESPAC-1 are available. Following its conclusion ESPAC-3 will randomise over 700 patients to receive either 5-FU or gemcitabine for six months

in comparison to control the group. Novel approaches are required as the benefits of conventional cytotoxics in pancreatic cancer are limited. Matrix metalloproteinases (MMPs) are a family of zinccontaining proteolytic enzymes that breakdown extracellular matrix proteins (ECM) essential for tumour invasion [86,87] Disruption in the regulation and expression of MMPs indicates an important role of MMPs in the spread of pancreatic cancer [88-90]. Marimastat is a highly potent but reversible inhibitor of MMP2, with weak activity against unrelated MMPs. Patients with unresectable stage II or HI disease treated with marimastat had a median survival of seven months compared to less than three months with stage IV disease [89,91]. Phase i n trials of marimastat and other MMP inhibitors are underway in advanced disease and as adjuvant therapy post resection. These studies include a double-blind randomised study comparing adjuvant chemoradiotherapy with or without marimastat in patients with resectable cancer, and a double-blind randomised study comparing gemcitibine with or without marimastat in patients with non-resectable pancreatic cancer. Gene therapy There are several approaches being tested for the treatment of pancreatic cancer. Gene replacement therapy restores the function of genes such as tumour suppressor genes. In appropriate cells this is pro-apoptotic or promotes cell cycle arrest. The wild type p53, p 16, p21 and retinoblastoma genes have been replaced in pancreatic cancer using either retrovirus or adenovirus vectors [92]. Anti-sense oligonucleotides directed at oncogenic ras have been shown to reduce tumourigenicity and anti-sense oligonucleotides directed at c-ErbB-2 decrease cancer cell growth. Genetic prodrug activation therapy (GPAT) involves transferring a gene encoding a specific enzyme into cancer cells which will convert a non-toxic prodrug into a cytotoxic metabolite. Neighbouring tumour cells and also distant tumour cells that do not express the enzyme may also die due to bystander effects. Several systems have been assessed such as the combination of P-nitroreductase and treatment with CB1954 (which is converted to powerful alkylating agents) [93]. Immunotherapy approaches, which utilise cytokine genes

Downloaded from http://annonc.oxfordjournals.org/ at Dalhousie University on May 17, 2015

S standard, E extended, R radical

289 such as IL2 or IL12, have been used alone or in combination with GPAT [94]. The principal limitation of gene therapy at present is targeting vectors to the appropriate tissue. The best way to introduce genes into mammalian cells is using disabled viruses. Unfortunately this often provokes an immune response. Tumour-specific targeting is not possible at present although the development of modified viruses or 'synthetic' viruses is ongoing. Tumour-specific expression is however possible by linking the promoter of a tumourspecific gene (such as CEA) in the gene constructs. Refinement of these strategies in conjunction with the development of novel techniques of delivery will allow the selective targeting of tumour cells.

17. 18.

19. 20. 21.

22.

23. 24.

At the present there are nearly 200,000 new cases of pancreatic cancer each year in the world. Most are dead within one year and surgery cures no patient. Outside of specialist centres the resection rates are less than 3% and the mortality is at least 20-30%. The type of surgery is poorly documented and there is no agreed standard of pathological reporting. Advanced cancer or adjuvant therapies have yet to be shown to be of benefit. In the New Millennium surgery will become a key component of treatment. • Management of cases will be in regional centres achieving resection rates of 30-50%. • The type of surgery and pathological reporting will be standardised. • Treatment will be multimodel hinging on a variety of gene therapies. • Molecular diagnosis will be in routine use and molecular prognosis will determine the type of treatment to be given to an individual patient.

25.

References 1.

2. 3. 4. 5. 6.

7. 8. 9. 10.

11. 12. 13. 14. 15.

16.

