in pancreatic cancer

8 downloads 171765 Views 138KB Size Report
for both the health service and the pharmaceutical industry is to harness ..... tumours than can be given by external beam tech- ... causative agents and mechanics underlying genetic ... anchorage independent manner168 through an auto-.
Aliment Pharmacol Ther 1998; 12: 949±964.

Review article: current treatment and optimal patient management in pancreatic cancer A. HAYCOX*, M. LOMBARD , J. NEOPTOLEMOS  & T . W ALLEY* Prescribing Research Group, Department of Pharmacology and Therapeutics, University of Liverpool; and  Pancreatic Research Group, University of Liverpool & Royal Liverpool University Hospital, Liverpool, UK Accepted for publication 26 May 1998

SUMMARY

This review analyses the current state of knowledge and understanding concerning the optimum treatment and therapeutic management of patients who suffer from pancreatic cancer. It outlines recent advances in scienti®c understanding and assesses their potential future value to clinicians in confronting this disease. Despite a signi®cant expansion in scienti®c knowledge relating to factors underlying the early development of pancreatic carcinoma, the clinician continues to be restricted to a severely limited therapeutic armoury for this disease. Local therapies (surgery and radiation) are inevitably of limited value in the face of a disease that is normally encountered at a stage where metastasis is already highly developed. Despite such limitations, however, surgery performed in specialist units may be of value for 10±20% of patients, with a 5-year survival rate in such units of between 10 and 24%. This may be improved even further by appropriate use of adjuvant treatment. The advanced stage of the disease when

INTRODUCTION

A recent study1 identi®ed a number of major problems underlying the quality of reporting of published results on the treatment of pancreatic cancer. Many studies accrued patients opportunistically rather than in accordance with a scienti®c entry criteria and the Correspondence to: Dr A. Haycox, Prescribing Research Group, The Inirmary, 70 Pembroke Place, Liverpool L69 3GF, UK. E-mail: [email protected] Ó 1998 Blackwell Science Ltd

normally encountered emphasizes the potential value of systemic treatment in this therapeutic area. Unfortunately systemic treatment (chemotherapy) has been found to be ineffective to date in signi®cantly extending survival, with a low rate and duration of remission being identi®ed in most trials. The challenge for both the health service and the pharmaceutical industry is to harness recent and future developments in scienti®c knowledge to the practical bene®t of clinicians. Where cure is possible it should be vigorously pursued; where it is not, in this ®eld above all others, clinicians have a duty of care. To achieve this it is necessary to abandon the therapeutic nihilism that has characterized the attitudes of clinicians towards this disease in the past. It is time that such nihilism was replaced by a recognition of the challenges and the opportunities available to clinicians in enhancing the quantity and quality of life available to patients. The dictum of `curing whenever possible but caring always' should be the future therapeutic philosophy used to guide clinicians in this important and rapidly changing therapeutic area.

majority of studies did not provide crucial data on patient and tumour characteristics (age, sex, disease stage, lymph node metastasis, tumour size, tumour site, histological grade) or undertake a histopathological veri®cation of pancreatic cancer. Studies frequently ignored long-term survival and made little or no effort to evaluate the level of quality of life experienced by patients. Perhaps of greatest concern was the fact that in one out of six of the studies examined patients were lost to follow-up without explanation. Such limitations in published studies cause signi®cant problems to both 949

950

A. HAYCOX et al.

researchers and clinicians given that progress depends upon careful evaluation of comparable evidence concerning the value of different therapeutic strategies. Only by standardizing the reporting of results is it possible to effectively evaluate the comparative value of the range of therapeutic strategies currently used or proposed for the treatment of pancreatic cancer. Despite these weaknesses, however, it is important that the extensive and rapidly growing body of research evidence generated in this area is effectively synthesized and reviewed to assess its practical value in informing current and future clinical decision-making with regard to treatment strategies for pancreatic cancer. Providing such a practically focused review is the purpose of this article. THE DUAL AIMS OF TREATMENT

The dual aims of treatment for pancreatic cancer are to maximize quality of life by ensuring effective symptom palliation for jaundice, pruritus, pain, diarrhoea and weight loss, and other complications, while simultaneously controlling or curing the underlying cancer. Surgery currently provides the best potential for cure in pancreatic cancer. However, this potential must be placed in the joint context of a relatively late presentation and the generally poor ability of the patient to withstand radical surgery due to general age and in®rmity. Effective palliation requires the sensitive management of the patient's physical symptoms in an integrated management strategy to improve survival and quality of life.2 Symptom palliation is often commenced during the course of making a diagnosis and consideration of the role of surgery or other treatment follows once the diagnosis is con®rmed. Given a median survival in pancreatic cancer of only 5±7 months,3 the speed and the manner in which palliation is achieved become crucial. Unfortunately, palliation involving courses of chemotherapy4, 5 or radiotherapy may take 6 weeks to administer. In such circumstances the time commitment and side-effects related to such courses would be likely to dominate remaining survival.6 The need to `trade off' improvements in `quantity' of life against the impact upon the `quality' of the patients' remaining life represents a key issue in identifying optimal palliative strategies for each patient. The range of strategies available to control the major symptoms of pancreatic cancer are outlined below.

ACHIEVING EFFECTIVE SYMPTOM PALLIATION

Weight loss Weight loss is a frequent symptom encountered in the early stages of pancreatic cancer and is often due to systemic cachexia caused by tumour growth. However, it is important not to overlook the possibility of malabsorption caused by pancreatic enzyme de®ciency. Pancreatic enzyme supplements may be of value in a minority of patients but are frequently omitted from the care package provided to the patient. Similarly these can be valuable in controlling diarrhoea due to pancreatic insuf®ciency but again are often forgotten. Pain Pain is another frequent and debilitating symptom with 70±80% of patients presenting with severe back or abdominal pain.7 Adherence to the WHO guidelines for cancer pain relief (oral administration of analgesic commencing with non-narcotics followed by weak and then strong narcotics in association with adjuvant drugs) provides adequate pain relief in approximately one-half of patients. Pain can also be effectively treated with chemical splanchnicectomy8 or through neurolytic coeliac blockade9 which provides effective relief for 3± 4 months.10, 11 A new approach to the management of intractable pain in pancreatic cancer is bilateral thoracic sympathectomy by excision or ablation of the splanchnic nerve. This innovative approach has been found to be an effective treatment for chronic pancreatic pain12 but requires further evaluation. Obstructive jaundice Obstructive jaundice occurs in 60±70% of patients.13 The role of surgical bypass has been challenged by the development of endoscopic or percutaneous transhepatic stenting which can relieve obstructive jaundice and pruritus in 90% of patients. The high rates of postoperative mortality together with limited post-operative survival combine to make the advantages of surgical bypass highly questionable.14 Equally however, the lower procedure-related mortality exhibited by stenting (»1%15) must be balanced against the need for frequent stent replacement in patients with lengthy post-operative survival. Newer metal stents which expand following deployment to a ®nal diameter of almost 10 mm are much less prone to occlusion from debris, which was a Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

REVIEW: TREATMENT OF PANCREATIC C ANCER

disadvantage of 10 and 12 French gauge plastic endoprostheses. However, tumour in-growth causing occlusion can still be a problem and these new stents are also much more expensive. From a cost-effectiveness perspective, while the initial costs associated with surgical intervention are higher, the bene®ts will probably last until the patient dies. As a consequence, surgical intervention is likely to be costeffective when targeted upon the minority of patients that are likely to experience more prolonged survival. However, a number of randomized studies16±19 have shown that stenting is superior for patients who are either a poor operative risk or who have a limited life expectancy. Bypass surgery should be restricted to patients who have longer anticipated survival, given that in such patients any advantages of stenting may be outweighed by the need for stent replacement due to occlusion, tumour in-growth or because of gastric outlet obstruction. The comparative impact of surgery and stenting must therefore be evaluated not only in terms of patient survival but also in terms of the post-operative quality of life experienced by patients. To date the only quality of life comparisons undertaken have been restricted to complication rates, hospital utilization or physical performance status.20±25 Gastric outlet obstruction Gastric outlet obstruction can develop in patients with pancreatic carcinoma provided with biliary bypass alone. The development of gastric outlet obstruction implies that the patient will subsequently require gastroenterostomy. It could be argued therefore that both biliary bypass and gastroenterology should be routinely performed together as a means of palliation. However, as the addition of a gastroenterostomy to surgical biliary bypass doubles the mortality rate but is ultimately needed by only about 10% of patients, some have concluded that patients should be palliated by biliary bypass alone.26, 27 THE ROLE OF SURGERY

The pancreas is dif®cult to access surgically because of its deep-seated location and close proximity to vital vascular structures. High post-operative mortality in the past has contributed to the notion that this was `the cancer which gave cancer a bad name'.28±30 Indeed, a long-term analysis of the outcome of partial pancreatÓ 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

