palliative treatment of neuroendocrine tumors

6
Seminars in Surgical Oncology 9453-458 (1993) Palliative Treatment of Neuroendocrine Tumors J. GREGORY McKINNON, MD From the Department of Surgery, University of Calgary, Calgary, Alberta, Canada Effective palliation of patients with incurable neuroendocrine tumors requires both control of hormonal overproduction symptoms as well as control of tumor growth. Several important advances have been made in recent years toward these two goals. Octreotide and ome- prazole have both been extremely effective in ameliorating hormonal symptoms of carcinoids, islet cell tumors and medullary thyroid carci- noma. Newer cytotoxic chemotherapy regimens and interferon have increased response rates over traditional therapy. More aggressive surgical extirpation of metastatic disease has also been beneficial. @ 1993 Wiley-Liss, Inc. KEY WORDS: APUDoma, carcinoid, islet cell tumor, pheochromocytoma, thyroid neoplasm INTRODUCTION Although neuroendocrine tumors present a myriad of different and challenging clinical problems, they tend to share a number of common characteristics. The natural history of these tumors is usually one of slow growth over a period of 10- 15 years, which may be marked by dramatic clinical syndromes of hormone overproduction [l]. Once it has been determined that a tumor cannot be resected for cure, the treating physi- cian is faced with two tasks. The first is to control symptoms produced by hormone over production, which may be immediately life-threatening. The sec- ond is to attempt prolongation of survival by limiting the growth or ablating metastatic tumor tissue. Occa- sionally the same therapeutic maneuver may accom- plish these two goals. This review attempts to outline remarkable advances made in the last decade toward these two goals. CONTROL OF HORMONAL SYMPTOMS Carcinoid Tumors The natural history of carcinoid tumors is quite variable depending on their site of origin. Metastatic rate is proportional to size and averages 70% for tu- mors of the small intestine that are 1-2 cm, and higher for tumors larger than 2 cm [2]. Colorectal carcinoids seem to have a more aggressive course, with the meta- static rate of tumors greater than 2 cm being 74% with an overall 5-year survival of 37% [3]. Once hepatic metastases have developed, outlook is dismal with a five year survival ranging from zero to 47% [4-61. Virtually all neuroendocrine tumors contain secre- tory granules, but most are not functionally significant [7]. Carcinoid tumors occasionally secrete a number of hormonally active peptides that are thought to medi- ate carcinoid syndrome, a constellation of symptoms including diarrhea, flushing, bronchoconstriction and tissue fibrosis. Although the mechanism of these symptoms is poorly understood, it is mediated at least in part by histamine, serotonin, prostaglandin E and F, and tachykinins [8]. Fortunately, the treatment is broadly effective and does not depend on complete delineation of hormonal effects. Estimates of inci- dence of carcinoid syndrome have varied widely de- pending on the location of the primary and how dili- gently it is searched for. Feldman suggested that 84% of all patients with carcinoids had elevated levels of serotonin, although not all were clinically significant [9]. Norheim reported carcinoid syndrome in 67% of patients with metastatic tumor at time of presentation [ 101. Small intestine carcinoids produce the syndrome only when metastasized to the liver. Foregut carcin- oids have been known to produce systemic symptoms Address reprint requests to Dr. J.G. McKinnon, 3330 Hospital Dr. N.W., Calgary, Alberta T2N 4N1, Canada. 0 1993 Wiley-Liss, Inc.

Upload: dr-j-gregory-mckinnon

Post on 06-Jul-2016

215 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: Palliative treatment of neuroendocrine tumors

Seminars in Surgical Oncology 9453-458 (1993)

Palliative Treatment of Neu roendocrine Tumors

J. GREGORY McKINNON, MD

From the Department of Surgery, University of Calgary, Calgary, Alberta, Canada

Effective palliation of patients with incurable neuroendocrine tumors requires both control of hormonal overproduction symptoms as well as control of tumor growth. Several important advances have been made in recent years toward these two goals. Octreotide and ome- prazole have both been extremely effective in ameliorating hormonal symptoms of carcinoids, islet cell tumors and medullary thyroid carci- noma. Newer cytotoxic chemotherapy regimens and interferon have increased response rates over traditional therapy. More aggressive surgical extirpation of metastatic disease has also been beneficial. @ 1993 Wiley-Liss, Inc.

KEY WORDS: APUDoma, carcinoid, islet cell tumor, pheochromocytoma, thyroid neoplasm

INTRODUCTION Although neuroendocrine tumors present a myriad

of different and challenging clinical problems, they tend to share a number of common characteristics. The natural history of these tumors is usually one of slow growth over a period of 10- 15 years, which may be marked by dramatic clinical syndromes of hormone overproduction [l]. Once it has been determined that a tumor cannot be resected for cure, the treating physi- cian is faced with two tasks. The first is to control symptoms produced by hormone over production, which may be immediately life-threatening. The sec- ond is to attempt prolongation of survival by limiting the growth or ablating metastatic tumor tissue. Occa- sionally the same therapeutic maneuver may accom- plish these two goals. This review attempts to outline remarkable advances made in the last decade toward these two goals.

