• No results found

Neuroendokrina Tunntarmstumörer (SI-NETs) är indolenta neoplasier med en årlig incidens kring 1 / 100,000 personer. De är ofta diagnocerade senare i sjukdomsförloppet, vilket begränsar behandlingens effektivitet. Syftet med denna avhandlingen är att undersöka pre-kliniska och kliniska aspekter av patienter med avancerad och / eller spridd sjukdom, med speciellt fokus på hantering av intra-abdominell fibros, rollen av lokoregional kirurgi och levertransplantation, samt ex vivo sensitivitet av cytotoxiska substanser på tumörprover. Dessutom undersöktes nya serum biomarkörer för diagnos och prognos av SI-NETs.

Bakgrunden i delarbete I var att serotonin och andra cytokiner, som utsöndras från SI-NET tumörceller kan orsaka fibros, vilket leder till karcinoid hjärtsjukdom och fibrotiska reaktioner intraabdominellt. Syftet med arbetet var att studera förekomsten, kliniska komplikationer och hantering av den fibrotiska reaktionen i buken. Trettiosex patienter identifierades med signifikanta radiologiska tecken och symtom på fibros.

Av dessa hade 20 patienter central mesenteriell fibros, som orsakade tarmischemi och 16 hade retroperitoneal fibros, som orsakade obstruktiv uropati med hydronefros. Palliativa ingrepp i form av stentinläggning i Vena mesenterica superior eller resektion av centrala mesenteriella metastaser och/eller perkutan nefrostomi och J-stent behandling var fördelaktiga hos majoriteten av patienterna. Slutsatsen från detta delarbetet var att tidig diagnos och minimalinvasiva åtgärder var effektiva för sjukdomspalliation.

I delabete II undersöktes rollen av lokoregional kirurgi vid diagnos hos asymtomatiska patienter med SI-NET och fjärrmetastaser. Data erhölls från vår Uppsala databas för SI-NET och det Nationella Patientregistret.

Trehundrasextiotre patienter med SI-NET utan lokala tumörrelaterade symptom vid diagnos uppdelades i två grupper, varav en grupp som opererades inom sex månader från diagnos och en kontrollgrupp, som antingen behandlades icke-kirurgiskt eller opererades senare i sjukdomsförloppet. ”Propensity score” matchning utfördes för att reducera risken för selektions-bias. De matchade grupperna visade ingen skillnad i total överlevnad, 30-dagars postoperativ mortalitet och morbiditet samt vårdtid vid sjukhus. Däremot visar studien att gruppen som opererades vid diagnos genomgick flera re-operationer pga tarmobstruktion. Slutsatsen är

att man generellt inte kan rekommendera lokoregional kirurgi i ett profylaktiskt syfte vid diagnos hos asymptomatiska SI-NET patienter vid stadium IV.

I delarbete III undersöktes rollen av levertransplantation (LTx) hos patienter med levermetastaserade SI-NET. Från Uppsaladatabasen för SI-NET identifierades 78 patienter, som uppfyllde befintliga LTx kriterier: <65 år, radikalt genomförd lokoregional kirurgi och avsaknad av extra-hepatisk sjukdom. Patienterna genomgick behandling enligt de kliniska protokollen vid vårt centrum, och under denna tidsperiod utfördes inte några LTx.

Resultaten i detta delarbete visar att de flesta unga patienter (<65 år) med SI-NET och levermetastaser har en lång överlevnad med standardiserad multimodal behandling, dvs utan LTx, och att de flesta överlevnadssiffror som rapporteras efter LTx för NET i andra centra inte överträffar dessa siffror.

I delarbete IV undersöktes nya diagnostiska och prognostiska biomarkörer vid SI-NET. Etthundrafyrtiofem biomarkörer i plasma hos 96 patienter med SI-NET och 23 friska kontrollpersoner undersöktes med metoderna PLA och PEA (proximity ligation assay och proximity extension assay, Olink bioscience). Nitton biomarkörer vid PLA och 17 vid PEA visade sig ha olika serumnivåer mellan patienter och friska kontroller. Tre markörer studerades vidare; DcR3, TFF3 och Midkine. Alla dessa proteiner uttrycktes i tumörvävnad enligt immunohistokemiskt analys. Resultatet visar att höga serum nivåer av DcR3 och TFF3 var associerat med en sämre överlevnad.

DcR3 är en markör för levermetastaser och TFF3 samt Midkine är nya diagnostiska biomarkörer vid SI-NET tidigt i sjukdomsförloppet.