Gudjonsson B. Carcinoma of the pancreas: critical analysis of costs, results of resections, and the need for standardized reporting. J Am Coll Surg 1995; 181:483-503. Sauve L. Des pancreaectomies et specialement de la pancreatectomie cephalique. Rev Chir 1908; 37: 335-85. Kausch W. Carcinom der Papilla Duodeni und seine radikale Entfernung. Beitr Klin Chir 1912; 78:439-86. Whipple AO, Parsons WB, Mullens CR. Treatment of carcinoma of the the ampulla of Vater. Ann Surg 1935; 102: 763-79. Whipple AO. Observations on radical surgery for lesions of pancreas. Surg Gynecol Obstet 1946; 82: 623-31. Bramhall SR, Allum WH, Jones AG et al. Treatment and survival in 13,560 patients with pancreatic cancer and incidence of the disease in the West Midlands: an epidemiologies] study. Br J Surg 1995; 57: 111-5. Gall FP, Kessler H, Hermanek P. Surgical treatment of ductal pancreatic carcinoma. Eur J Surg Oncol 1991; 17: 173-81. Sperti C, Pasquali C, Piccoli A et al. Survival after resection for ductal adenocarcinoma of the pancreas. Br J Surg 1996; 83:625-31. Grace PA, Pitt HA, Tompkins RK et al. Decreased mortality and morbidity after pancreaticoduodenectomy. Am J Surg 1986; 151: 141-9. Crist DW, Sitzmann JV, Cameron JL. Improved hospital morbidity, mortality, and survival after the Whipple procedure. Ann Surg 1987; 206: 358-65. TredcM. Survival after pancreatoduodenectomy: 118consecutiveresections without an operative mortality. Ann Surg 1990; 211:447-58. Yeo JC, Cameron JL, Sohn TA et al. Six hundred fifty consecutive pancreaticoduodenectomies in the 1990s. Ann of Surg 1997; 226: 248-60. Sosa JA, Bowman HM, Gordon TA et al. Importance of hospital volume in the overall management of pancreatic cancer. Ann Surg 1998; 228:429-38. Glasgow RE, Mulvihiil SJ. Hospital volume influences outcome in patients undergoing pancreatic resection for cancer. West J Med 1996; 165:294-300. Lieberman MD, Kilbum H, Lindsey M et al. Relation of perioperative deaths to hospital volume among patients undergoing pancreatic resection for malignancy. Ann Surg 1995; 222: 638-45. Neoptolemos JP, Russell RC, Bramhall S et al. Low mortality following resection for pancreatic and periampullary tumours in 1026 patients: UK

26. 27.

28.

29. 30. 31.

32.

33.

34.

35.

36. 37. 38.

39.

40.

41.

42. 43. 44.

45.

46.

47.

48.

Downloaded from http://annonc.oxfordjournals.org/ at Dalhousie University on May 17, 2015