CONTROL OF HORMONAL SYMPTOMS Carcinoid Tumors

The natural history of carcinoid tumors is quite variable depending on their site of origin. Metastatic rate is proportional to size and averages 70% for tu- mors of the small intestine that are 1-2 cm, and higher for tumors larger than 2 cm [2]. Colorectal carcinoids seem to have a more aggressive course, with the meta- static rate of tumors greater than 2 cm being 74% with

an overall 5-year survival of 37% [3]. Once hepatic metastases have developed, outlook is dismal with a five year survival ranging from zero to 47% [4-61.

Virtually all neuroendocrine tumors contain secre- tory granules, but most are not functionally significant [7]. Carcinoid tumors occasionally secrete a number of hormonally active peptides that are thought to medi- ate carcinoid syndrome, a constellation of symptoms including diarrhea, flushing, bronchoconstriction and tissue fibrosis. Although the mechanism of these symptoms is poorly understood, it is mediated at least in part by histamine, serotonin, prostaglandin E and F, and tachykinins [8]. Fortunately, the treatment is broadly effective and does not depend on complete delineation of hormonal effects. Estimates of inci- dence of carcinoid syndrome have varied widely de- pending on the location of the primary and how dili- gently it is searched for. Feldman suggested that 84% of all patients with carcinoids had elevated levels of serotonin, although not all were clinically significant [9]. Norheim reported carcinoid syndrome in 67% of patients with metastatic tumor at time of presentation [ 101. Small intestine carcinoids produce the syndrome only when metastasized to the liver. Foregut carcin- oids have been known to produce systemic symptoms

Address reprint requests to Dr. J.G. McKinnon, 3330 Hospital Dr. N.W., Calgary, Alberta T2N 4N1, Canada.

0 1993 Wiley-Liss, Inc.

Page 2: Palliative treatment of neuroendocrine tumors

454 McKinnon

in the absence of metastases, although this is unusual [lo].

The most important advance made in the treatment of carcinoid syndrome in recent years has been the development of octreotide, the long-acting synthetic analogue of somatostatin [ 1 11. Since the introduction of this drug, numerous trials have assessed its efficacy in virtually all neuroendocrine tumors, both for symp- tom reduction and for antitumor toxicity. In carcinoid syndrome, octreotide has been extremely successful in ameliorating or abolishing flushing and diarrhea with an 80- 100% complete response. This has been accom- panied by a concomitant reduction in urine SHIAA [12-141. Octreotide is given as a chronic subcutaneous injection of 50-100 pg three times daily. Side effects have been minimal except for pain at the injection site and the development of gallstones [ 151. Recently, somatostatin receptors have been identified in carci- noid liver metastases by needle biopsy. Their presence may prove predictive of response to somatostatin treatment [16].

Anecdotal reports have suggested that carcinoid syndrome refractory to octreotide alone may be re- sponsive to a combination of Interferon-a and octreo- tide [ 171. Recently, cyproheptadine, a serotonin inhibi- tor, was shown to be successful in alleviating flushing and diarrhea although it was not tolerated in all pa- tients [18].

Gastrinoma The Zollinger-Ellison syndrome is the result of un-

controlled tumor production of gastrin from islet cell tumors. The syndrome is characterized by peptic ul- ceration and diarrhea with their associated complica- tions. It has been estimated that with an aggressive approach to localization and resection, at least 20% of patients with sporadic disease should be cured by sur- gical exploration [19]. Gastrinoma in multiple endo- crine neoplasia-type I (MEN-I) syndrome tends to be multicentric and is difficult or impossible to cure with resection [20]. These patients tend to become chronic management problems. Fortunately, malignancy rate in MEN associated tumors tends to be less than in the sporadic form, and survival with metastatic disease somewhat longer [20,21]. In those patients who are noncurable by surgical means, success in controlling gastrin induced acid hypersecretion has led to long- term survival. Ultimate mortality is usually due to malignant tumor progression. In a study of noncura- ble islet-cell tumors, Thompson reported a 3-year sur- vival of 56% [22]. Gastrinomas had a significantly bet- ter prognosis than nonfunctioning islet cell tumors [221.

Complete control of acid secretion is now possible

with the introduction of the H + K + ATPase inhibi- tor, omeprazole [23]. This drug has proved extraor- dinarily effective for long-term control of hypergas- trinemia and has largely supplanted the role of H-2 blockers and gastrectomy. Although initial animal studies suggested that long term use might result in the formation of gastric enterochromaffin-like tumors (ECL), omeprazole has been proved safe in long-term human trials [24-271.