951

icoduodenectomy (Whipple's resection) concluded that the impact on survival was so small that this procedure should be abandoned.31 Such nihilism, however, is seriously outdated as recent improvements in surgical techniques and technology32 have signi®cantly enhanced safety and ef®cacy so that 5-year survival of between 19%33 and 24%34 can be attained with postoperative mortality being reduced from 18% to less than 5%.35, 36 The situation has improved because of better patient selection and peri-operative and post-operative management together with the increasing concentration of pancreatic surgery in the hands of specialists who can achieve minimal operative mortality.37 In the UK, this trend is exempli®ed by a reduction of 30-day post-operative mortality from 45% in the ®rst decade and 28% in the second decade of a longitudinal analysis of 13 560 patients38 to 5.9% in a recent UKPACA study.39 An improvement in the surgical management of pancreatic cancer is also demonstrated by a Norwegian study40 which identi®ed an overall operative mortality of 9%. Only » 10% of patients have disease con®ned to the pancreas upon initial assessment41 and even when the disease is detected at this stage, the general in®rmity of the patient population may contraindicate radical surgery.42±44 Age by itself, however, should not represent a contraindication to surgical intervention. A recent review found advanced age insigni®cant in predicting surgical outcome,45 which contradicted the ®ndings of earlier studies. Size of tumour at presentation, histological grade and clear resection margins at surgery are the key features which signi®cantly determine outcome.33, 46±48 A study which recruited patients from the whole of Japan found that patients with small carcinomas exhibited a resectability rate of 99%, an operative mortality of only 4% and a 5-year survival of 30%.49 Furthermore, the early onset of obstructive jaundice in ampullary and lower bile duct carcinomas is likely to lead to more rapid diagnosis and a much higher 5-year survival following resection of 25±30%.50 Conversely, symptoms of tumours located in the head, corpus and tail of the pancreas are more commonly identi®ed at a much later stage, by which time often the tumour has in®ltrated adjacent structures. Alternative operations have been proposed with the aim of either improving long-term survival51 (total pancreatectomy, regional pancreatectomy, extended or subtotal resection) or improving quality of life52 (pylorus-preserving surgery). In general there is a paucity of

952

A. HAYCOX et al.

published data on the quality of palliation provided by the more extensive operations but they appear to provide little bene®t in terms of enhanced survival while signi®cantly increasing intra-operative and postoperative complications.40, 53 The less radical operations proposed54, 55 have been found to provide signi®cant bene®ts for only a small subset of the patient population. In general therefore the role of these newer procedures remains controversial, and further evidence is required concerning their impact upon survival and quality of life in broader subgroups of patients. For locally advanced disease, no generally accepted guidelines are available concerning the boundaries for resectability and the approach taken will be dependent

on the attitude of the surgeon.46, 56±61 The only clearcut contraindication to surgical intervention is when distant metastases are demonstratively present. However, even in these circumstances bene®cial results have been obtained in the surgical treatment of solitary liver metastasis.62 One study however, found that physical performance following pancreatectomy was signi®cantly better than following palliative bypass surgery,63 potentially justifying its use as a planned palliative intervention. More recently, laparoscopic bypass has been utilized in an attempt to achieve good surgical palliation whilst avoiding the disadvantages of major debilitating abdominal surgery in these patients.64

Table 1. Results of surgery Reference

Patient group analysed

Outcome of surgery

Gudjonsson31

Patients provided with Whipple resection

5-year survival (3.8%) abandon curative resection

Patients provided with palliative bypass

1970sÐmortality 17% survival 5 months 1980sÐmortality 14% survival 7 months

Cameron45 Cameron45 Pellegrini34 Pellegrini34 Livingston55 Satake68 Satake68 Bakkevold40 Bakkevold40 Edge65

Patients Patients Patients Patients Patients Patients Patients Patients Patients Patients

5-year survival of 19% 5-year survival of 57% 5-year survival of 24% 5-year survival of 57% 5-year survival of 9% 80% surgical cure rate 17% surgical cure rate Median survival 17 months Median survival 18 months Post-operative mortality 6% Major complications 21%

Wade48

Patients in Stage I±II provided with resection Patients in Stage III provided with resection

Additional 9-month survival No survival advantage

Bakkevold40

Patients with carcinoma of the pancreas and papillar of Vater

Operative mortality 9%

Bramhall38

Patients treated in the West Midlands of the UK over 20-year period

Post operative mortality First decade 45% Second decade 28%

Tsuchiya49

Patients with carcinoma of less than 2 cm

Resectability rate 99% Operative mortality 4% 5-year survival 30%

Watanapa32

Patients provided with operations for surgical palliation (1971±90)

First decade: Mortality 17%, survival 5 months Second decade: Mortality 14%, survival 7 months

provided with Whipple resection without metastasis provided with Whipple resection without metastasis treated surgically in the USA with carcinomas of less than 2 cm in stage IV of their disease provided with Whipple resection provided with total pancreatectomy provided with surgical resection

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

REVIEW: TREATMENT OF PANCREATIC C ANCER

Multi-institution analyses of outcomes for pancreatic surgery are of value in identifying the extent to which improvements in outcome achieved in single specialist centres can be generalized throughout the hospital sector. Such studies inevitably combine a wide range of surgical practice and expertise and must therefore be interpreted with caution. One such analysis of American University Centres65 identi®ed a post-operative mortality rate of 6% and a major complication rate of 21%, which coincided with results achieved in another recent series.66 In this study no association could be identi®ed between post-operative mortality and either the surgeons' caseload or patients' age,67 and while resection was found to provide signi®cant bene®ts for localized pancreatic cancer, such bene®ts were not apparent when the cancer was more extensive.48 In evaluating surgical outcomes it is imperative that outcome is assessed in the context of the patient population being analysed. In general, despite recent improvements, the prospect of bene®cial surgical intervention in the treatment of pancreatic carcinoma for the majority of patients remains poor, and therefore it is important to evaluate the effects of more systematic means of treating this aggressive carcinoma (Table 1). THE ROLE OF CHEMOTHERAPY

Fluorouracil (5-FU) remains the most effective and most frequently used single chemotherapy agent in the treatment of pancreatic carcinoma. 5-FU works partly by interference with enzymes such as thymidylate synthase, and partly by incorporation of metabolites into RNA and DNA interfering with cell proliferation. Continuous infusion of 5-FU or 5-FUdR (5-¯uorouracil deoxyribose) is more effective than administration by bolus and less toxic.69±71 The use of circadian infusion has also shown further improvement in the therapeutic/ toxic ratio which may facilitate further dose intensi®cation.72±74 Most studies analysing 5-FU and the simultaneous use of folinic acid75±79 have identi®ed a marginal survival bene®t over 5-FU alone but on no occasion was the resulting improvement found to be signi®cant. The addition of recombinant human granulocyte-macrophage colony-stimulating factor (rhGMCSF) to this combination80, 81 however, led to severe toxicity and no improvement in survival or quality of life.82 Conversely the combination of 5-FU with cisplatin is highly active in advanced pancreatic cancer with an acceptable level of toxicity in the majority of cases.83 Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

953

Only one randomized controlled trial has shown signi®cant survival bene®t using 5-FU in combination with doxorubicin and mitomycin C.84 Gemcitabine is a ¯uorinated pyramidine nucleoside analogue that inhibits DNA synthesis through a number of mechanisms but essentially is incorporated into DNA in place of dCTP and interferes with cell division. A Phase II trial of gemcitabine identi®ed signi®cant improvements in cancer-related symptoms and performance status for a number of patients with pancreatic carcinoma.85 A prospective, multicentre Phase II trial was initiated in a cohort of patients who had failed to respond to 5-FU.86 In this trial an outcome dimension was developed entitled `clinical bene®t response' to evaluate the palliative impact of gemcitabine. A clinical bene®t response was de®ned as having occurred if patients experienced a 50% reduction in pain, a 50% reduction in analgesic requirement or a 20 point improvement in Karnofsky performance score for over 4 weeks without any adverse change in any other measured characteristic. Seventeen out of 63 patients entered into the trial (27%) displayed a clinical bene®t response, which appears to indicate that gemcitabine can play a signi®cant role as a palliative agent in the treatment of pancreatic carcinoma. A further randomized trial that compared gemcitabine and 5-FU as ®rst line therapy in advanced pancreatic carcinoma87 found that average survival was 5.65 months for gemcitabine in comparison to 4.41 months for 5-FU. In addition, 24% of gemcitabine patients were alive at 9 months in comparison to only 6% of 5-FU patients. The proportion of patients displaying an improvement in `clinical bene®t response' was 24% for gemcitabine in comparison to only 5% in 5-FU. Unfortunately, gemcitabine was also associated with signi®cantly higher toxicity. A wide range of other single agents have been evaluated to assess their effectiveness in the treatment of pancreatic cancer. However, all such trials to date have met with very limited success. The majority of single agents evaluated have been found to be either ineffective or to lead to signi®cant levels of toxicity which makes their routine use of limited value.88±107 A selection of the results obtained in these trials is provided in Table 2. The lack of activity displayed by single agents is re¯ected in the results obtained for combination chemotherapy. None of the principal combinations evaluated to date have been found to be consistently superior to 5-FU alone while all exhibit