SI-NET anses vara allmänt resistenta mot systemisk behandling förutom somatostatinanaloger och alfa-interferon. I delarbete V undersökte vi denna tumörtyp med hänsyn till sensitivitet ex vivo mot en panel av kemoterapeutiska och riktade medel med användning av ”fluorometric microculture cytotoxicity assay” (FMCA). Tumörceller från patientprover med njurcancer, kolorektal cancer, äggstockscancer och kronisk lymfocytär leukemi användes som jämförelse. SI-NET-proverna analyserades även i förhållande till kliniska-patologiska variabler (ålder vid diagnos, stadium, peritoneal carcinomatos, extra-abdominella metastaser och grad) samt befintliga biomarkörer, (serum-Chromogranin A och urin-5HIAA).

Resultaten av detta delarbetet visar att tumörceller från SI-NET uppvisar variabel men generellt intermediär sensitivitet ex vivo jämfört med andra tumördiagnoser. Kliniska-patologiska faktorer och befintliga biomarkörer var inte associerade till sensitivitet ex vivo och utmanade dessa kriterier för behandlingsbeslut i SI-NET. Den stora variationen i ex vivo sensitivitet

innebär att det sannolikt finns utrymme för individuellt anpassad behandling där fler faktorer inklusive ex vivo sensitiviteten vägs in.

Acknowledgements

I would like to express my sincere gratitude to the following people, who have helped and supported me along the way.

Professor Peter Stålberg, my main supervisor, for introducing me to both basic and clinical scientific research, for the highly educative conversations we had, and most importantly for his support and exceptional guidance throughout all parts of this thesis.

Professor Per Hellman, my co-supervisor, for his encouragement and kindness and for sharing his invaluable expertise with me.

Associate Professor Olov Norlén, my co-supervisor, for his rapid and insightful advice, outstanding statistical skills and also his positive spirit.

Professor Peter Nygren, my co-supervisor, for concise and detailed instructions, as well as excellent support in the final article in this thesis.

Katarina Edfeldt, my co-author, for valuable instructions and help in the laboratory in the fourth article in the thesis.

Andreas Karakatsanis, my co-author, for his valuable help, enthusiasm and stimulating conversations.

Birgitta Bondeson for her great technical assistance in tissue preparation and immunohistochemical staining in the laboratory.

My other co-authors: Eva Tiensuu Janson, Kjell Öberg, Staffan Welin, Heather C Stuart, Ola Hessman, Göran Åkerström, Gunnar Westin and Christopher Bäcklin for valuable contributions to the articles in this thesis.

Claes Juhlin and Kristiina Kask, former and present Head of the Department of Surgery, as well as Professor Olle Nilsson and Professor Per Hellman, former and current Chair of the Department of Surgical Sciences, for providing the means for clinical work and scientific research.

Ola Hessman, Head of the Endocrine Section and former clinical supervisor, for teaching me his surgical skills, sharing his deep knowledge in the field and supporting me.

My sister Argyro for artistic contributions in the front page and some of the figures in the thesis and also my little sister Despoina for her support.

My parents Markos and Euthalia for supporting every step in my life, for the principles they inspired in me and for their unconditional love.

My two sons Markos and Aristides, for bringing light and joy into my life.

My wife Julia, for her love and understanding. It is her patience and strength that have made this thesis possible.

References

1. Kloppel G. Oberndorfer and his successors: from carcinoid to neuroendocrine carcinoma. Endocr Pathol. 2007;18(3):141-144.

2. Modlin IM, Shapiro MD, Kidd M. Siegfried Oberndorfer: origins and perspectives of carcinoid tumors. Hum Pathol. 2004;35(12):1440-1451.

3. Williams ED, Sandler M. The classification of carcinoid tum ours. Lancet.

1963;1(7275):238-239.

4. Rindi G, Kloppel G, Alhman H, et al. TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system.

Virchows Arch. 2006;449(4):395-401.

5. Gress D, Edge S, Greene F, Washington M, Asare E, Brierley J. Principles of cancer staging

AJCC Cancer Staging Manual (8th ed.). Springer, New York; 2017.

6. Niederle B, Pape UF, Costa F, et al. ENETS Consensus Guidelines Update for Neuroendocrine Neoplasms of the Jejunum and Ileum.

Neuroendocrinology. 2016;103(2):125-138.