Octreotide has been assessed for its effect on gas- trinoma and been found to control symptoms in the majority of patients [28]. However it was not effective in those patients with metastatic tumor with high lev- els of gastrin production [28]. Its role in gastrinoma therefore, is restricted to those patients who cannot tolerate omeprazole or high dose H-2 blockers. It should also be remembered that in patients with MEN-I syndrome, control of acid hypersecretion can sometimes be greatly facilitated by control of any pre- existing hyperparathyroidism [20,21].

Insulinoma Insulin secreting islet cell tumors are quite rare. Of

those that occur, the majority are benign, that is, no metastases are detected at time of surgical exploration and resection of tumor results in long-term cure in approximately 90% of cases [29]. In patients with metastasized or unresectable disease, control of severe hypoglycemia is problematic. Traditional methods of hypoglycemia control include diet and use of diazox- ide. The mechanism of diazoxide is unknown and its efficacy somewhat unpredictable [30]. Nevertheless it controls symptoms in approximately 60% of patients [31]. As with other islet cell tumors, octreotide has been reported to have a beneficial effect in patients with unresectable insulinoma [32], although resistance to therapy eventually develops with progression of disease [33]. Other therapies necessitate cytoreduction, as discussed later.

Other Islet Cell Tumors VIPoma, previously known as Verner-Morrison

syndrome, is characterized by uncontrolled release of vasoactive intestinal peptide. This results in a severe form of diarrhea with dehydration, hypokalemia and acidosis, aptly described as pancreatic cholera [34]. Glucagonoma is a rare tumor where high levels of glucagon lead to a syndrome of migratory necrolytic rash, mild diabetes and severe muscle wasting [31]. If followed long enough, both of these islet cell tumors are usually found to be malignant, unlike insulinomas. Both syndromes are responsive to octreotide, particu- larly VIPomas [34-371. Treatment of glucagonoma with octreotide has been less well documented but

Page 3: Palliative treatment of neuroendocrine tumors

Neuroendocrine Tumors 455

appears to be effective in controlling all the syn- drome’s symptoms [38,39]. The extremely rare somatostatinoma is characterized by diarrhea, stea- torrhea, cholelithiasis, and weight loss. The majority are reported to be malignant and treatment of meta- static disease is cytoreductive [40]. Not surprisingly, octreotide has been tried without success [41].

Medullary Carcinoma of the Thyroid This disease may be indolent, with the tumor dor-

mant for many years [42]. However, overall 10-year survival is only 50% [43]. Unresectable metastatic tumor may be nonfunctional or may result in uncon- trollable watery diarrhea. As with other neuroendo- crine tumors, these symptoms may be controlled with octreotide [44]. However results have not been consist- ent [45].

Pheochromocytoma Malignant pheochromocytoma is a rare disease. In

a large series of pheochromocytomas unrelated to MEN syndromes, 13% were eventually found to be malignant [46]. Metastases may occur in bone, lung, mediastinum or liver and are characterized by local symptoms and excess catecholamine release [47]. Con- trol of tumor-induced catecholamine effects are possi- ble with standard a- and P-adrenergic blockade with agents such as phenoxybenzamine and propranolol. Overall long term survival once metastases are present is 44% [48].

CYTOTOXIC THERAPY The alternative strategy for palliation of incurable

neuroendocrine tumors, which may be complemen- tary to hormone blockade, is reduction or elimination of tumor burden. The usual anticancer strategies of surgery, radiation and cytotoxic chemotherapy can all be effective in the proper circumstances. In addition to these more traditional forms of treatment, there are more specific strategies that may be useful, such as radioisotope therapy.

Chemotherapy Treatment of disseminated neuroendocrine car-

cinomas with cytotoxic chemotherapy has not been very encouraging, with poor response rates and seri- ous toxicity [49]. For carcinoids and islet cell tumors, streptozotocin has been long used with varying suc- cess. More recently this drug has been used in combi- nation with other cytotoxic and biological agents. A multicenter trial demonstrated streptozotocin + dox- orubicin to result in 69% tumor response rate [50]. A new derivative of streptozotocin, chlorozotocin was as effective in combination with doxorubicin but with

fewer side effects [SO]. However, another recent trial showed a modest response in only 19% of patients with advanced neuroendocrine tumors with combined streptozotocin and doxorubicin [5 11. A combination of etoposide and cisplatin has been reported to result in 67% response rate in tumors classified as anaplastic [52]. Although side effects were severe, duration of response ranged from 8-24 months. Continuous infu- sion of 5FU has resulted in complete response of islet cell tumors but these reports are anecdotal [53].

Cytotoxic chemotherapy for metastatic pheo- chromocytoma has had limited success with a different group of agents [54,55]. Averbruch et al. reported a complete and partial response rate of 57% with a com- bination of cyclophosphamide, vincristine, and dacar- bazine, a regimen originally used for pediatric neuro- blastoma [56].