Surgery for pancreatic cancer in the new millennium

survey of specialist pancreatic units. UK Pancreatic Cancer Group. Br J Surg 1997; 84: 1370-6. DiCarlo V, Balzano G, Zerbi A et al. Pancreatic cancer resection in elderly patients. Br J Surg 1998; 85: 607-10. Magistrelli P, Masetti R, Coppola R et al. Pancreatic resection for periampullary cancer in elderly patients. Hepato-Gastroenterology 1998;45: 242-7. Lai EC, Mok FP, Fan ST et al. Preoperative andoscopic drainage for malignant obstuctive jaundice. BrJ Surg 1994; 81: 1195-8. Warshaw AL, Gu ZY, Wittenberg J et al. Preoperative staging and assessment of resectability of pancreatic cancer. Arch Surg 1990; 125:230-3. Minnard EA, Conlon KL, Hoos A et al. Laparoscopic ultrasound enhances standard laparoscopy in the staging of pancreatic cancer. Ann Surg 1998; 228: 182-7. Legmann P, Vignaux O, Dousset B et al. Pancreatic tumors: comparison of dual-phase helical CT and endoscopic sonography. Am J f Roentgentrerol 1998; 170: 1315-22. McCarthy MJ, Evans J, SagarG et al. Prediction of resectability of pancreatic malignancy by computed tomography. Br J Surg 1998; 85: 320-5. Bluemke DA, Fishman EK. CT and MR evaluation of pancreatic cancer. Surgical Oncology Clinics of North America 1998; 7: 103-24. Warshaw AL. Implications of peritoneal cytology for staging of early pancreatic cancer. Am J Surg 1991; 161: 26-9. Finch M, Howes N, Ellis I et al. Hereditary pancreatitis and familial pancreatic cancer. Digestion 1997; 58: 564-9. Lowenfels AB, Maisonneuve P, Dimango EP et al. Hereditary pancreatitis and the risk of pancreatic cancer. Journal of the National Cancer Institute 1997; 89:442-6. Pellegata NS, Sessa F, Renault B et al. K-ras and p53 gene mutations in pancreatic cancer: ductal and non ductal tumours progress through different genetic lesions. Cancer Research 1994; 54: 1556-60. Hruban RH, van Mansfield ADM, Offerhaus GJA et al. K-ras mutations in adenocarcinoma of the pancreas. Am J Pathol 1993; 143: 545-54. Schaeffer BK, Glasner S, Kuhlmann E et al. Mutated c-K-ras in small pancreatic adenocarcinomas. Pancreas 1994; 9: 161-5. Caldas C, Hahn SA, Hruban RH et al. Detection of K-ras mutations in the stool of patients with pancreatic adenocarcinoma and pancreatic duct hyperplasia. Cancer Research 1994; 54: 3568-73. Yamanda TM, Nakamori S, Ohzato S et al. Detection of K-Ras nutations in plasma DNA of patients with pancreatic adenocarcinoma: Correlation with Clinopathological features. Clinical Cancer Research 1998; 4: 1527-32. Van Laethem JL, Bourgeois V, Parma J et al. Relative contribution of Ki-ras gene analysis and brush cytology during ERCP for