954

A. HAYCOX et al.

Table 2. Outcome of chemotherapy in pancreatic cancer Reference

Drug regimen

Outcome

Rougier83

5-FU and cisplatin

Overall response rate 26% Mean duration of response 10 months Palliative effect in 45% of patients Concluded to be active and well tolerated

Tajiri74

5-FU and leucovorin

Response rate 7% Median duration of response 3 months Concluded no therapeutic bene®t over 5-FU alone

Cascinu79

5-FU and leucovorin

Response rate 8% Median survival 5 months Severe toxicity led to trial being stopped

Weinerman77

5-FU and leucovorin

Mean survival of treated group 16 weeks Mean survival in matched controls 12 weeks

Moore87

Gemcitabine vs. 5-FU

Gemcitabine survival 6 months, clinical bene®t 24% 5-FU mean survival 4 months, clinical bene®t 5%

Hubbard90 Jones91 Moore82 Mahjoubi92 Leichmann94 Ebert99

Iproplatin Merbarone 5FU ± rhGM-CSF Pirarubicin Amona®de Octreotide

Concluded to be ineffective Concluded to be ineffective No survival or quality of life advantage over 5-FU alone Concluded to be ineffective Severe toxicity and ineffective Low dose: median survival 3 months (ineffective) High dose: median survival 6 months. No signi®cant side-effects and improved quality of life

Friess103 Casper104 Jenkins106 Kajanti107

Octreotide Edatrexate Piroxantrone Oral carmofur

Concluded to be ineffective at low dose Minor therapeutic activityÐineffective as a single agent Signi®cant toxicity and ineffective Median survival 8 months (3% partial response, 23% stable disease). Moderate toxicity

Decaprio108

5-FU & folinic acid

Median time to progression 3 months (7% partial response, 47% stable disease). No improvement over 5-FU alone

signi®cantly greater levels of toxicity.108±114 Again the results obtained for the principal combinations evaluated to date are provided in Table 2. Research evidence linking pancreatic cancer to sex hormones has led to signi®cant interest in the potential role of anti-oestrogen and anti-androgen treatments.115 The potential value of hormone therapy in the regulation of pancreatic cancer has been investigated98 with the hormone CCK being of particular interest.116 The CCK receptor antagonist MK-329 is highly selective for the predominant receptor present in pancreatic tissue117±119 and signi®cantly enhances the effect of cisplatin in animal models but at the cost of increased toxicity.120 Application of this approach to humans requires greater understanding of the role of CCK receptor blockade and further control of levels of toxicity. In general, analyses undertaken to date

emphasize that while certain patients may bene®t from such treatments,121 there is little evidence of a generalized improvement in survival.122 The generally disappointing results obtained from chemotherapy arise in part because it is dif®cult to increase the intratumour concentration of an anticancer drug to a therapeutic level by systematic administration alone.123, 124 Early experience with the intratumoural administration of anti-cancer agents has found them to be more effective and less toxic than systemic administration.125 Early work in animal models has shown that intratumoural administration was successful in completely suppressing tumour growth at a dose that would be ineffective via the intravenous route.126 Whilst such early results are encouraging they require much greater validation prior to their generalized use in pancreatic carcinoma. Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

REVIEW: TREATMENT OF PANCREATIC C ANCER

THE ROLE OF RADIOTHERAPY OR CHEMORADIOTHERAPY

The survival bene®ts provided by radiotherapy alone have been generally disappointing.127 The location of the pancreas (surrounded by crucial radiosensitive structures) limits access and the intensity of radiotherapy that can be applied. Radiotherapy is therefore of greatest value in patients who present with disease localized to the pancreatic bed.128 For such patients a number of small studies have identi®ed survival and palliation ®gures for radiotherapy that are comparable to those obtained through surgery, although no direct comparison of these modalities has been undertaken to date.129±132 Adjuvant use of external beam radiotherapy after surgical resection has been reported to signi®cantly improve patient survival133 while the combination of 5-FU with external beam radiotherapy has also been found to extend survival in comparison to either 5-FU or radiotherapy alone.134, 135 Unfortunately the majority of comparative analyses of patient outcome have been generated in comparatively small studies using a non-randomized research design. It is imperative that high-quality randomized controlled trials of the comparative outcome of radiotherapy are undertaken to effectively determine its role and value in the treatment of pancreatic carcinoma. The combination of radiotherapy and chemotherapy aims to combine the enhanced local control obtained through radiotherapy with the wider systemic bene®ts of chemotherapy.136±139 Unfortunately while 5-FU signi®cantly increases the radiosensitivity of pancreatic tumours the tolerable dose of 5-FU in combined therapy is reduced below optimum levels and such suboptimal dosing may adversely affect patient outcome.140 The combination of 5-FU and split course radiotherapy has led to modest improvements in palliation and survival and has become ®rst line therapy in many hospitals. A range of variations of combined modality therapy have been evaluated including radiotherapy and simultaneous multi-drug chemotherapy consisting of 5-FU, streptozotocin and cisplatin (RT-FSP).141 The RT-FSP trial con®rms the ability of clinicians to achieve bene®cial therapeutic goals for patients with locally advanced pancreatic cancer using chemotherapy (Table 3). Further trials are essential to evaluate the potential contribution of new drugs to the radiotherapy/5-FU combination.

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

955

A range of other strategies have been proposed to enhance the effectiveness of radiotherapy in improving survival or palliation in patients with pancreatic cancer. Such strategies include wide area radiation,149 radioactive implants,150 intra-operative radiation therapy,143 conformal radiation techniques151 and hyperfractionated radiation therapy.152 Wide Area Radiation Therapy (WART) was developed to more effectively meet the treatment needs of the 95% of patients with pancreatic cancer that develop hepatic metastases. A number of non-randomized trials142, 143 have identi®ed signi®cantly increased survival in certain patient groups through the use of chemoradiation to the primary tumour supported by wide ®eld irradiation. The potential bene®ts of this approach, however, while promising, require con®rmation in large-scale randomized trials. Direct implantation of radioactive sources enables the delivery of intensive but localized doses of radiation which, when combined with external beam radiation therapy and chemotherapy, has been found to produce signi®cant improvements in local disease control. One study144 found that implantation led to enhanced survival (Table 3), and signi®cantly improved local control of the disease and effective palliation of symptoms. Unfortunately, radioactive implantation is associated with complications such as deep vein thrombosis, gastrointestinal haemorrhage and sepsis and also exhibits an operative mortality as high as 33%.153 Thus although implantation radiotherapy may improve local control of pancreatic cancer, its associated morbidity and mortality make it a potentially dangerous procedure.154 Pre-operative radiotherapy aims to reduce the size and spread of tumour so as to enhance the possibility and ef®cacy of curative resection. Pre-surgical radiotherapy has the theoretical advantages of increased vulnerability of the cancer cells to radiotherapy given that vascularity has not been interrupted by surgery and patients are likely to be in better general health. Hoffman et al.155 concluded that the method was safe and effective in downstaging lymph nodes and resection margins but that the implications of such changes upon patient survival requires further evaluation in the form of a controlled clinical trial. This result mirrors other studies146, 147, 156, 157 that have supported the value of pre-operative irradiation in increasing the overall resectability rate and thus improving patient survival.

956

A. HAYCOX et al.

Table 3. Outcome of radiotherapy/chemoradiotherapy in pancreatic cancer Reference

Nature of treatment

Outcome

Komaki142 Gunderson143 Goertz144

Wide ®eld irradiation Local vs. wide ®eld irradiation Implantation and XRT compared to XRT and surgical resection

Survival (1 year, 67%; 2 year, 47%) 2-year survival 6% (local), 20% (wide ®eld) Median survival 8 months (Implant & XRT) 7 months (Surgery) 5 months (XRT alone)

Sindelar145

IORT compared to conventional therapy External beam irradiation following resection

Major complications (35%) No signi®cant difference in survival Local recurrence rate 50% Disease free survival 12 months Overall survival 23 months

Pre-operative radiotherapy on unresectable patients Preoperative chemoradiation on unresectable patients IORT with post-operative chemotherapy compared to XRT Surgery vs. split course radiotherapy and 5FU

35% of patients found at 6 week assessment to be resectable (2 survived 5 years and 1 for 7 years) 48% of patients provided with resection leading to a `signi®cant increase in survival' SurvivalÐno signi®cant difference Local relapse rate IORT (25%)Ðcontrol (55%) Median survival: Surgery 11 months CombinationÐ20 months 2-year survival: SurgeryÐ15% CombinationÐ42%

Moertle137

EBRT vs. combination therapy

Blitzer136

Radiotherapy vs. combined therapy

Median survival: Radiotherapy aloneÐ6 months Combination therapyÐ10 months One year survival: Radiation aloneÐ25% Combination therapyÐ44%

Gastrointestinal Tumour Study Group134

Adjuvent chemoradiation following resection

Median survival: No chemoradiationÐ11 months ChemoradiationÐ21 months Two year survival: No chemoradiationÐ18% ChemoradiationÐ43%