Radiation Therapy External beam radiotherapy is rarely indicated in

neuroendocrine tumors, except for palliation of bone metastases. However there has been some success with specific isotope therapy, particularly in metastatic pheochromocytoma. * 311-MIBG accumulates in cate- cholamine storage granules and is selectively taken up in pheochromocytoma tumor tissue in a manner simi- lar to l3II in thyroid carcinoma. Recent reports sug- gest a 60% response rate [57,58]. There is some hope of using the same strategy in other tumors that take up I-MIBG such as medullary thyroid carcinoma, but no clinical trials have been reported yet.

Interferon Interferon has been investigated as a new and excit-

ing agent for treating neuroendocrine tumors. Early results reported by Oberg were extremely encourag- ing, with objective tumor response and amelioration of symptoms seen in the majority of patients [59-611. A prospective randomized trial of 20 patients showed human leukocyte interferon to be superior to strep- tozotocin and 5FU with 10 out of 10 patients treated with interferon showing regression or stable disease [62]. However, other investigators using recombinant interferon-a have been less successful. An ameliora- tion in hormonal symptoms is sometimes seen, al- though not as complete as with octreotide, but with little or no objective tumor response [63-651. Efficacy may be limited by the formation of neutralizing anti- bodies in patients as early as 3 months into treatment [66]. These antibodies have been reported to occur more often, with interferona2b than with interferon-a 2a [67]. Side effects such as fever, anorexia, and weight loss have also been serious [68,69], and there has been

Page 4: Palliative treatment of neuroendocrine tumors

456 McKinnon

one reported case of induction of a systemic lupus erythematosus (SLE)-like syndrome with human leu- kocyte-derived interferon [70].

In experimental human xenografts, interferon plus dimethylfluorornithine was superior to IFN alone. This has not reached human trials as yet [71].

Octreotide Octreotide's remarkable success in symptom con-

trol of neuroendocrine tumors led researchers to in- vestigate its possible role as a cytoreductive agent [72,- 731. Unfortunately, an observed effect in animal models and anecdotal reports has not been borne out in rigorous trials [13,15,74]. It has been tested in meta- static medullary carcinoma of the thyroid, with some fall in calcitonin levels seen but no measurable tumor response [45].

SURGICAL THERAPY Liver Metastasis Resection

Unlike most other visceral cancers, neuroendocrine malignancies tend to be slow growing, with much of their morbidity caused by autonomous hormone pro- duction. For these two reasons, cytoreductive surgery, that is resection or debulking of metastatic disease, is often beneficial and sometimes curative. Norton re- ported a series of five patients with metastatic gas- trinoma who had resection of all gross disease, includ- ing both liver and pancreas and followed by postoperative chemotherapy [75]. Four of five patients had all gross disease resected and subsequently re- quired decreased dosage of antisecretory medication. Two remained free of disease at 14 and 32 months. McEntee et al. recommended palliative resective sur- gery for islet cell tumors, even if some gross disease was left behind [76]. Eight of 16 patients with sympto- matic endocrinopathies obtained complete relief after incomplete resection of disease. Six of seven under- going curative resection obtained relief [76].

Insulinoma is slow growing with symptoms that are difficult to control medically. It is therefore particu- larly attractive to attempt ablation of gross metastatic disease. Danforth et al. reported a small series of met- astatic insulinomas which were treated with curative resection [77]. The median disease-free survival after curative resection was 5 years. The recurrence rate was 63%, with the median interval to recurrence being 2.8 years, and the median survival with recurrent tumor 19 months. Palliative resection was associated with a median survival of 4 years, and biopsy only, 11 months.

The traditional criteria and technical limitations of resective surgery of the liver have been expanded by new techniques. Ravikumar et al. reported a small series of cryosurgical ablation of metastatic neuroendocrine tu- mors [78]. Information on follow-up is limited.

Recently, a number of groups have begun using liver transplantation as a therapeutic modality for pa- tients with unresectable metastatic disease [79]. Starzl reported a series of five patients who underwent resec- tion of the primary along with liver transplantation, for metastatic disease from neuroendocrine tumors of gut origin, two of which were functional [80]. Three of five are alive and free of disease at a median of 12 months follow-up. Other reports are anecdotal but are encouraging, at least in the short term [81,82].

Hepatic Artery Embolization When other surgical and medical treatments prove

to be ineffective and metastases are limited to the liver, which is usually the case with carcinoids and islet cell tumors, hepatic arterial embolization may provide useful palliation. Because liver tumors are preferen- tially supplied by the hepatic artery and the normal liver parenchyma survives with portal circulation alone, interruption of the arterial supply can lead to ischemia and infarction of the metastases. Maton re- ported a series of 13 patients with carcinoid liver metastases treated with hepatic artery embolization, with reduction in tumor bulk and a decrease in hor- monal production [83]. Side effects, however, were se- rious with one mortality and major morbidity in the majority of patients. Other investigators have re- ported similar results in other islet cell tumors [84,85]. Selective embolization appears to be more effective than hepatic artery interruption.