7. Lloyd RV, Osamura RY, Klöppel G, et al. WHO classification of tumours of endocrine organs. 4th edition. ed.

8. Velayoudom-Cephise FL, Duvillard P, Foucan L, et al. Are G3 ENETS neuroendocrine neoplasms heterogeneous? Endocr Relat Cancer.

2013;20(5):649-657.

9. Dasari A, Shen C, Halperin D, et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA Oncol. 2017.

10. Yao JC, Hassan M, Phan A, et al. One hundred years after "carcinoid":

epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26(18):3063-3072.

11. Bilimoria KY, Bentrem DJ, Wayne JD, Ko CY, Bennett CL, Talamonti MS.

Small bowel cancer in the United States: changes in epidemiology, treatment, and survival over the last 20 years. Ann Surg. 2009;249(1):63-71.

12. Ito T, Sasano H, Tanaka M, et al. Epidemiological study of gastroenteropancreatic neuroendocrine tumors in Japan. J Gastroenterol.

2010;45(2):234-243.

13. Guo Z, Li Q, Wilander E, Ponten J. Clonality analysis of multifocal carcinoid tumours of the small intestine by X-chromosome inactivation analysis. J Pathol. 2000;190(1):76-79.

14. Norlen O, Stalberg P, Oberg K, et al. Long-term results of surgery for small intestinal neuroendocrine tumors at a tertiary referral center. World J Surg.

2012;36(6):1419-1431.

15. Landerholm K, Falkmer S, Jarhult J. Epidemiology of small bowel carcinoids in a defined population. World J Surg. 2010;34(7):1500-1505.

16. Modlin IM, Oberg K, Chung DC, et al. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol. 2008;9(1):61-72.

17. Institute NC. SEER Registry Groupings for Analyses.

https://seer.cancer.gov/registries/terms.html. Accessed 16 October 2017.

18. Zar N, Garmo H, Holmberg L, Rastad J, Hellman P. Long-term survival of patients with small intestinal carcinoid tumors. World J Surg.

2004;28(11):1163-1168.

19. Talback M, Rosen M, Stenbeck M, Dickman PW. Cancer patient survival in Sweden at the beginning of the third millennium--predictions using period analysis. Cancer Causes Control. 2004;15(9):967-976.

20. Frilling A, Sotiropoulos GC, Radtke A, et al. The impact of 68Ga-DOTATOC positron emission tomography/computed tomography on the multimodal management of patients with neuroendocrine tumors. Ann Surg.

2010;252(5):850-856.

21. Yao JC, Pavel M, Phan AT, et al. Chromogranin A and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with everolimus. J Clin Endocrinol Metab. 2011;96(12):3741-3749.

22. Meijer WG, Kema IP, Volmer M, Willemse PH, de Vries EG. Discriminating capacity of indole markers in the diagnosis of carcinoid tumors. Clin Chem.

2000;46(10):1588-1596.

23. Formica V, Wotherspoon A, Cunningham D, et al. The prognostic role of WHO classification, urinary 5-hydroxyindoleacetic acid and liver function tests in metastatic neuroendocrine carcinomas of the gastroenteropancreatic tract. Br J Cancer. 2007;96(8):1178-1182.

24. Lollgen RM, Hessman O, Szabo E, Westin G, Akerstrom G. Chromosome 18 deletions are common events in classical midgut carcinoid tumors. Int J Cancer. 2001;92(6):812-815.

25. Stalberg P, Westin G, Thirlwell C. Genetics and epigenetics in small intestinal neuroendocrine tumours. J Intern Med. 2016.

26. Edfeldt K, Ahmad T, Akerstrom G, et al. TCEB3C a putative tumor suppressor gene of small intestinal neuroendocrine tumors. Endocr Relat Cancer. 2014;21(2):275-284.

27. Karpathakis A, Dibra H, Pipinikas C, et al. Prognostic Impact of Novel Molecular Subtypes of Small Intestinal Neuroendocrine Tumor. Clin Cancer Res. 2016;22(1):250-258.

28. Launay JM, Birraux G, Bondoux D, et al. Ras involvement in signal transduction by the serotonin 5-HT2B receptor. J Biol Chem.

1996;271(6):3141-3147.

29. Lundin L, Norheim I, Landelius J, Oberg K, Theodorsson-Norheim E.

Carcinoid heart disease: relationship of circulating vasoactive substances to ultrasound-detectable cardiac abnormalities. Circulation. 1988;77(2):264-269.

30. Kidd M, Modlin I, Shapiro M, et al. CTGF, intestinal stellate cells and carcinoid fibrogenesis. World J Gastroenterol. 2007;13(39):5208-5216.

