Herramientas para el abordaje diagnóstico de los tumores neuroendocrinos de páncreas

Autores/as

  • Yenny Fernanda Guzmán Universidad de los Andes
  • Rocío del Pilar López Universidad de los Andes
  • Alonso Vera Universidad de los Andes
  • Deyanira González Devia Universidad de los Andes

DOI:

https://doi.org/10.30944/20117582.50

Palabras clave:

páncreas, neoplasias pancreáticas, tumores neuroendocrinos, diagnóstico, biomarcadores de tumor

Resumen

Introducción. Los tumores neuroendocrinos de páncreas son relativamente raros y heterogéneos. Sin embargo, su incidencia se ha incrementado a nivel mundial, y los avances en el diagnóstico y el tratamiento han mejorado la supervivencia. Tienen un pronóstico más favorable que el adenocarcinoma de páncreas, pero el reconoci- miento y el abordaje diagnóstico son complejos y requieren un equipo humano multidisciplinario entrenado. Objetivo. Actualizar al médico en el abordaje clínico, patológico, imaginológico y genético, y en la evaluación hormonal basada en la evidencia disponible, brindando herramientas y recomendaciones específicas para las diferentes circunstancias clínicas.

Conclusión. La incidencia de los tumores neuroendocrinos de páncreas en los últimos 40 años ha aumentado en más del 600 %, y corresponden a la segunda neoplasia pancreática con gran mortalidad. Actualmente, dispo- nemos de múltiples biomarcadores para caracterizarlos y plantear un tratamiento más personalizado.

Descargas

Los datos de descargas todavía no están disponibles.

Biografía del autor/a

Yenny Fernanda Guzmán, Universidad de los Andes

Médica, Universidad de los Andes, Bogotá, D.C., Colombia

Rocío del Pilar López, Universidad de los Andes

Médica patóloga, Departamento de Patología y Laboratorios; jefe, Sección de Biología Molecular, Hospital Universitario Fundación Santa Fe de Bogotá; profesora clínica, Facultad de Medicina, Universidad de los Andes, Bogotá, D.C., Colombia

Alonso Vera, Universidad de los Andes

Médico, cirujano general, cirujano de trasplantes; jefe, Servicio de Trasplantes y Cirugía Hepatobiliar, Hospital Universitario Fundación Santa Fe de Bogotá; profesor clínico, Facultad de Medicina, Universidad de los Andes, Bogotá, D.C., Colombia

Deyanira González Devia, Universidad de los Andes

Médica internista, endocrinóloga, Hospital Universitario Fundación Santa Fe de Bogotá; profesora clínica, Facultad de Medicina, Universidad de los Andes, Bogotá, D.C., Colombia

Referencias bibliográficas

Halperin D. Neuroendocrine Tumors. En: Kantarji- an H, Wolff R, editors. The MD Anderson Manual of Medical Oncology. Third edition. New York: The Mc- Graw-Hill Companies, Inc; 2016.

Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017;77030:1-8. doi: 10.1001/jamaoncol.2017.0589.

https://doi.org/10.1001/jamaoncol.2017.0589

Gonzalez-Devia D, López-Panqueva R, Taboada-Bar- rios L. Experiencia diagnóstica en tumores neuroen- docrinos del Hospital Universitario FSFB, 2001-2010. Bogotá: Fundación Santa Fe de Bogota; 2011.

Chiruvella A, Kooby DA. Surgical management of pancreatic neuroendocrine tumors. Surg Oncol Clin N Am. 2016;25:401-21. doi: 10.1016/j.soc.2015.12.002.

https://doi.org/10.1016/j.soc.2015.12.002

Mougery A, Adler DG. Neuroendocrine tumors: Re- view and clinical update. Hosp Physician. 2007;51:12- 21.