Combination Therapy In view of the many different categories of therapy

available for neuroendocrine tumors, several groups have advocated an organized combination of thera- pies for patients with disseminated disease using surgi- cal, hormonal and cytotoxic therapy. It is difficult to evaluate the relative merit of these regimens because of the rarity of the disease and the lack of randomized trials. However some have reported improved survival compared to historical controls [86-881.

CONCLUSIONS The goals of therapeutic intervention for palliative

care of patients with incurable neuroendocrine tumors must be clear before embarking on potentially toxic therapy. Control of hormonal mediated symptoms and secondly, inhibition of growth or ablation of tumor are the common endpoints. The means to achieve these ends and the price to be paid by the patient must be individualized according to the type and behavior of the tumor and the overall status of the patient. Several important strides have been made in recent years in therapy for these tumors. The develop- ment of octreotide and l3*I-MIBG, the more effective use of cytotoxic chemotherapy and the more aggres-

Page 5: Palliative treatment of neuroendocrine tumors

Neuroendocrine Tumors 457

sive use of cytoreductive surgery have all been of bene- fit. It seems clear that most of these patients have relatively indolent tumors with a long natural history, and that an aggressive approach to treatment with all available means will continue to improve the length and quality of their lives.

REFERENCES 1. Eriksson B, Oberg, Skogseid B: Neuroendocrine pancreatic

tumors. Clinical findings in a prospective study of 84 patients. Acta Oncol 28:373-377, 1989.

2. Moertel CG: Karnofsky memorial lecture: An odyssey in the land of small tumors. J Clin Oncol 5:1503, 1987.

3. Ballantyne GH, Savoca PE, Flannery JT, et al: Incidence and mortality of carcinoids of the colon. Data from the Connecti- cut Tumor Registry. Cancer 69:2400-2405, 1992.

4. Nwiloh JO, Pillarisetty S, Moscovic EA, Freeman-HP: Carci- noid tumors. J Surg Oncol45:261-241, 1990.

5. Welch J P, Malt R A: Management of carcinoid tumors of the gastrointestinal tract. Surg Gynecol Obst, 145:223-227, 1977.

6. Thompson GB, van Heerden JA, Martin J K Carcinoid tu- mors of the gastrointestinal tract: Presentation, management, and prognosis. Surgery, 98:1054-1062, 1985.

7. Wood SM, Polak JM, Bloom SR: Gut hormone secreting tu- mours. Scand J Gastroenterol Suppl, 82:165-179, 1983.

8. Arnold R: Therapeutic strategies in the management of endo- crine GEP tumours. Eur J Clin Invest Suppl I:S82-90, 1990.

9. Feldman JM: Carcinoid tumors and syndrome. Semin Oncol 14:237-246, 1987.

10. Norheim I, Oberg K, Theodorsson-Norheim E: Malignant car- cinoid tumors. Ann Surg 206:115-125, 1987.

11. Bauer W, Brimer U, Deopfner W, et al: SMS 201-995: a very potent and selective octapeptide analogue of somotostatin with prolonged action. Life Sci 31:1133-1141, 1982.

12. Vinik A, Moattari AR: Use of somatostatin analog in manage- ment of carcinoid syndrome. Dig-Dis-Sci (3 Suppl):14S-27S, 1989.

13. Kvols LK, Buc-M, Moertel CG, et al: Treatment of metastatic islet cell carcinoma with a somatostatin analogue (SMS 201- 995). Ann Intern Med 107:162-168, 1987.

14. Gorden P. Comi RJ. Maton PN. Go VL: NIH conference.

15.

16.

17.

18.

19,

20.

21.

22.

23.