Bruckner141

Combination therapy (RT-FSP)

Remission: completeÐ33%, partialÐ33% Signi®cantly improved local control

Bossett127

Pilepich146 Coia147 Fossati148 Kalser135

Unfortunately none of the studies was undertaken in the form of a randomized control trial and therefore the results of such studies should be interpreted as being indicative rather than providing conclusive evidence. Intraoperative radiation therapy (IORT) facilitates the safe delivery of higher radiation doses to pancreatic tumours than can be given by external beam techniques.158±161 It has been suggested that this technique may lead to improved survival and a reduction in local recurrence rates151 without signi®cantly increasing surgical complications.162 A number of studies have also found signi®cant improvements in the palliation of severe pain.148, 160, 163 Local control rates may be further improved by the use of a boost technique such as intraoperative electron beam irradiation (IOEBRT); however, a prospective randomized study comparing IOEBRT with conventional radiotherapy145 found no

signi®cant difference in either local control or median survival. However, the combination of extended radical pancreatectomy with intraoperative radiation therapy seems to signi®cantly increase long-term survival.164 Unfortunately, IORT may also inhibit the activity of tumour in®ltrating lymphocytes. The restriction of this natural killer activity means that IORT is best provided with biological response modi®ers to counter such inhibition of local immunity165 if the theoretical advantages provided by IORT are to be fully translated into survival bene®t for patients. THE ROLE OF TUMOUR CELL BIOLOGY

The development of an enhanced knowledge of the causative agents and mechanics underlying genetic mutation has provided valuable clues to tumorigenesis Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

REVIEW: TREATMENT OF PANCREATIC C ANCER

and the process of tumour development. However, despite the fact that recent studies have begun to generate information concerning the genetic pro®le underlying pancreatic adenocarcinoma, little is currently known of the genetic alterations arising in the very earliest stages of such tumours. The identi®cation of `early' tumours that are amenable to resection still remain genetically at a late stage of tumorigenesis and hence can provide little indication of preceding changes. The development of a detailed understanding of the process and time sequence of molecular changes in preneoplastic lesions and early pancreatic tumorigenesis represents a crucial gap in our knowledge. Improved understanding of such molecular changes would provide the potential to develop entirely new therapeutic strategies to correct or assuage the damage caused by such molecular mutations.166 Interference with regulatory factors and signals is another potential strategy for future treatments. It is known, for example, that pancreatic cancer overexpresses epidermal growth factor receptor (EGFR)167 as well as producing transforming growth factor a (TGF-a). This combination enables cells to proliferate in an anchorage independent manner168 through an autocrine loop which provides cancer cells with a signi®cant growth advantage. Their normal function becomes usurped by the cancer cells, leading to activation of growth-promoting pathways. Growth control and dif-

957

ferentiation is in¯uenced by components of the extracellular matrix and within this matrix cytokines regulate the recruitment of effector cells in tumour tissue by changing cell adhesion and chemotaxis. Combined treatment with cytokine TNF-a and an EGFR-speci®c monoclonal antibody can modulate antigen expression and provoke an effective immunotherapeutic response.169 A range of further pharmacological interventions directed at changing the function of this growth regulatory gene are currently under development.170 Promising results obtained in mice171 have stimulated trials in patients suffering from unresectable pancreatic cancer. Such trials strongly suggest that cytokines can modulate antigen expression and provoke an effective immunotherapeutic response with a monoclonal antibody in pancreatic cancer patients.172 The role of angiogenesis in tumour sustenance173 also provides possibilities of adjuvant treatment by its inhibition by cytokines.174 Tumour invasion requires in®ltration of cells into adjacent tissues and degradation of extracellular barriers represents an important feature underlying the aggressive nature of pancreatic cancer.175 The process of overcoming such barriers involves matrix metalloproteinases (MMP) which are a family of 15 proteolytic enzymes which enable abnormal cells to spread by overcoming the barriers formed by the ECM. A range of biotechnology companies are currently engaged in

Table 4. Outcome of palliative treatment in pancreatic cancer Reference

Nature of palliative treatment

Outcome

Klassen139

Biliary by-pass

Post-operative mortality 19% Median survival 5 months

Huibregtse15

Endoscopic stents

Post-operative mortality 1% Successful palliation 85%

Sharp10 Ventafridda9

Operative celiac plexus block WHO guidelines Subsequent use of neurolytic coeliac block Operative celiac plexus block

Successful palliation: (1±2 months duration 83%) Successful palliation 49% Successful palliation 100% Successful palliation 85% Pain free at death 50%

Bypass alone compared with bypass plus gastroenterostomy

Successful palliation by biliary bypass alone 90% (little additional bene®t from gastroenterostomy)

Analysis of post-operative mortality for operations of surgical palliation

Whipples 5±10% Biliary by-pass 12% Gastrojejunostomy 14%

Gemcitabine

`Clinical bene®t response' experienced by 27% of patients

Costamagna11 Holbrook26 Watanapa

Casper85

32

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

958

A. HAYCOX et al.

developing MMP inhibitors with the aim of reducing the ability of cancer cells to invade adjacent cells. One of the ®rst synthetic compounds to enter clinical trials is marimastat (British Biotech) which is being tested in a variety of protocols in the treatment of advanced pancreatic cancer as well as in the adjuvant setting. This is a novel approach with interesting possibilities for extending survival176 in a therapeutic area characterized by the extreme aggressiveness of the underlying growth of the carcinoma. CONCLUSIONÐCONFRONTING THE CLINICAL CHALLENGES

The short-term challenge is to alter clinical attitudes away from therapeutic nihilism, in order to take full advantage of advances in drug therapy177 and other new therapeutic innovations.86 The extent of this shortterm challenge is emphasized by studies that question the reliability of even the minimal 5-year survival rates for pancreatic adenocarcinoma presented in the literature. One study178 found that the majority of such `survivors' had a histologically uncon®rmed tumour with diagnosis being based entirely upon macroscopical ®ndings, or radiological or clinical evidence alone. The authors concluded that the true 5-year survival rate for pancreatic (extra-ampullary) adenocarcinoma is close to zero. The medium-term challenge is to optimize the use of currently available therapeutic strategies. Most patients will continue to present, for the foreseeable future, with advanced disease, posing the dual therapeutic challenge of local and distant tumour control. Improving local control requires effective surgical, chemotherapeutic and radiotherapeutic interventions. Therapeutic strategies must also take adequate account of the impact of treatment strategies upon the quality of a patient's remaining life. Quality of life evaluation must therefore become integral to the evaluation of therapeutic strategies for this patient group. The long-term challenge is to effectively translate scienti®c advance into clinical practice. In this way innovative therapeutic strategies can be developed to interfere with the fundamental pathways that initiate and promote pancreatic carcinoma. Improved understanding of the role of mutated ras proteins and autocrine/paracrine growth factors offers examples of areas in which an improved understanding of methods of

modulating pancreatic carcinoma cell growth offers the potential for signi®cant improvements in patient care. In confronting these challenges it is important that clinicians make optimal use of the skills and expertise of other health professionals and scientists. This is best achieved through close collaboration and af®liation with an expert multidisciplinary team. Undoubtedly, further advances in unravelling the biology of pancreatic cancer and improving both palliative and curative treatment options will depend on channelling and concentrating patients through protocols in multidisciplinary centres of excellence. In this way a `bespoke' treatment can be tailored to the individual patient which provides the best method of improving the quality and quantity of life experienced by the current and future patient population in this important therapeutic area (Table 4). ACKNOWLEDGEMENTS

The authors gratefully acknowledge the provision of an education grant from Eli Lilly which supported and facilitated this research. REFERENCES 1 Andren-Sandberg A, Cedercrantz C. Review of standards for reporting results of treatment of exocrine pancreatic cancer. Int J Pancreatol 1993; 14(3): 213±17. 2 Krige JE, Bornman PC, Terblanche J. Optimal palliation of pancreatic carcinoma. South African Med J 1992; 81: 238± 40. 3 Connolly MM, Dawson PJ, Michelassi F, Moossa AR, Lowenstein F. Survival in 1001 patients with carcinoma of the pancreas. Am Surg 1987; 206(3): 366±73. 4 Bukowski RM, Fleming TR, MacDonald JS, Oishi N, Taylor SA, Baker LH. Evaluation of combination chemotherapy and Phase II agents in pancreatic adenocarcinoma. Cancer 1993; 71(2): 322±5. 5 Cullinan SA, Moertel CG, Fleming TR, et al. A comparison of three chemotherapeutic regimens in the treatment of advanced pancreatic and gastric carcinoma. J Am Med Assoc 1985; 253(14): 2061±7. 6 Pelley RJ. Role of chemotherapy in the palliation of gastrointestinal malignancies. Seminars Oncol 1995; 22(2(Suppl. 3): 45±52. 7 Krech R, Walsh D. Symptoms of pancreatic cancer. J Pan Symptom Manage 1991; 6: 360±7. 8 Lillemoe KD, Cameron JL, Kaufman HS, Yeo CJ, Pitt HA, Sauter PK. Chemical splanchnicectomy in patients with unresectable pancreatic cancer: a prospective randomised trial. Ann Surg 1993; 217(5): 447±57.