the diagnosis of biliary and pancreatic disease. Gastrointestinal Endoscopy 1998; 47:479-85. Furuya N, Kawa S, Akamatsu T et al. Long-term follow up of patients with chronic pancreatitis and k-ras gene mutation detected in pancreatic juice. Gastroenterology 1997; 113: 593-8. Tada M, Ohashi M, Shiratori Y et al. Analysis of K-ras mutation in hyperplastic duct cells of the pancreas without pancreatic disease. Gastroenterology 1996; 110: 227-31. Barton CM, Staddon SL, Hughes CM et al. Abnormalities of the p53 tumour suppressor gene in human pancreatic cancer. BrJ Cancer 1991; 64:1076-82. Hahn SA, Schutte ATM, Hoque S et al. DPC4, A candidate tumour suppressor gene at human chromosome 18q21.1. Science 1996; 271:350-43. Caldas C, Hahn SA, da Costa LT et al. Frequent somatic mutations and homozygous deletions of the pl6 (MTS1) gene in pancreatic adenocarcinoma. Nature Genetics 1994; 8: 27-32. Kawesha A, Ghaneh P, Evans JD et al. Overexpression of C-erbB2, C-crbB3 and cyclin D and loss or inactivation of p53, pl6 and p21WAFl do not correlate with prognosis in pancreatic cancer. Digestion 1998; 59: 222. Gansauge F, Gansauge S, Schmidt E, et al. Prognostic significance of molecular alterations in human pancreatic carcinoma-an immunological study. Lang Arch Surg 1998; 383: 152-5. Coppola D,LuL,Fruehauf JPetal. Analysis of p53,p21 (WAFI)andTGF beta 1 in human ductal adenocarcinoma of the pancreas- TGF beta 1 protein expression predicts longer survival. Am J Gin Pathology 1998; 110:16-23. Friess H, Lu Z, Graber HU el al. Bax but not bcl-2 influences the prognosis of human pancreatic cancer. Gut 1998; 43: 414-21. Evans JD, Comford PA, Dodson A, et al. Expression of bcl-XL correlates with survival in pancreatic cancer. Int J of Pancreatol 1998; 24:72. Friess H, Zhao L, Andren-Sandberg A et al. Moderate activation of the apoptosis inhibitor bcl-XL worsens the prognosis in pancreatic cancer. Ann Surg 1998; 228: 780-7. Smith AC, Dowsett JF, Russell RC et al. Randomised trial of endoscopic stenting versus surgical bypass in malignant low bile duct obstruction. Lancet 1994; 344: 1655-60. Anderson JR, Sorensen SM, Kruse A et al. Randomised trial of endoscopic endoprosthesis versus operative bypass in malignant obstructive jaundice. Gut 1989; 30: 1132-5. Shepard AH, Royle G, Ross APR. Endoscopic biliary endoprosthesis in the palliation of malignant obstruction of the distal bile duct: A randomised trial. BrJ Surg 1988; 75: 1166-9. Raiker GK, Melin MN, Ress A et al. Cost effectiveness of surgical palliation versus endoscopic stenting in the management of unresectable pancreatic cancer. Ann Surg Oncol 1996; 3: 470-5.