31. Druce M, Rockall A, Grossman AB. Fibrosis and carcinoid syndrome: from causation to future therapy. Nat Rev Endocrinol. 2009;5(5):276-283.

32. Hellman P, Hessman O, Akerstrom G, et al. Stenting of the superior mesenteric vein in midgut carcinoid disease with large mesenteric masses.

World J Surg. 2010;34(6):1373-1379.

33. Norlen O, Stalberg P, Zedenius J, Hellman P. Outcome after resection and radiofrequency ablation of liver metastases from small intestinal neuroendocrine tumours. Br J Surg. 2013;100(11):1505-1514.

34. Elias D, Lefevre JH, Duvillard P, et al. Hepatic metastases from neuroendocrine tumors with a "thin slice" pathological examination: they are many more than you think. Ann Surg. 2010;251(2):307-310.

35. Garcia-Carbonero R, Salazar R, Sevilla I, Isla D. SEOM clinical guidelines for the diagnosis and treatment of gastroenteropancreatic neuroendocrine tumours (GEP NETS). Clin Transl Oncol. 2011;13(8):545-551.

36. Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 Trial of 177Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med. 2017;376(2):125-135.

37. Caplin ME, Pavel M, Ruszniewski P. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(16):1556-1557.

38. Rinke A, Muller HH, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol. somatostatin analog in patients with gastroenteropancreatic neuroendocrine carcinoma: single agent or combination? Ann Oncol. 2007;18(1):13-19.

41. Molina-Cerrillo J, Alonso-Gordoa T, Martinez-Saez O, Grande E. Inhibition of Peripheral Synthesis of Serotonin as a New Target in Neuroendocrine Tumors. Oncologist. 2016;21(6):701-707.

42. Sarmiento JM, Heywood G, Rubin J, Ilstrup DM, Nagorney DM, Que FG.

Surgical treatment of neuroendocrine metastases to the liver: a plea for resection to increase survival. J Am Coll Surg. 2003;197(1):29-37.

43. Makridis C, Oberg K, Juhlin C, et al. Surgical treatment of mid-gut carcinoid tumors. World J Surg. 1990;14(3):377-383; discussion 384-375.

44. Frilling A, Akerstrom G, Falconi M, et al. Neuroendocrine tumor disease: an evolving landscape. Endocr Relat Cancer. 2012;19(5):R163-185.

45. Ohrvall U, Eriksson B, Juhlin C, et al. Method for dissection of mesenteric metastases in mid-gut carcinoid tumors. World J Surg. 2000;24(11):1402-1408.

46. Lardiere-Deguelte S, de Mestier L, Appere F, et al. Toward Preoperative Classification of Lymph-Node Metastases in Patients with Small Intestine Neuroendocrine Tumours in the Era of Intestinal-Sparing Surgery.

Neuroendocrinology. 2015.

47. Makridis C, Rastad J, Oberg K, Akerstrom G. Progression of metastases and symptom improvement from laparotomy in midgut carcinoid tumors. World J Surg. 1996;20(7):900-906; discussion 907.

48. Niederle B, Pape UF, Costa F, et al. ENETS Consensus Guidelines Update for Neuroendocrine Neoplasms of the Jejunum and Ileum.

Neuroendocrinology. 2016;103(2):125-138.

49. Capurso G, Rinzivillo M, Bettini R, Boninsegna L, Delle Fave G, Falconi M.

Systematic review of resection of primary midgut carcinoid tumour in patients with unresectable liver metastases. Br J Surg. 2012;99(11):1480-1486.

50. Pape UF, Perren A, Niederle B, et al. ENETS Consensus Guidelines for the management of patients with neuroendocrine neoplasms from the jejuno-ileum and the appendix including goblet cell carcinomas.

Neuroendocrinology. 2012;95(2):135-156.

51. National Comprehensive Cancer Network.

https://www.nccn.org/professionals/physician_gls/pdf/neuroendocrine.pdf.

2017.

52. Lehnert T. Liver transplantation for metastatic neuroendocrine carcinoma: an analysis of 103 patients. Transplantation. 1998;66(10):1307-1312.

53. Le Treut YP, Gregoire E, Klempnauer J, et al. Liver transplantation for neuroendocrine tumors in Europe-results and trends in patient selection: a 213-case European liver transplant registry study. Ann Surg.

2013;257(5):807-815.