Somatosta’tin and somatostatin analogue (SMS 201-995) in treatment of hormone-secreting tumors of the pituitary and gastrointestinal tract and non-neoplastic diseases of the gut. Ann Intern Med 110:35-50, 1989. Wynick D, Bloom SR: Clinical Review 23: The use of the long-acting somatostatin analog octreotide in the treatment of gut neuroendocrine tumors. J Clin Endocrinol Metab 73: 1-3, 1991. Reubi JC, Kvols LK, Waser B, et al: Detection of somatostatin receptors in surgical and percutaneous needle biopsy samples of carcinoids and islet cell carcinomas. Cancer Res 50:5969- 5977, 1990. Joensuu H, Katka K, Kujari H: Dramatic response of a meta- static carcinoid tumour to a combination of interferon and octreotide. Acta Endocrinol Copenh 126: 184- 185, 1992. Moertel CG, Kvols LK, Rubin J: A study of cyproheptadine in the treatment of metastatic carcinoid tumor and the malig- nant carcinoid syndrome. Cancer 67:33-36, 1991. Berg CL, Wolfe MM: Zollinger-Ellison syndrome. Med Clin North Am 75:903-921, 1991. Sheppard BC, Norton JA, Doppman JL, et al: Management of islet cell tumors in patients with multiple endocrine neoplasia: A prospective study. Surgery. 106: 1108-1 117, 1989. Lamers CB: Gastrinoma in multiple endocrine neoplasia type 1. Acta Oncol 30:489-492, 1991. Thompson GB, van Heerden JA, Grant CS, et al: Islet cell carcinomas of the pancreas: A twenty-year experience. Sur- gery, 104:1011-1023, 1988. Zdon MJ, Ballantyne GH, Schafer DE, et al: Proton pump

inhibition-the ultimate control of acid secretion? J Surg Res 40:353-361, 1986.

24. Ekman L, Hansson E, Havu N, et al: Toxicological studies on omeprazole. Scand J Gastroenterol Suppl 108:53-69, 1985.

25. McArthur KE, Collen MJ, Maton PN, et al: Omeprazole: Effective, convenient therapy for Zollinger-Ellison syndrome. Gastroenterology 88:939-944, 1985.

26. Delchier JC, Soule JC, Mignon M, et al: Effectiveness of ome- prazole in seven patients with Zollinger-Ellison syndrome re- sistant to histamine HZreceptor antagonists. Dig Dis Sci, 31 : 693-699, 1986.

27. Maton PN, Vinayek R, Frucht H, et al: Long-term efficacy and safety of omeprazole in patients with Zollinger-Ellison syn- drome: A prospective study. Gastroenterology 972327-836, 1989.

28. Vinik Al, Tsai S, Moattari AR, Cheung P: Somatostatin ana- logue (SMS 201-995) in patients with gastrinomas. Surgery

29. Rothmund-M, Angelini-L, Brunt-LM, et al: Surgery for be- nign insulinoma: An international review. World J Surg 14:

30. Arnold RT: Therapeutic strategies in the management of endo- crine GEP tumours. Eur J Clin Invest 20 (Suppl I):S82-90, 1990.

31. Boden G Insulinoma and glucagonoma. Semin Oncol 14:253- 262, 1987.

32. Glaser B, Rosler A, Halperin Y: Chronic treatment of a benign insulinoma using the long-acting somatostatin analogue SMS 201-995. Isr J Med Sci 26:16-19, 1990.

33. Wynick D, Anderson JV, Bloom SR: Resistance of metastatic pancreatic endocrine tumors after long term treatment with the somatostatin analogue octreotide (SMS 201-995). Clin Endo- crinol 30:385-388, 1988.

34. Maton PN, O’Dorisio TM, Howe BA, et al: Effect of a long- acting somatostatin analogue (SMS 201-995) in a patient with pancreatic cholera. N Engl J Med 312:17-21, 1985.

35. Kraenzlin ME, Ch’ng JL, Wood SM, et al: Long-term treat- ment of a VlPoma with somatostatin analogue resulting in remission of symptoms and possible shrinkage of metastases. Gastroenterology 88: 185-187, 1985.

36. Maton PN, Gardner JD, Jensen RT: Use of long-acting somatostatin analog SMS 201-995 in patients with pancreatic islet cell tumors. Dig Dis Sci 34(3 Suppl):28S-39S, 1989.

37. O’Dorisio TM, Gaginella TS, Mekhjian HS, et al: Somatosta- tin and analogues in the treatment of VIPoma. Ann NY Acad Sci, 527:528-535, 1988.

38. Moattari AR, Cho K, Vinik Al: Somatostatin analogue in treatment of coexisting glucagonoma and pancreatic pseudo- cyst: Dissociation of responses. Surgery 108:581-587, 1990.

39. Boden G, Ryan IG, Eisenschmid BL, et al: Treatment of inop- erable glucagonoma with the long-acting somatostatin ana- logue SMS 201-995. N Engl J Med 314:1686-1689, 1986.

40. Vinik Al, Strodel WE, Eckhauser FE, et al: Somatostatinomas, PPomas, neurotensinomas. Semin Oncol 14:263-28 I , 1987.

41. Davis TM, Bray G, Domin J, Bloom SR: A case of somato- statinoma: Responses to food and SMS 201-995 administra- tion. Pancreas 3:729-733, 1988.

42. Ahuja S, Ernst H: Dormant metastases in medullary thyroid carcinoma. A case report. Exp Clin Endocrinol 98:37-41, 1991.

43. Tenvall J, Biorklund JL, Moller T, et al: Prognostic factors of papillary, follicular and medullary carcinoma of the thyroid gland. Acta Radio1 Oncol 24:17-24, 1985.