290 49. 50.

51.

52.

53.

54.

55.

56. 57. 58.

60. 61.

62.

63. 64.

65. 66. 67. 68.

69.

70. 71.

72. 73.

74.

75. 76.

77.

78.

79.

80.

81.

82.

83. 84.

85.

86. 87.

88.

89. 90.

91.

92.

93.

94.

95. 96.

97.

98.

99.

100. 101.

102.

103.

104.

supervoltage radium therapy of locally unresectable gastrointestinal cancer. Lancet 1969; ii: 865-7. Cullinan S, Moertel CG, Wieand HS et al. A phase III trial on the therapy of advanced pancreatic carcinoma: evaluation of the Mallinson regime and combined 5-flourouracil, doxorubicin and cisplatinum. Cancer 1990; 65: 2207-12. Gastrointestinal Tumour Study Group. Therapy of locally unresectable pancreatic carcinoma: a randomised comparison of high dose (6,000 rads) radiation alone, moderate dose radiation (4,000 rads) and high dose radiation and 5-flourouracil. Cancer 1981; 48: 1705-10. Gastrointestinal Tumour Study Group. Further evidence of effective adjuvant combined radiation and chemotherapy following curative resection of pancreatic cancer. Cancer 1987; 59: 2006-10. Jeekel J. Adjuvant or neoadjuvant therapy for pancreatic carcinoma? Digestion 1997; 58: 533-5. Bakkevold KE, Arneso B, Dhal O et al. Adjuvant combination chemotherapy (AMF) following radical resection of carcinoma of the pancreas and papilla of Vater;resultsof a prospective, randomised multi-center study. Eur J Surg 1993; 29: 698-703. Neoptolemos JP, Kerr DJ, Beger H et al. ESPAC-1 trial progress report: the European randomized adjuvant study comparing radiochemotherapy, 6 months chemotherapy and combination therapy versus observation in pancreatic cancer. Digestion 1997; 58: 570-7. Matrisan L. Metalloproteinases and their inhibitors in matrix remodelling. Trends in Genetics 1990; 6: 1-8. Liotta LA, Steeg PS, Stetler-Stevenson WG. Cancer metastasis and angiogenesis: an imbalance of positive and negative regulation. Cell 1991 1991; 64: 327-36. Mollenhauer J, Roether I, Kem HF. Distribution of extracellular matrix proteins in pancreatic ductal adenocarcinoma and its influence on tumour cell proliferation in vitro. Pancreas 1987; 2: 14-24. Evans JD, Ghaneh P, Kawesha A et al. Role of matrix metalloproteinases and their inhibitors in pancreatic cancer. Digestion 1997; 58: 520-8. Bramhall S, Neoptolemos JP, Stamp G et al. Inbalance of expression of matrix metalloproteinases (MMPs) in human pancreatic cancer. J Pathology 1997; 182:347-55. Evans JD, Bramhall SR, Carmichael J et al. A phase II trial of marimastat (BB-2516) in advanced pancreatic cancer. Ann Oncol 7 1996; Supple 5: A239P. Bouvet M, Bold RJ, Lee J et al. Adenovirus mediated wild type p53 tumour suppressor gene therapy induces apoptosis and suppresses growth of human pancreatic cancer. Ann Surg Oncol 1998; 5: 681-688. Green NK, Youngs DJ, Neoptolemos JP et al. Sensitization of colorectal and pancreatic cancer cell lines to the prodrug 5-(aziridin-l-yl)-2,4dinitrobenzamide (CB1954) by retroviral transduction and expression of the E. coli nitroreductase gene. Cancer Gene Ther 1997; 4: 229-38. Yoshida Y, Tasaki K, Kimurai M et al. Antitumor effect of human pancreatic cancer cells transduced with cytokine genes which activate Thl helper T cells. Anticancer Res 1998; 18: 333-5. Geer RJ, Brennan MF. Prognostic indicators of survival after resection of pancreatic adenocarcinoma. Am J Surg 1993; 165: 68-72. Nitecki SS, Sarr MG, Colby TV et al. Long-term survival after resection for ductal adenocarcinoma of the pancreas. Is it really improving? Ann Surg 1995; 221: 59-66. Russell RCG. Unusual pancreatic tumours. In Neoptolemos JP, Lemoine NR, eds. Pancreatic Cancer Molecular and Clinical Advances, Vol. 308-207. Oxford: Blackwell Science Publications, 1996. Mosca F, Giulianotti PC, Balestracci Tet al. Long-term survival in pancreatic cancer pylorus-preserving versus Whipple pancreatoduodenectomy. Surgery 1997; 122:553-66. Former JG. Regional pancreatectomy for cancer of the pancreas, ampulla, and otherrelatedsites. Tumor staging andresults.Ann Surg 1984; 199:41825. Manabe T, Ohshio G, Baba N et al. Radical pancreatectomy for ductal cell carcinoma of the head of the pancreas. Cancer 1989; 64: 1132-7. Hiraoka T. Extended radical resection of cancer of the pancreas with intraoperative radiotherapy. Baillieres Clinical Gastroenterology 1990; 4: 985-93. Ishikawa O, Ohhigashi H, Sasaki Y et al. Practical usefulness of lymphatic and connective tissue clearance for the carcinoma of the pancreas head. Ann Surg 1988; 208: 215-20. Hanyu F, Imaizumi T, Suzuki N et al. ExtendedradicalWhipple resection for carcinoma of the head of the pancreas. In: Reber HA, ed. Pancreatic carcinoma. New Jersey: Humana Press Inc, 1998: 201-12. Naganuma T, Kararada Y. Extended pancreatic resection for carcinoma of the pancreas. In: Reber HA, (Ed) Pancreatic Carcinoma. New Jersey: Humana Press Inc, 1998: 213-33.