54. Le Treut YP, Gregoire E, Belghiti J, et al. Predictors of long-term survival after liver transplantation for metastatic endocrine tumors: an 85-case French multicentric report. Am J Transplant. 2008;8(6):1205-1213.

55. Frilling A, Malago M, Weber F, et al. Liver transplantation for patients with metastatic endocrine tumors: single-center experience with 15 patients. Liver Transpl. 2006;12(7):1089-1096.

56. Bonaccorsi-Riani E, Apestegui C, Jouret-Mourin A, et al. Liver transplantation and neuroendocrine tumors: lessons from a single centre experience and from the literature review. Transpl Int. 2010;23(7):668-678.

57. Mazzaferro V, Pulvirenti A, Coppa J. Neuroendocrine tumors metastatic to the liver: how to select patients for liver transplantation? J Hepatol.

2007;47(4):460-466.

58. O'Toole D, Grossman A, Gross D, et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: biochemical markers.

Neuroendocrinology. 2009;90(2):194-202.

59. Turner GB, Johnston BT, McCance DR, et al. Circulating markers of prognosis and response to treatment in patients with midgut carcinoid tumours. Gut. 2006;55(11):1586-1591.

60. Korse CM, Taal BG, Vincent A, et al. Choice of tumour markers in patients with neuroendocrine tumours is dependent on the histological grade. A marker study of Chromogranin A, Neuron specific enolase, Progastrin-releasing peptide and cytokeratin fragments. Eur J Cancer. 2012;48(5):662-671.

61. Kema IP, de Vries EG, Muskiet FA. Measurement of 5-HIAA in urine. Ann Clin Biochem. 1995;32 ( Pt 1):102-104.

62. Sun W, Lipsitz S, Catalano P, Mailliard JA, Haller DG, Eastern Cooperative Oncology G. Phase II/III study of doxorubicin with fluorouracil compared with streptozocin with fluorouracil or dacarbazine in the treatment of advanced carcinoid tumors: Eastern Cooperative Oncology Group Study E1281. J Clin Oncol. 2005;23(22):4897-4904.

63. Faure M, Niccoli P, Autret A, Cavaglione G, Mineur L, Raoul JL. Systemic chemotherapy with FOLFOX in metastatic grade 1/2 neuroendocrine cancer.

Mol Clin Oncol. 2017;6(1):44-48.

64. Kulke MH, Hornick JL, Frauenhoffer C, et al. O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors. Clin Cancer Res. 2009;15(1):338-345.

65. Qian ZR, Ter-Minassian M, Chan JA, et al. Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol. 2013;31(27):3418-3425.

66. Scoazec JY. Angiogenesis in neuroendocrine tumors: therapeutic applications. Neuroendocrinology. 2013;97(1):45-56.

67. Yao JC, Fazio N, Singh S, et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet.

2016;387(10022):968-977.

68. Pavel ME, Hainsworth JD, Baudin E, et al. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet. 2011;378(9808):2005-2012.

69. Anthony LB, Pavel ME, Hainsworth JD, et al. Impact of Previous Somatostatin Analogue Use on the Activity of Everolimus in Patients with Advanced Neuroendocrine Tumors: Analysis from the Phase III RADIANT-2 Trial. Neuroendocrinology. RADIANT-2015;10RADIANT-2(1-RADIANT-2):18-RADIANT-25.

70. Lundberg M, Thorsen SB, Assarsson E, et al. Multiplexed homogeneous proximity ligation assays for high-throughput protein biomarker research in serological material. Mol Cell Proteomics. 2011;10(4):M110 004978.

71. Assarsson E, Lundberg M, Holmquist G, et al. Homogenous 96-plex PEA immunoassay exhibiting high sensitivity, specificity, and excellent scalability. PLoS One. 2014;9(4):e95192.

72. Blokzijl A, Nong R, Darmanis S, Hertz E, Landegren U, Kamali-Moghaddam M. Protein biomarker validation via proximity ligation assays.

Biochim Biophys Acta. 2014;1844(5):933-939.

73. Blumenthal RD. An overview of chemosensitivity testing. Methods Mol Med.

2005;110:3-18.

74. Cashin PH, Mahteme H, Graf W, Karlsson H, Larsson R, Nygren P. Activity ex vivo of cytotoxic drugs in patient samples of peritoneal carcinomatosis with special focus on colorectal cancer. BMC Cancer. 2013;13:435.

75. Rosenbaum PR, Rubin DB. The Central Role of the Propensity Score in Observational Studies for Causal Effects. Biometrika. 1983;70(1):41-55.