44. Jerkins TW, Sacks HS, ODorisio TM, et al: Medullary carci- noma of the thyroid, pancreatic nesidioblastosis and mi- croadenosis, and pancreatic polypeptide hypersecretion: A new association and clinical and hormonal responsees to long- acting somatostatin analogue SMS-201-995. J Clin Endocrinol Metab 641313-1319, 1987.

45. Modigliani E, Cohen R, Joannidis S, et al: Results of long-term continuous subcutaneous octreotide administration in 14 pa- tients with medullary thyroid carcinoma. Clin Endocrinol Oxf

46. Scott HW, Halter SA: Oncologic aspects of pheo-

104:834-842, 1988.

393-398, 1990.

36:183-6, 1992.

Page 6: Palliative treatment of neuroendocrine tumors

458 McKinnon

chromocytoma: The importance of follow-up. Surgery 96: 1061-1066, 1984.

47. Lewi HJ, Reid R, Mucci B, et al: Malignant phaeo- chromocytoma. Br J Urol 57:394-398. 1985.

48. Averbuch SD, Steakley CS; Young RC: Malignant pheo- chromocytoma: Effective treatment with a combination of cy- clophosphamide, vincristine, and dacarbazine. Ann Intern Med 109:267-273, 1988.

49. Rougier P, Oliveira J, Ducreux M. et al: Metastatic carci- noid and islet cell tumours of the pancreas: A phase 11 trial of the efficacy of combination chemotherapy with 5-fluorouracil, doxorubicin and cisplatin. Eur J Cancer 27: 1380-1382, 1991.

50. Moertel CG, Lefkopoulo M, Lipsitz S, et al: Streptozocin- doxorubicin, streptozocin-fluorouracil or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med 326:519-523, 1992.

51. Frame J. Kelsen D, Kemeny N, et al: A phase I1 trial of strcptozotocin and adriamycin in advanced APUD tumors. Am J Clin Oncol 11:490-495, 1988.

52. Moertel CG, Kvols LK. O’Connell MJ, Rubin JTI: Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplas- tic variants of these neoplasms. Cancer 68:227-232, 1991.

53. Hanscn R, Helm J, Wilson JF. Wilson S: Nonfunctioning islet cell carcinoma of the pancreas. Complete response to continu- ous 5-fluorouracil infusion. Cancer 62: 15- 17, 1988.

54. Srimuninnimit V, Wampler GL: Case report of metastatic fa- milial pheochromocytoma treated with cisplatin and 5-fluorouracil. Cancer Chemother Pharmacol 28:217-219. 1991.

55. Siddiqui MZ, Von-Eyben FE, Spanos GTI: High-voltage irra- diation and combination chemotherapy for malignant pheo- chromocytoma. Cancer 62:686-690, 1988.

56. Averbuch SD, Steakley CS, Young RC. et al: Malignant pheo- chromocytoma: Effective treatment with a combination of cy- clophosphamidc, vincristine. and dacarbazine. Ann Intern Med 109:267-273, 1988.

57. Krempf M, Lumbroso J, Mornex R, et al: Use of m-[l311]i- odobenzylguanidine in the treatment of malignant pheo- chromocytoma. J Clin Endocrinol Metab 72:455 -461, 1991.

58. Khafagi FA, Shapiro B, Fischer MTI, et al: Phaeo- chromocytoma and functioning paraganglioma in childhood and adolescence: Role of iodine 13 1 metaiodobenzylguanidine. Eur J Nucl Med 18:191-198, 1991.

59. Eriksson B, Oberg K , Alm G, et al: Treatment of malignant endocrine pancreatic tumours with human leucocyte inter- feron. Lancet, 2:1307-9, 1986.

60. Oberg K , Eriksson B: Medical treatment of neuroendocrine gut and pancreatic tumors. Acta Oncol 28:425-431, 1989.

61. Sheehan-Dare RA, Simmons AV, Cotterill JA, Janke PG: He- patic tumors with hyperglucagonemia. Response to treatmcnt with human lymphoblastoid interferon. Cancer 62:912-~914, 1988.

62. Oberg K, Norheim I, Alm G, et al: Treatment of malignant carcinoid tumors: a randomized controlled study of streptozo- cin plus 5-FU and human leukocyte interferon. Eur J Cancer Clin Oncol 25:1475-1479, 1989.

63. Creutzfeldt W, Bartsch HH, Jacubaschke U, Stockmann F: Treatment of gastrointestinal endocrine tumours with inter- feron-alpha and octreotide. Acta Oncol 30529-535, 1991.

64. Doberauer C. Mengelkoch B. Kloke 0, et al: Treatment of metastatic carcinoid tumors and the carcinoid syndrome with recombinant interferon alpha. Acta Oncol 30:603-605, 1991.