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

REVIEW: TREATMENT OF PANCREATIC C ANCER 9 Ventafridda GV, Caraceni AT, Sbanotto AM, Barletta L. Pain treatment in cancer of the pancreas. Eur J Surg Oncol 1990; 16: 1±6. 10 Sharp KW, Stevens EJ. Improving palliation in pancreatic cancer: intraoperative celiac plexus block for pain relief. Southern Med J 1991; 84(4): 469±71. 11 Costamagna G, Gabbrielli A, Mutignani M. Treatment of `obstructive' pain by endoscopic drainage in patients with pancreatic head carcinoma. Gastrointest Endosc 1993; 39: 774±7. 12 Worsey J, Ferson PF, Keenan RJ, Julian JB, Landreneau RJ. Thoracoscopic pancreatic denervation for pain control in irresectable pancreatic cancer. Br J Surg 1993; 80(8): 105± 12. 13 Sarr MG, Cameron JL. Surgical palliation of unresectable carcinoma of the pancreas. World J Surg 1984; 8: 906±18. 14 Sarr MG, Cameron JL. Surgical management of unresectable carcinoma of the pancreas. Surgery 1982; 91: 123±33. 15 Huibregtse K, Katon RM, Coene PP, Tytgat GNJ. Endoscopic palliative treatment in pancreatic cancer. Gastrointest Endosc 1986; 32(5): 334±8. 16 Bornman PC, Tobias R, Harries-Jones EP, Vanstiegmann G. Prospective controlled trial of transhepatic biliary endoprosthesis versus bypass surgery for incurable carcinoma for head of pancreas. Lancet 1986; 1(8472): 69±71. 17 Speer AG, Cotton PB, Russell RCG, et al. Randomised trial of endoscopic versus percutaneous stent insertion in malignant obstructive jaundice. Lancet 1987; 2(8550): 57±62. 18 Shepherd HA, Diba A, Ross AP, Arthur M, Royle G, ColinJones D. Endoscopic biliary endoprosthesis in the palliation of malignant obstruction of the distal common bile duct: a randomised trial. Br J Surg 1988; 75(10): 1166±8. 19 Anderson JR, Sorenson SM, Kruse A, Rokkjaer M, Matzen P. Randomised trial of endoscopic endoprosthesis versus operative bypass in malignant obstructive jaundice. Gut 1989; 30(8): 1132±5. 20 Shapiro G. Adenocarcinoma of the pancreas: a statistical analysis of biliary bypass versus Whipple resection in good risk patients. Ann Surg 1975; 182: 715±21. 21 Lygidakis NJ, Brummelkamp WH, Tytgat GH, et al. Periampulary and pancreatic head carcinoma: facts and factors in¯uencing mortality, survival and quality of post-operative life. Am J Gastroenterol 1986; 81(10): 968±74. 22 Crist DW, Sitzmann JV, Cameron JL. Improved hospital morbidity, mortality and survival after Whipple procedure. Ann Surg 1987; 206(3): 358±65. 23 Pliam MB, Remine WH. Further evaluation of total pancreatectomy. Arch Surg 1975; 110: 506±12. 24 Strachura J, Tomaszewska R, Pietron M, Kulig J. Does microhistology improve the cytological diagnosis of liver and pancreatic tumours? Cytopathology 1991; 2: 39±42. 25 Yasue M, Sakamoto J, Morimoto T, et al. Evaluation of the effects of pancreatic resection in advanced pancreatic cancer with special reference using hospital free survival as a measure of quality of life. Jap J Clin Oncol 1995; 25: 37± 45.

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

959

26 Holbrook AG, Chester JF, Britton DC. Surgical palliation for pancreatic cancer: will biliary bypass alone suf®ce? J Royal Soc Med 1990; 83: 12±14. 27 Weaver DW. Gastrojejunostomy: is it helpful for patients with pancreatic cancer? Surgery 1987; 102: 608±13. 28 Kinsella TJ. Adjuvant radiation therapy in pancreatic carcinoma: a reappraisal. Cancer Invest 1988; 6(6): 745±6. 29 Baylor SN, Berg JW. Cross-classi®cation and survival characteristics of 5000 cases of cancer of the pancreas. J Surg Oncol 1973; 5: 335±58. 30 Cubilla AL, Fitzgerald PJ, Fortner JG. Pancreas cancer±duct cell adenocarcinoma: survival in relation to site, size, stage and type of therapy. J Surg Oncol 1978; 10: 465±82. 31 Gudjonsson B. Cancer of the pancreas: 50 years of surgery. Cancer 1987; 9: 2284±303. 32 Watanapa P, Williamson RCN. Surgical palliation for pancreatic carcinoma: developments during the past two decades. Br J Surg 1992; 79: 8±20 33 Cameron JL, Crist DW, Sitzmann JV, et al. Factors in¯uencing survival after pancreaticoduodenectomy for pancreatic cancer. Am J Surg 1991; 161(1): 120±5. 34 Pellegrini CA, Heck CF, Raper S, Way LW. An analysis of the reduced morbidity and mortality rates after pancreaticoduodenectomy. Arch Surg 1989; 124(7): 778±81. 35 Lieberman MD, Kilburn H, Lindsey M, Brennan MF. Relation of perioperative deaths to hospital volume among patients undergoing pancreatic resection for malignancy. Ann Surg 1995; 222(5): 638±45. 36 Neiderhuber JE, Brennan MF, Menck HR. The national cancer database report on pancreatic cancer. Cancer 1995; 76: 1671±7. 37 Fernandez-del Castillo C, Rattner DW, Warshaw AL. Standards for pancreatic resection in the 1990s. Arch Surg 1995; 130: 295±300. 38 Bramhall SR, Allum WH, Jones AG, Allwood A, Cummins C, Neoptolemos JP. Treatment and survival in 13,560 patients with pancreatic cancer, and incidence of the disease in the West Midlands: an epidemiological study. Br J Surg 1995; 82(1): 111±15. 39 Neoptolemos JP, Russell RCG, Bramhall S, Theis B. Low mortality following resection for pancreatic and periampullary tumours in 1026 patients: UK survey of pancreatic specialist units. Br J Surg 1997; 84: 1370±6. 40 Bakkevold KE, Kambestad B. Long term survival following radical and palliative treatment of patients with carcinoma of the pancreas and papillar of vaterÐthe prognostic factors in¯uencing the long term results: a prospective multicentre study. Eur J Surg Oncol 1993; 19: 147±61. 41 Armstrong B, Doll R. Environmental factors and cancer incidence and mortality in different countries, with special reference to dietary practices. Int J Cancer 1975; 15: 617± 31. 42 Gordis L, Gold EB. Epidemiology of pancreatic cancer. World J Surg 1984; 8: 808±21. 43 Lillemoe KD. Pancreatic disease in the elderly patient. Surg Clinics North Am 1994; 74: 317±44.

960

A. HAYCOX et al.

44 Macmahon B. Risk factors for cancer of the pancreas. Cancer 1982; 50: 2676±82. 45 Cameron JL, Pitt HA, CJY, et al. 145 consecutive pancreatoduodenectomies without mortality. Ann Surg 1993; 217(5): 430. 46 Trede M, Schwall G, Saeger HD. Survival after pancreatoduodenectomy: 118 consecutive resections without an operative mortality. Ann Surg 1990; 211: 447±58. 47 Klempnauer J, Ridder GJ, Bektas H, Pichlmayer R. Surgery for exocrine pancreatic cancer: who are the 5 and 10 year survivors? Oncology 1995; 52(5): 353±9. 48 Wade TP, Kraybill WG, Virgo KS, Johnson FE. Pancreatic cancer treatment in the US veteran from 1987 to 1991: effects of tumour stage on survival. J Surg Oncol 1995; 58: 104±111. 49 Tsuchiya R, Noda T, Harada N, et al. Collective review of small carcinomas of the pancreas. Ann Surg 1986; 203(1): 77±81. 50 Michelassi F, Erroi F, Dawson PJ, et al. Experience with 647 consecutive tumours of the duodenum, ampulla, head of the pancreas and distal common bile duct. Am Surg 1989; 20(4): 544±56. 51 van Heerden JA. Pancreatic resection for carcinoma of the pancreas: Whipple versus total pancreatectomyÐan institutional perspective. World J Surg 1984; 8: 880±7. 52 Braasch JW, Deziel DJ, Rossi RL, Watkins E, Winter PF. Pyloric and gastric preserving pancreatic resection. Ann Surg 1986; 204(4): 411±16. 53 van Heerden JA, McIlrath DC, Ilstrup DM, Weiland LH. Total pancreatectomy for ductal adenocarcinoma of the pancreas: an update. World J Surg 1988; 12(5): 658±62. 54 Grace PA, Pitt HA, Longmire WP. Pylorus preserving pancreaticoduodenectomy: an overview. Br J Surg 1990; 77(9): 968±74. 55 Livingston EH, Welton ML, Reber HA. Surgical treatment of pancreatic cancer. Int J Pancreatol 1991; 9: 153±7. 56 Jones BA, Langer B, Taylor BR, Girotti M. Periampullary tumours: which ones should be resected? Am J Surg 1985; 149(1): 46±52. 57 Monabe T, et al. Evaluation of en block radical pacreatectomy for carcinoma of the head of the pancreas involving the adjacent vessels. Dig Surg 1985; 2: 27±30. 58 Trede M. The surgical treatment of pancreatic carcinoma. Surgery 1985; 97: 28±35. 59 Matsuno S, Sato T. Surgical treatment for carcinoma of the pancreasÐexperience in 272 patients. Am J Surg 1986; 152: 499±503. 60 Funovics JM, Karner J, Pratschner T, Fritsch A. Current trends in the management of carcinoma of the pancreatic head. Hepatogastroenterology 1989; 31(6): 450±5. 61 Jeekel J. Surgery of pancreatic cancer. Ann Oncol 1994; 5(Suppl. 3): 73±4. 62 Takamori H, Hiraoka T, Kanemitsu K, et al. Treatment strategies for hepatic metastases from pancreatic cancer in patients previously treated with radical resection combined with intraoperative radiation therapy. HPB Surg 1994; 8: 107±10.