Thakker RV. Molecular and cellular endocrinology multiple endocrine neoplasia type 1 (MEN1) and type 4 (MEN4). Mol Cell Endocrinol. 2014;386:2-15. doi: 10.1016/j.mce.2013.08.002.

https://doi.org/10.1016/j.mce.2013.08.002

Kulke MH, Benson AB 3rd, Bergsland E, Berlin JD, Blaszkowsky LS, Choti MA, et al. Neuroendocrine tu- mors: Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2012;10:724-64.

https://doi.org/10.6004/jnccn.2012.0075

Agarwal SK, Mateo CM, Marx SJ. Rare germline mu- tations in cyclin-dependent kinase inhibitor genes in multiple endocrine neoplasia type 1 and related states. J Clin Endocrinol Metab. 2009;94:1826-34. doi: 10.1210/jc.2008-2083.

https://doi.org/10.1210/jc.2008-2083

Minnetti M, Grossman A. Somatic and germline mu- tations in NETs: Implications for their diagnosis and management. Best Pract Res Clin Endocrinol Metab. 2017;30:115-27. doi: 10.1016/j.beem.2015.09.007.

https://doi.org/10.1016/j.beem.2015.09.007

Tamm EP, Bhosale P, Lee JH, Rohren EM. State-of- the-art imaging of pancreatic neuroendocrine tu- mors. Surg Oncol Clin N Am. 2016;25:375-400. doi: 10.1016/j.soc.2015.11.007.

https://doi.org/10.1016/j.soc.2015.11.007

Bosman FT, Carneiro F, Hruban RH, Theise ND. WHO Classification of tumours of the digestive system. Fourth edition. Int Agency Res Cancer. 2010;3:382-95.

Halfdanarson TR, Rabe KG, Rubin J, Petersen GM. Pancreatic neuroendocrine tumors (PNETs): Inci- dence, prognosis and recent trend toward improved survival. Ann Oncol. 2008;19:1727-33. doi: 10.1093/an- nonc/mdn351.

https://doi.org/10.1093/annonc/mdn351

Liu JB, Baker M. Surgical management of pan- creatic neuroendocrine tumors. Surg Clin N Am. 2016;96:1447-68. doi: 10.1016/j.suc.2016.07.002.

https://doi.org/10.1016/j.suc.2016.07.002

Tang LH, Untch BR, Reidy DL, Reilly EO, Dhall D, Jih L, et al. Well differentiated neuroendocrine tu- mors with a morphologically apparent high grade component: A pathway distinct from poorly differen- tiated neuroendocrine carcinomas. Clin Cancer Res. 2016;22:1011-7. doi: 10.1158/1078-0432.

https://doi.org/10.1158/1078-0432.CCR-15-0548

Chi C, Klimstra D. Somatostatin receptor expres- sion related to TP53 and RB1 alterations in pancre- atic and extrapancreatic neuroendocrine neoplasms with a Ki67-index above 20%. Semin Diagn Pathol. 2014;31:498-511. doi: 10.1038/modpathol.2016.217.

https://doi.org/10.1038/modpathol.2016.217

Milione M, Spada F, Sessa F, Capella C, La S. The clinicopathologic heterogeneity of grade 3 gastro- enteropancreatic neuroendocrine neoplasms: Mor- phological differentiation and proliferation identify different prognostic categories. Neuroendocrinology. 2017;104:85-93. doi: 10.1159/000445165.

https://doi.org/10.1159/000445165

Perren A, Couvelard A, Scoazec J, Costa F, Borbath I, Delle Fave G, et al. ENETS consensus guidelines for the standards of care in neuroendocrine tumors. Pa- thology: Diagnosis and prognostic stratification. Neu- roendocrinology. 2017;105:196-200.

https://doi.org/10.1159/000457956

Lloyd R, Osamura R, Klöpel G, Rosai J. WHO clas- sification of tumours of endocrine organs. Fourth edition. Vol. 10. Lyon, France: IARC Classification of Tumours; 2017. p. 209-39.

Smith S, Brick A, O'Hara S, Normand C. Evidence on the cost and cost-effectiveness of palliative care: A literature review. Palliat Med. 2014;28:130-50. doi: 10.1177/0269216313493466.

https://doi.org/10.1177/0269216313493466

Luo G, Jin K, Cheng H, Guo M, Lu Y. Pancreatology revised nodal stage for pancreatic neuroendocrine tumors. Pancreatology. 2017;17:1-6. doi: 10.1016/j. pan.2017.06.003.

Anderson CW, Bennett JJ. Clinical presentation and diagnosis of pancreatic neuroendocrine tumors. Surg Oncol Clin N Am. 2016;25:363-74. doi: 10.1016/j. soc.2015.12.003.

https://doi.org/10.1016/j.soc.2015.12.003

Alshaikh OM, Yoon JY, Chan BA, Krzyzanowska MK, Butany J, Asa SL, et al. Pancreatic neuroendocrine tumor producing insulin and vasopressin. Endocr Pathol. 2017;28:1-6. doi: 10.1007/s12022-017-9492-5.

https://doi.org/10.1007/s12022-017-9492-5

Jensen RT. Endocrine tumors of the gastrointesti- nal tract and pancreas. In: Harrison ́s Principles of Internal Medicine. 19th edition. New York: The Mc- Graw-Hill Companies, Inc.; 2015.