65. Valimaki M, Jarvinen H, Salmela P, et al: Is the treatment of metastatic carcinoid tumor with interferon not as successful as suggested? Cancer 67:547-549, 1991.

66. Oberg K, Alm GV: Development of neutralizing interferon antibodies after treatment with recombinant interferon-alpha 2b in patients with malignant carcinoid tumors. J Interferon Res. 9(Suppl l):S45-9, 1989.

67. Grander D, Oberg K, Lundqvist ML, et al: Interferon-induced enhancement of 2’,5’-oligoadenylate synthetase in mid-gut car- cinoid tumours. Lancet 336:337-340, 1990.

68. Basser RL, Lieschke GJ, Sheridan WP, et al: Recombinant alpha-2b interferon in patients with malignant carcinoid tu- mow. Aust NZ J Med 21375-878, 1991.

69. Ahren B, Engman K, Lindblom A: Treatment of malignant midgut carcinoid with a highly purified human leukocyte alpha-interferon. Anticancer Res 12: 129-133, 1992.

70. Ronnblom LE, Alm GV, Oberg KE: Possible induction of systemic lupus erythematosus by interferon-alpha treatment in a patient with a malignant carcinoid tumour. J Intern Med 227207-10, 1990.

71. Evers BM. Hurlbut SC, Tyring SK, et al: Novel therapy for the treatment of human carcinoid. Ann Surg 213:411-416, 1991.

72. Shepherd JJ, Senator GB: Regression of liver metastases in patient with gastrin-secreting tumour treated with SMS 201- 995. (Letter.) Lancet 2574, 1986.

73. Boden G, Ryan IG, Eisenschmid BL, et al: Treatment of inop- erable glucagonoma with the long-acting somatostatin ana- logue SMS 201-995. N Engl J Med 314:1686-1689, 1986.

74. Maton PN, Gardner JD, Jensen RT: Use of long-acting somatostatin analog SMS 201-995 in patients with pancreatic islet cell tumors. Dig Dis Sci 34(3 Suppl):28S-39S, 1989.

75. Norton JA, Sugarbaker PH, Doppman JL, et al: Aggressivc resection of metastatic disease in selected patients with malig- nant gastrinoma. Ann Surg 203:352-359, 1986.

76. McEntee GP, Nagorney DM, Kvols LK, et al: Cytoreductivc hepatic surgery for neuroendocrine tumors. Surgery 108:1091- 1096, 1990.

77. Danforth DN Jr, Gorden P; Brennan MF: Metastatic insulin- secreting carcinoma of the pancreas: clinical course and the role of surgery. Surgery 96:1027-1037, 1984.

78. Ravikumar TS, Kane R, Cady B, et al: A five year study of cryosurgery in the treatment of liver tumors. Arch Surg 126: 1520-1524, 1991.

79. Alsina AE, Bartus S, Hull D, et al: Liver transplant for meta- static ncuroendocrine tumor. J Clin Gastroenterol, 12533- 537, 1990.

80. Makowka L, Tzakis AG, Mazzaferro V, et al: Transplantation of the liver for metastatic endocrine tumors of the intestine and pancreas. J Clin Gastroenterol I2:533-537, 1990.

81. Penn I: Hepatic transplantation for primary and metastatic cancers of the liver. Surgery 110:726-734, 1991.

82. Lobe TE, Vera SR, Bowman LC, et al: Hepdticopan- creaticogastroduodenectomy with transplantation for meta- static islet cell carcinoma in childhood. J Pediatr Surg 27:227- 229, 1992.

83. Maton PN, Camilleri M, Griffin G, et al: Role of hepatic arterial embolisation in the carcinoid syndrome. Br Med J Clin Res 287:932-935, 1983.

84. Marlink RG, Lokich JJ, Robins JR, CIouse ME: Hepatic ar- terial embolization for metastatic hormone-secreting tumors. Technique, effectiveness, and complications. Cancer 65:2227- 2232, 1990.

85. Ajani JA, Carrasco CH, Charnsdngavej C, et al: Islet cell tu- mors metastatic to the liver: Effective palliation by sequential hepatic artery embolization. Ann Intern Med 108:340-344, 1988.

86. Hajarizadeh H, Ivancev K, Mueller CR, et al: Effective pallia- tive treatment of metastatic carcinoid tumors with intra-arter- ial chemotherapyichemoembolization combined with octreo- tide acetate. Am J Surg, 163:479-483, 1992.

87. Hanssen LE, Schrumpf E, Jacobsen MB, et al: Extended expe- rience with recombinant alpha-2b interferon with or without hepatic artery embolization in the treatment of midgut carci- noid tumours. A preliminary report. Acta Oncol, 30523-527, 1991.

88. Ahlman H, Wangberg B, Jansson S, et al: Management of disseminated midgut carcinoid tumours. Digestion 49:78-96, 1991.