Correspondence to: Professor J.P. Neoptolemos Department of Surgery University of Liverpool 5th floor UCD Daulby Street Liverpool, L69 3GA United Kingdom

Downloaded from http://annonc.oxfordjournals.org/ at Dalhousie University on May 17, 2015

59.

Dotter CT. Transluminally-placed coilspring endarterial tube grafts. Invest Radiol 1969; 4: 329. Davids PH, Groen AK, Rauws EAJ et al. Randomised trial of self expanding metal scents for distal malignant biliary obstruction. Lancet 1992; 340:148892. Knyrim K, Wagner HJ, Pausch J et al. A prospective, randomised controlled trial of metal stents for malignant obstruction of the bile duct. Endoscopy 1993; 25: 207-12. Carr-Locke DL, Ball TJ, Conners PJ. Multicenter randomised trial of Wallstent biliary prosthesis versus plastic stents. Gastrointest Endosc 1993; 39:310. Keymling M, Wagener JH, Vakil N et al. Relief of malignant duodenal obstruction by percutaneous insertion of a metal stem. Gastrointest Endosc 1993; 39: 439-41. Tamasky PR, England RE, Lail LM et al. Cystic duct patency in malignant obstructive jaundice; An ERCP based study relevant to the role of laparoscopic cholecystojejunostomy. Ann Surg 1995; 221:265-71. Bergamaschi R. Marvik R, Thoresen JE et a]. Open versus laparoscopic gastrojejunostomy for palliation in advanced pancreatic cancer. Surg Lap Endosc 1998; 8: 92-6. Jones L, Russell P, Mosca F et al. The Standard Kausch-Whipple Pancreatoduodenectomy. Digestive Surgery 1999 (in press). Harrison LE, Brennan MF. Portal vein resection for pancreatic adenocarcinoma. Surg Oncol Clin N Am 1998; 7: 165-81. Leach SD, Lee JE, Chamsangavej C et al. Survival following pancreaticoduodenectomy with resection of the superior mesenteric-portal vein confluence for adenocarcinoma of the pancreatic head. Br J Surg 1998; 85:611-7. Mayumi T, Nimura Y, Kamiya J et al. Distal pancreatectomy with en bloc resection of the celiac artery for carcinoma of the body and tail of the pancreas. Int J of Pancreatol 1997; 22: 15-21. Watson K. Carcinoma of the ampulla of Vater: successful radical resection. BrJ Surg 1944; 31: 368-73. Roder JD, Stein HJ, Huttl W et al. Pylorus-preserving versus standard pancreatico-duodenectomy: an analysis of 110 pancreatic and periampullary carcinomas. BrJ Surg 1992; 79: 152-5. -Klinkenbijl JH, van der Schelling GP, Hop WC et al. The advantages of pylorus-preserving pancreatoduodenectomy in malignant disease of the pancreas and periampullary region. Ann of Surg 1992; 216: 142-5. Grace PA, Pitt HA, Longmire WP. Pylorus preserving pancreatoduodenectomy: an overveiw. BrJ Surg 1990; 77: 968-74. van Berge Henegouwen MI, Moojen TM, van Gulik TM et al. Postoperative weight gain after standard Whipple's procedure versus pylorus-preserving pancreatoduodenectomy: the influence of tumour status. BrJ Surg 1998; 85: 922-6. Brooks JR, Culebras JM. Cancer of the pancreas. Palliative operation, Whipple procedure, or total pancreatectomy? Am J Surg 1976; 131:516-20. Ihse I, Lilja P, Arnesjo B, Bengmark S. Total pancreatectomy for cancer. An appraisal of 65 cases. Ann Surg 1977; 186: 675-80. Moossa AR, Lewis MH, Mackie CR. Surgical treatment of pancreatic cancer. Mayo Clinic Proceedings 1979; 54: 468-74. van Heerden JA. Pancreatic resection for carcinoma of the pancreas: Whipple versus total pancreatectomy~an institutional perspective. World Journal of Surgery 1984; 8: 880-8. van Heerden JA, Mcllrath DC, Ilstrup DM et al. Total pancreatectomy for ductal adenocarcinoma of the pancreas: an update. World Journal of Surgery 1988; 12: 658-62. Cubilla AL, Former J, Fitzgerald PJ. Lymph node involvement in carcinoma of nthe head of the pancreas. Cancer 1978; 4: 880-7. Former JG, Kim DK, Cubilla A et al. Regional pancreatectomy: en bloc pancreatic, portal vein and lymph node resection. Ann Surg 1977; 186:4250. Nimura Y. Pancreatic surgery: cutting-edge developments and technology. Pancreas 1998; 16: 227-32. Mukaiya M. Hirata K, Satoh T et al. Lack of survival benefit of extended lymph node dissection for ductal adenocarcinoma of the head of the pancreas: retrospective multi-institutional analysis in Japan. World Journal of Surgery 1998; 22: 248-52. Hirata K, Sato T, Mukaiya M et al. Results of 1001 pancreatic resections for invasive ductal adenocarcinoma of the pancreas. Arch Surg 1997; 132:7716. Nagakawa T, Nagamori M, Futakami F et al. Results of extensive surgery for pancreatic carcinoma. Cancer 1996; 77: 640-5. Henne-Bruns D, Kremer B, Meyer-Pannwitt U et al. Partial duodenopancreatectomy with radical lymphadenectomy in patients with pancreatic and periampullary carcinomas: initial results. HepatoGastroenterology 1993; 40: 145-9. Pedrazzoli S, DiCarlo V, Dionigi R et al. Standard versus extended lymphadenectomy associated with pancreaticoduodenectomy in the surgical treatment of adenocarcinoma of the head of the pancreas. Ann Surg 1998; 228:508-17. Neoptolemos JP, Bramhall S, Kerr DJ. Adjuvant treatment in pancreatic cancer combined treatment modalities. In: Beger HG, Warshaw A, CarrLocke DC, Russell RCG, Buchler M, Neoptolemos JP, Sarr M (Eds) The Pancreas. Boston, Blackwell Scientific 1998: 1104-19. Moertel CG, Childs DS, Reitermeier RJ et al. Combined 5-flourouracil and