63 Miyata M, Nakao K, Takao T, et al. An appraisal of pancreatectomy for advanced cancer of the pancreas based on survival rate and postoperative physical performance. J Surg Oncol 1990; 45: 33±9. 64 Targaroma E, Pera M, Martinez J, Balague C, Trias M. Laparoscopic treatment of pancreatic disorders: diagnosis and staging, palliation of cancer and treatment of pancreatic pseudocysts. Int Surg 1996; 81(1): 1±5. 65 Edge SB, Schmieg RE, Rosenlof LK, Wilhelm MC. Pancreas cancer resection outcome in American university centres in 1989±90. Cancer 1993; 71(11): 3502±8. 66 Grace PA. Decreased morbidity and mortality after pancreatoduodenectomy. Am J Surg 1986; 151: 141±9. 67 Forrest JF, Longmire WP. Carcinoma of the pancreas and periampullary region: a study of 279 patients. Ann Surg 1979; 189: 129±38. 68 Satake K, Nishiwaki H, Yokomatsu H, et al. Surgical curability and prognosis for standard versus extended resection for T1 carcinoma of the pancreas. Surg Gynecol Obstet 1992; 175: 259±65. 69 Bruckner HW, Crown J, McKenna A, Hart R. Leucovorin and 5 ¯uorouracil as a treatment for disseminated cancer of the pancreas and unknown primary tumours. Cancer Res 1988; 88(19): 5570±2. 70 Hansen R, Quebbeman R, Ausman R, et al. Continuous systemic 5-¯uorouracil infusion in advanced colorectal cancer: results in 91 patients. J Surg Oncol 1989; 40(3): 177±81. 71 Lokich JJ, Ahlgren JD, Gullo JJ, Philips JA, Fryer JG. A prospective randomised comparison of continuous infusion of ¯uorouracil with a conventional bolus schedule in metastatic colorectal carcinoma: a Mid Atlantic Oncology Program study. J Clin Oncol 1989; 7(4): 425±32. 72 von Roemeling R, Hrushesky WJM. Circadian patterning of continuous ¯oxuridine infusion reduces toxicity and allows higher dose intensity in patients with widespread cancer. J Clin Oncol 1989; 7(1): 1710±19. 73 Hrushesky WJM, Vonroemeling R, Lanning RM, Rabati JT. Circadian shaped infusion of ¯oxuridine for progressive metastatic renal cell carcinoma. J Clin Oncol 1990; 8(9): 1504±13. 74 Tajiri H, Yoshimori M, Okazaki N, Miyati M. Phase II study of continuous venous infusion of 5-¯uorouracil in advanced pancreatic cancer. Oncology 1991; 48: 18±21. 75 Bleyer WA. New vistas for leucovorin in cancer chemotherapy. Cancer 1989; 63: 995±1007. 76 O'Connell MJ. A Phase III trial of 5-¯uorouracil and leucovorin in the treatment of advanced colorectal cancer. Cancer 1989; 63: 1026±30. 77 Weinerman BH, MacCormick RE. A Phase II survival comparison of patients with adenocarcinoma of the pancreas treated with 5-¯uorouracil and calcium leucovorin versus a matched tumour registry control population. Am J Clin Oncol 1994; 17(6): 467±9. 78 Decaprio JA, Mayer RJ, Gonin R, Arbuck SG. Fluorouracil and high dose leucovorin in previously untreated patients with advanced adenocarcinoma of the

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

REVIEW: TREATMENT OF PANCREATIC C ANCER

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

pancreas: results of a Phase II trial. J Clin Oncol 1991; 9(12): 2128±33. Cascinu S, Fedeli A, Fedeli SL, Catalano G. 5-¯uorouracil, leucovorin and interferon alpha 2B in advanced pancreatic cancer: a pilot study. Ann Oncol 1993; 4(1): 83±4. Gabrilove JL, Jakubowski A, Scher H, et al. Effect of granulocyte colony-stimulating factor on neutropenia and associated morbidity due to chemotherapy for transitional cell carcinoma of the urothelium. New Engl J Med 1988; 318(22): 1414±22. Sheridan WP, Wolf M, Lusk J, et al. Granulocyte colonystimulating factor and neutrophil recovery after high dose chemotherapy and autologous bone marrow transplantation. Lancet 1989; 2(8668): 891±4. Moore DF, Pazdur R. Phase I study of 5-¯uorouracil with folinic acid combined with recombinant human granulocytemacrophage colony-stimulating factor. Am J Clin Oncol 1992; 15(6): 464±6. Rougier P, Zarba JJ, Ducreaux M, et al. Phase II study of cisplatin and 120-hour continuous infusion of 5-¯uorouracil in patients with advanced pancreatic adenocarcinoma. Ann Oncol 1993; 4: 333±6. Palmer KR, Kerr M, Knowles G, Cull A, Carter DC, Leonard RCF. Chemotherapy prolongs survival in operable pancreatic carcinoma. Br J Surg 1994; 81: 882±5. Casper ES, Green MR, Kelsen DP, et al. Phase II trial of gemcitabine in patients with adenocarcinoma of the pancreas. Invest New Drugs 1994; 12(1): 29±34. Rothenberg MC, et al. Gemcitabine: effective palliative therapy for pancreas patients failing 5-FU. Proc Am Soc Clin Oncol 1995; 14: 470. Moore M, et al. A randomised trial of gemcitabine versus 5FU as ®rst line therapy in advanced pancreatic cancer. Proc Am Soc Clin Oncol 1995; 14: 473. Bruera E, Macmillan K, Kuehn N, Hanson J, MacDonald RN. A controlled trial of megastrol acetate in appetite, caloric intake, nutritional status and other symptoms in patients with advanced cancer. Cancer 1990; 66(6): 1279±82. Marsh R, Manyam V, Bewsher C, Youngblood M. Circadian rhythm modulated 5-FUdR infusion with megace in the treatment of advanced pancreatic cancer. J Surg Oncol 1994; 57: 25±9. Hubbard KP, Pazdur R, Ajani JA, et al. Phase II evaluation of iproplatin in patients with advanced gastric and pancreatic cancer. Am J Clin Oncol 1992; 15(6): 524±7. Jones DV, Ajani JA, Winn RJ, Daugherty KR, Levin B, Krakoff IH. A Phase II study of merbarone in patients with adenocarcinoma of the pancreas. Cancer Invest 1993; 11(6): 667±9. Mahjoubi M, Rougier P, Oliviera J, Herait P, Tigaud JM, Droz JP. Phase II trial of pirarubicin in the treatment of advanced pancreatic cancer. Cancer Invest 1994; 12(4): 403±5. Linke K, Pazdur R, Abbruzzese JL, et al. Phase II study of amona®de in advanced pancreatic adenocarcinoma. Invest New Drugs 1991; 9: 353±6. Leichman CG, Tangen C, Macdonald JS, Leimert T, Fleming TR. Phase II trial of amona®de in advanced pancreas cancer:

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

95

96

97 98 99

100 101

102 103

104

105

106

107

108

109

110

111

961

a Southwest Oncology Group Study. Invest New Drugs 1993; 11(2±3): 219±21. Buchler M, Friess H, Klempa I, et al. Role of octreotide in the prevention of postoperative complications following pancreatic resection. Am J Surg 1992; 163(1): 125±31. Upp JR, Olson D, Poston GJ, Alexander RW, Townsend CM, Thompson JC. Inhibition of growth of two human pancreatic carcinomas in vivo by somatostatin analogue SMS 201-995. Am J Surg 1988; 155(1): 29±35. Liebow C, Hierowski M, duSpain K. Hormonal control of pancreatic cancer growth. Pancreas 1986; 1: 44±8. Poston GJ, Gillespie J, Guillou PJ. Biology of pancreatic cancer. Gut 1991; 32(7): 800±12. Ebert M, Friess H, Beger HG, Buchler MW. Role of octreotide in the treatment of pancreatic cancer. Digestion 1994; 55(S1): 48±51. Stoscheck CM, King LE. Role of epidermal growth factor in carcinogenesis. Cancer Res 1986; 46: 1030±7. Mascardom RN, Sherline P. Somatostatin inhibits rapid centrosomal separation and cell proliferation induced by epidermal growth factor. Endocrinology 1982; 111: 1394±6. Poston GJ. Somatostatin and the treatment of pancreatic cancer. Ann R Col Surg Eng 1994; 76: 172±4. Friess H, Buchler M, Beglinger C, et al. Low dose octreotide treatment is not effective in patients with advanced pancreatic cancer. Pancreas 1993; 8(5): 540±5. Casper ES, Schwartz GK, Johnson B, Kelson DP. Phase II trial of edatrexate in patients with advanced pancreatic adenocarcinoma. Invest New Drugs 1992; 10(3): 313±16. Schmid FA, Sirotnak FM, Otter GM, Degraw JI. Combination chemotherapy with a new folate analog: activity of 10-ethyl10-deaza-aminopterin compared to methotrexate with 5-FU and alkylating agents against advanced metastatic disease in murine tumor models. Cancer Treat Rep 1987; 71: 727±32. Jenkins TR, Tangen C, Macdonald JS, Weiss G, Chapman R, Hantel A. A Phase II trial of piroxantrone in adenocarcinoma of the pancreas: a Southwest Oncology Group Study. Invest New Drugs 1993; 11(4): 329±31. Kajanti MJ, Pyrhonen SO. Phase II trial of oral carmofur in advanced pancreatic carcinoma. Ann Oncol 1991; 2: 765±6. Decaprio JA, et al. Phase II of weekly 5 ¯uorouracil (5-FU) with folinic acid (FA) in previously untreated patients with unresectable measurable pancreatic adenocarcinoma. Proc Am Soc Clin Oncol 1989; 8:388. Bukowski RM, Balcerzak SP, O'Bryan RM, Bonnet JD, Chen TT. Randomised trial of 5 ¯uorouracil and mitomycin C with or without streptozotocin for advanced pancreatic cancer: a Southwest Oncology Group Study. Cancer 1983; 52(9): 1577±82. Oster MW. Chemotherapy for advanced pancreatic cancerÐ a comparison of ¯uorouracil, adriamycin and mitomycin (FAM) with ¯uorouracil, streptotozotocin and mitomycin (FSM). Cancer 1986; 57: 29±33. Group GTS. Phase II studies of drug combinations in advanced pancreatic carcinomaЯuorouracil plus doxoru-

962

112

113

114 115 116

117 118

119

120

121

122

123

124

125

126

127

A. HAYCOX et al. bicin plus mitomycin C and two regimens of streptotozotocin plus mitomycin C plus ¯uorouracil. J Clin Oncol 1986; 4: 1794±8. Kelsen D, Hudis C, Niedzwiecki D, et al. A Phase III comparison trial of streptozocin, mitomycin and 5 ¯uorouracil with cisplatin, cytosine, arabinocide and caffeine in patients with advanced pancreatic carcinoma. Cancer 1991; 68(5): 965±9. Crown J, Casper ES, Botet J, Murray P, Kelsen DP. Lack of ef®cacy of high dose leucovorine and ¯uorouracil in patients with advanced pancreatic adenocarcinoma. J Clin Oncol 1991; 9(9): 1682±6. Schilder RJ, et al. Phase II trial of PALA/5 ¯uorouracil (5-FU) in pancreatic cancer. Proc Am Soc Clin Oncol 1990; 9: 109. Greenway BA. Carcinoma of the exocrine pancreas: a sex hormone responsive tumour? Br J Surg 1987; 74: 441±2. Enochs MR, Johnson MR. Trophic effects of gastrointestinal hormones: physiological implications. Fed Proc 1977; 36: 1942±7. Woodruff GN, Hughes J. Cholecystokinin antagonists. Ann Rev Pharmacol Toxicol 1991; 31: 469±501. Duggan KA, Hams G, MacDonald GJ. Modi®cation of renal and tissue cation transport by the cholecystokinin octapeptide in the rabbit. J Physiol 1988; 397: 527±38. Pinho E, et al. Synergistic enhancement of cisplatin cytotoxicity by the cholecystokinin antagonist MK-329 in pancreatic cancer. Proc Am Assoc Cancer Res 1992; 33: 551. Jamshidipour R, Pinho EB, Howell SB. In vivo modulation of cisplatin cytotoxicity by the cholecystokinin antagonist MK329 in human pancreatic cancer xenografts. Anti Cancer Res 1994; 14: 2611±16. Wong A, Chan A, Arthur K. Tamoxifen therapy in unresectable adenocarcinoma of the pancreas. Cancer Treatment Rep 1987; 71(7±8): 749±50. Bakkevold KE, Pettersen A, Arnesjo B, Espehaug B. Tamoxifen therapy in unresectable adenocarcinoma of the pancreas and the papilla of vater. Br J Surg 1990; 77: 725± 30. Moriai T, Makino TT, Ishii K. Intratumoral treatment of pancreatic cancer with mitomycin-C absorbed to activated carbon particles. a clinical trial on ®fteen cases. Anti Cancer Res 1989; 9: 1799±804. Otsuji E, Yamaguchi T, Tsuruta H, et al. The effect of intravenous and intratumoral chemotherapy using a monoclonal antibody±drug conjugate in a xenograft model of pancreatic cancer. Eur J Surg Oncol 1995; 21: 61±5. Matsuoka H, Sugimachi K, Kuwano H, Yano K. Intratumoral injection of calcium channel blocker enhances cytotoxicity of adriamycin in Ehrlich ascites solid tumor. Eur J Surg Oncol 1989; 15(3): 224±31. Deurloo MJM, Bohlken S, Kop W, et al. Intratumoral adminstration of cisplatin in slow release devices. Cancer Chemother Pharmacol 1990; 27(2): 135±40. Bosset JF, Pavy JJ, Gillet M, Mantion G, Pelissier E, Schraub S. Conventional external irradiation alone as adjuvant treatment in resectable pancreatic cancer: results of a prospective study. Radiother Oncol 1992; 24(3): 191±4.

128 Dobelbower RR, Dronn DG. Radiotherapy in the treatment of pancreatic cancer. Bailire's Clin Gastroenterol 1990; 4: 969± 83. 129 Dobelbower RR, Borgelt BB, Strubler KA, Kutcher GJ, Suntharalingam N. Precison radiotherapy for cancer of the pancreas: techniques and results. Int J Radiation Oncol, Biol, Phys 1980; 6: 1127±33. 130 Nguyen TD, Bugat R, Combes PF. Postoperative irradiation of carcinoma of the head of the pancreas areaÐshort-term tolerance and results to precise high dose technique in 18 patients. Cancer 1982; 50(1): 53±6. 131 Treurniet-Donker AD, van Mierlo MJM, van Putten WLJ. Localised unresectable pancreatic cancer. Int J Radiation Oncol, Biol, Phys 1990; 18: 59±62. 132 Whittington R, Dobelbower RR, Mohiuddin M, Rosato FE, Weiss SM. Radiotherapy of unresectable pancreatic carcinomas: a six-year experience with 104 patients. Int J Radiation Oncol, Biol, Phys 1981; 7(12): 1639±44. 133 Group GTS. Further evidence of effective adjuvant combined radiation and chemotherapy following curative resection of pancreatic cancer. Cancer 1987; 59: 2006±10. 134 Group GTS. Treatment of locally unresectable carcinoma of the pancreas: comparison of combined modality therapy to chemotherapy alone. J Nat Cancer Inst 1988; 80: 751±5. 135 Kalser MH, Ellenberg SS. Pancreatic cancer: adjuvant combined radiation and chemotherapy following curative resection. Arch Surg 1985; 120: 899±903. 136 Blitzer PH, Dosoretz DE, Katin MJ, et al. Radiation and chemotherapyÐtreatment of localised pancreatic carcinoma. Florida Med Assoc 1994; 81(6): 403±7. 137 Moertel CG, Childs DS, Reitemeier RJ, Colby MY, Holbrook MA. Combined 5-¯uorouracil and super voltage radiation therapy of locally unresectable gastrointestinal cancer. Lancet 1969; 2: 865±7. 138 Moertel CG, Frytak S, Hahn RG, et al. Therapy of locally unresectable pancreatic carcinoma: a randomised comparison of high dose (6000 rads) radiation alone, moderate dose radiation (4000 rads) plus 5-¯uorouracil and high dose radiation plus 5-¯uorouracil. Cancer 1981; 48(8): 1705± 10. 139 Klaassen DJ, MacIntyre JM, Catton GE, Engstrom PF, Moertel CG. Treatment of locally unresectable cancer of the stomach and pancreas: a randomised comparison of 5-¯uorouracil alone with radiation plus concurrent and maintenance 5¯uorouracil. An Eastern Cooperative Oncology Group Study. J Clin Oncol 1985; 3(3): 373±8. 140 Bramhall SR, Neoptolemos JP. Adjuvant treatment of pancreatic cancer. In: Neoptolemos JP and Lemoine NR, eds. Pancreatic Cancer: Molecular and Clinical Advances. Oxford: Blackwell, 1996. 141 Bruckner HW, Kalnicki S, Dalton J, et al. Combined modality therapy increasing local control of pancreatic cancer. Cancer Invest 1993; 11(3): 241±6. 142 Komaki R, Cox JD, Hansen R, Anderson T, Wilson JF. Phase I±II study of prophylactic hepatic irradiation with local irradiation and systemic chemotherapy for adenocarcinoma of