76. Horton KM, Kamel I, Hofmann L, Fishman EK. Carcinoid tumors of the small bowel: a multitechnique imaging approach. AJR Am J Roentgenol.

2004;182(3):559-567.

77. Janssen W, Schymura Y, Novoyatleva T, et al. 5-HT2B receptor antagonists inhibit fibrosis and protect from RV heart failure. Biomed Res Int.

2015;2015:438403.

78. Svejda B, Kidd M, Giovinazzo F, et al. The 5-HT(2B) receptor plays a key regulatory role in both neuroendocrine tumor cell proliferation and the modulation of the fibroblast component of the neoplastic microenvironment.

Cancer. 2010;116(12):2902-2912.

79. Pantongrag-Brown L, Buetow PC, Carr NJ, Lichtenstein JE, Buck JL.

Calcification and fibrosis in mesenteric carcinoid tumor: CT findings and pathologic correlation. AJR Am J Roentgenol. 1995;164(2):387-391.

80. Hellman P, Lundstrom T, Ohrvall U, et al. Effect of surgery on the outcome of midgut carcinoid disease with lymph node and liver metastases. World J Surg. 2002;26(8):991-997.

81. Nebigil CG, Launay JM, Hickel P, Tournois C, Maroteaux L. 5-hydroxytryptamine 2B receptor regulates cell-cycle progression: cross-talk with tyrosine kinase pathways. Proc Natl Acad Sci U S A. 2000;97(6):2591-2596.

82. Kulke MH, O'Dorisio T, Phan A, et al. Telotristat etiprate, a novel serotonin synthesis inhibitor, in patients with carcinoid syndrome and diarrhea not adequately controlled by octreotide. Endocr Relat Cancer. 2014;21(5):705-714.

83. Jian B, Xu J, Connolly J, et al. Serotonin mechanisms in heart valve disease I: serotonin-induced up-regulation of transforming growth factor-beta1 via G-protein signal transduction in aortic valve interstitial cells. Am J Pathol.

2002;161(6):2111-2121.

84. Nilsson CL, Brodin E, Ekman R. Substance P and related peptides in porcine cortex: whole tissue and nuclear localization. J Chromatogr A.

1998;800(1):21-27.

85. Yamakado K, Tanaka N, Nakatsuka A, Matsumura K, Takase K, Takeda K.

Clinical efficacy of portal vein stent placement in patients with hepatocellular carcinoma invading the main portal vein. J Hepatol. 1999;30(4):660-668.

86. Zhou ZQ, Lee JH, Song KB, et al. Clinical usefulness of portal venous stent in hepatobiliary pancreatic cancers. ANZ J Surg. 2014;84(5):346-352.

87. Mertens S, Zeegers AG, Wertheimer PA, Hendriksz TR, van Bommel EF.

Efficacy and complications of urinary drainage procedures in idiopathic retroperitoneal fibrosis complicated by extrinsic ureteral obstruction. Int J Urol. 2014;21(3):283-288.

88. Givi B, Pommier SJ, Thompson AK, Diggs BS, Pommier RF. Operative resection of primary carcinoid neoplasms in patients with liver metastases yields significantly better survival. Surgery. 2006;140(6):891-897; discussion 897-898.

89. Ahmed A, Turner G, King B, et al. Midgut neuroendocrine tumours with liver metastases: results of the UKINETS study. Endocr Relat Cancer.

2009;16(3):885-894.

90. van der Horst-Schrivers AN, Post WJ, Kema IP, et al. Persistent low urinary excretion of 5-HIAA is a marker for favourable survival during follow-up in patients with disseminated midgut carcinoid tumours. Eur J Cancer.

2007;43(18):2651-2657.

91. Strosberg J, Gardner N, Kvols L. Survival and prognostic factor analysis of 146 metastatic neuroendocrine tumors of the mid-gut. Neuroendocrinology.

2009;89(4):471-476.

92. Soreide O, Berstad T, Bakka A, et al. Surgical treatment as a principle in patients with advanced abdominal carcinoid tumors. Surgery.

1992;111(1):48-54.

93. Stump R, Haueis S, Kalt N, et al. Transplantation and surgical strategies in patients with neuroendocrine liver metastases: protocol of four systematic reviews. JMIR Res Protoc. 2013;2(2):e58.

94. Caplin ME, Pavel M, Cwikla JB, et al. Anti-tumour effects of lanreotide for

94. Caplin ME, Pavel M, Cwikla JB, et al. Anti-tumour effects of lanreotide for

Related documents