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

REVIEW: TREATMENT OF PANCREATIC C ANCER

143

144

145

146 147

148

149

150

151

152

153

154 155

156

157

158

the pancreas. Int J Radiation Oncol, Biol, Phys 1988; 15(2): 1447±52. Gunderson LL, Martin JK, Kvols LK, et al. Intraoperative and external beam irradiation plus 5-FU for locally advanced pancreatic cancer. Int J Radiation Oncol, Biol, Phys 1987; 13(3): 319±29. Goertz SR, Ali MM, Parker GA. Local management of pancreatic carcinoma: iodine-125 implantation. Clin Oncol 1990; 2: 22±6. Sindelar WF. Intraoperative radiotherapy in carcinoma of the stomach and pancreas. Recent Results Cancer Res 1988; 110: 226±43. Pilepich MV, Miller HH. Preoperative irradiation in carcinoma of the pancreas. Cancer 1980; 46: 1945±9. Coia L, Hoffman J, Scher R, et al. Preoperative chemoradiation for adenocarcinoma of the pancreas and duodenum. Int J Radiation Oncol, Biol, Phys 1994; 30(1): 161±7. Fossati V, Cattaneo GM, Zerbi A, et al. The role of intraoperative therapy by electron beam and combination of adjuvant chemotherapy and external radiotherapy in carcinoma of the pancreas. Tumori 1995; 81(1): 23±31. Merrick HW, Dobelbower RR. Aggressive therapy for cancer of the pancreas: does it help? Gastroenterol Clin North Am 1990; 19: 935±62. Dobelbower RR, Merrick HW, Ahuja RK, Skeel RT. 125 Interstitial implant, precision high dose external beam therapy and 5-FU for unresectable adenocarcinoma of the pancreas and extrahepatic biliary tree. Cancer 1986; 58(10): 2185± 92. Shipley WU, Wood WC, Tepper JE, et al. Intraoperative electron beam irradiation for patients with unresectable pancreatic carcinoma. Ann Surg 1984; 200(3): 289±96. Seydel HG, Stablein DM, Leichman LP, Kinzie JJ, Thomas PRM. Hyperfractionated radiation and chemotherapy for unresectable localised adenocarcinoma of the pancreas. The GITSG experience. Cancer 1990; 65(7): 1478±82. Dobelbower RR, et al. Interstitial implant, precision high-dose external beam therapy and 5-FU for unresectable adenocarcinoma of pancreas and extrahepatic biliary tree. Cancer 1986; 58: 2185±95. Nori D, Hilaris BS. Intraoperative brachytherapy in pancreatic cancer. Cancer Invest 1988; 6(6): 695±704. Hoffman JP, Weese JL, Solin LJ, et al. A single institutional experience with preoperative chemoradiotherapy for stage I± III pancreatic adenocarcinoma. Am Surg 1993; 59(11): 772±81. Ishikawa O, Kitamura T, Wada A, Sasaki Y, Imaoka S, Iwanaga T. Clinical and histopathological appraisal of preoperative irradiation for adenocarcinoma of the pancreatoduodenal region. J Surg Oncol 1989; 40(3): 143±51. Weese JL, Nussbaum ML, Paul AR, et al. Increased resectability of locally advanced pancreatic and periampullary carcinoma with neoadjuvant chemoradiotherapy. Int J Pancreatol 1990; 7(1±3): 177±85. Rich TA, Evans DB, Curley SA, Ajani JA. Adjuvant radiotherapy and chemotherapy for biliary and pancreatic cancer. Ann Oncol 1994; 5: 75±80.

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964

963

159 Dobelbower RR, Konski AA, Merrick W, Bronn DG, Schifeling D, Kamen C. Intra-operative electron beam radiation therapy for carcinoma of the exocrine pancreas. Int J Radiation Oncol, Biol, Phys 1991; 20(1): 113±19. 160 Heijmans HJ, Hoekstra HJ, Mehta DM. Is adjuvant intraoperative radiotherapy for resectable and unresectable pancreatic carcinoma worthwhile? Hepatogastroenterology 1989; 36(6): 474±7. 161 Bagne FR, Dobelbower RR, Milligan AJ, Brown DG. Treatment of cancer of the pancreas by intraoperative electron beam therapy: physical and biological aspects. Int J Radiation Oncol, Biol Phys 1989; 16(1): 231±42. 162 Dirk-Noyes R, Weiss SM, Krall JM, et al. Surgical complications of intraoperative radiation therapy: the Radiation Therapy Oncology Group experience. J Surg Oncol 1992; 50(4): 209±15. 163 Roldan GE, Gunderson LL, Nagorney DM, et al. External beam versus intraoperative and external beam irradiation for locally advanced pancreatic cancer. Cancer 1988; 61(6): 1110±16. 164 Hiraoka T, Uchina R, Kanemitsu K, et al. Combination of intraoperative radiation with resection of cancer of the pancreas. Int J Pancreatol 1990; 7(1±3): 201±7. 165 Yamaue H, Tanimura H, Aoki Y, et al. Clinical and immunological evaluation of intraoperative radiation therapy for patients with unresectable pancreatic cancer. J Surg Oncol 1992; 49: 10±15. 166 Yang L, Hwang R, Pandit L, Gordon EM, Anderson F, Parekh D. Gene therapy of metastatic pancreatic cancer with intraperitoneal injections of concentrated retroviral herpes thymidine kinase vector supernatant and gancyclovir. Ann Surg 1996; 224: 405±17. 167 Lemoine NR, Hughes CM, Barton CM, et al. The epidermal growth factor receptor in human pancreatic cancer. J Pathol 1992; 166(1): 7±12. 168 Friess H, Buchler MW, Korc M. Growth factors and growth factor receptors in pancreatic cancer. In: Neoptolemos JP, Lemoine NR, eds. Pancreatic Cancer: Molecular and Clinical Advances. Oxford: Blackwell, 1996: 51±60. 169 Smith JJ, Derynck R, Korc M. Production of transforming growth factor a in human pancreatic cells: evidence for a superagonist autocrine cycle. Proc Natl Acad Sci 1987; 84(21): 7567±70. 170 Ura H, Obara T, Nishino N, Tanno S, Okamura K, Namiki M. Cytotoxicity of simvastatin to pancreatic adenocarcinoma cells containing mutant ras gene. Jpn J Cancer Res 1994; 85(6): 633±8. 171 Schmiegel W, Schmielau J, Heringlake S. Immunotherapeutical potential and mechanism of TNF-a enhanced EGFreceptor expression in pancreatic cancer cells. Digestion 1990; 46: 176. 172 Watanabe N, Yamauchi N, Maeda M, et al. Recombinant human tumor necrosis factor causes regression in patients with advanced malignancies. Oncology 1994; 51: 360±5. 173 Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other diseases. Nat Med 1995; 1: 27±31.

964

A. HAYCOX et al.

174 Voest EE, Kenyon BM, Truitt G, Folkman J. Inhibition of angiogenesis in vivo by interleukin 12. J Natl Cancer Inst 1995; 87: 581±6. 175 Bramhall SR, Stamp GWH, Lemoine N, Neoptolemos JP. The matrix metalloproteinases and their inhibitors in pancreatic cancer. In: Neoptolemos JP, Lemoine NR, eds. Pancreatic Cancer: Molecular and Clinical Advances. Oxford: Blackwell, 1996: 68±91. 176 Evans JD, Bramhall SR, Stark A, et al. A phase II trial of marimastat (BB-2516) in advanced pancreatic cancer. Int J Pancreatol 1996; 19: 218A.

177 Boscoff WH, et al. Continuous infusion 5 ¯uorouracil, calcium leucovorin, mitomycin C and dipyrididamole treatment for patients with locally advanced pancreatic cancer. Proc Am Soc Clin Oncol 1995; 14: 471. 178 Alanen KA, Joensuu H. Long term survival after pancreatic adenocarcinoma Ð often a misdiagnosis? Br J Cancer 1993; 68: 1004±5.

Ó 1998 Blackwell Science Ltd, Aliment Pharmacol Ther 12, 949±964