Garbrecht N, Anlauf M, Schmitt A, Henopp T, Sipos B, Raffel A, et al. Somatostatin-producing neuroen- docrine tumors of the duodenum and pancreas: In- cidence, types, biological behavior, association with inherited syndromes, and functional activity. Endo- cr Relat Cancer. 2018;15:229-41. doi: 10.1677/ERC-07- 0157.

https://doi.org/10.1677/ERC-07-0157

Hulka B. Epidemiological studies using biological markers: Issues for epidemiologists. Cancer Epidemi- ol Biomarkers Prev. 1991;1:13-29.

Oberg K, Modlin IM, Herder W De, Pavel M, Klim- stra D, Frilling A, et al. Consensus on biomarkers for neuroendocrine tumour disease. Lancet Oncol. 2015;16:435-46. doi: 10.1016/S1470-2045(15)00186-2.

https://doi.org/10.1016/S1470-2045(15)00186-2

Jianu CS, Fossmark R, Syversen U, Hauso Ø, Waldum HL. A meal test improves the specificity of chromogr- anin A as a marker of neuroendocrine neoplasia. Tumor Biol. 2010;31:373-80. doi: 10.1007/s13277-010- 0045-5.

https://doi.org/10.1007/s13277-010-0045-5

Stridsberg M, Eriksson B, Öberg K, Janson ET. A com- parison between three commercial kits for chromogr- anin A measurements. J Endocrinol. 2003;177:337-41.

https://doi.org/10.1677/joe.0.1770337

Stridsberg M, Oberg K, Li Q. Measurements of chro- mogranin a, chromogranin b (secretogranin i), chro- mogranin c (secretogranin ii) and pancreastatin in plasma and urine from patients with carcinoid tu- mours and endocrine pancreatic tumours. J Endocri- nol. 1995;144:49-59. doi: 10.1677/joe.0.1440049.

https://doi.org/10.1677/joe.0.1440049

Ito T, Igarashi H, Jensen R. Serum pancreastatin: The long sought for universal, sensitive, specific tumor marker for neuroendocrine tumors (nets)? Pancreas. 2012;41:505-7. doi: 10.1097/MPA.0b013e318249a92a.

https://doi.org/10.1097/MPA.0b013e318249a92a

Bech PR, Martin NM, Ramachandran R, Bloom SR. The biochemical utility of chromogranin A , chro- mogranin B and cocaine- and amphetamine-regulat- ed transcript for neuroendocrine neoplasia. Ann Clin Biochem. 2013;51:8-21. doi: 10.1177/0004563213489670.

https://doi.org/10.1177/0004563213489670

Baudin E, Gigliottil A, Ducreux M, Ropers J, Comoy E, Sabourin JC, et al. Neuron-specific enolase and chro- mogranin A as markers of neuroendocrine tumours. Br J Cancer. 1998;78:1102-7. doi: 10.1038/bjc.1998.635.

https://doi.org/10.1038/bjc.1998.635

Langstein H, Norton J, Chiang V. The utility of circu- lating levels of human pancreatic polypeptide as a marker for islet cell tumors. Surgery. 1990;108:1109-15.

Walter T, Chardon L, CHopin-laly X, Raverot V, Caffin A, Chayvialle J, et al. Is the combination of chromogranin A and pancreatic polypeptide serum determinations of interest in the diagnosis and fol- low-up of gastro-entero-pancreatic neuroendocrine tumours ?. Eur J Cancer. 2012;48:1766-73. doi: 10.1016/j. ejca.2011.11.005.

https://doi.org/10.1016/j.ejca.2011.11.005

César S, Medrano-Guzmán R, López-García SC, Torres-Vargas S, González-Rodríguez D, Alvara- do-Cabrero I. Resecabilidad del tumor primario neuroendocrino gastroenteropancreático como fac- tor pronóstico de supervivencia. Cir Cir. 2011;79:498- 504.

Khan MS, Tsigani T, Rashid M, Rabouhans JS, Yu D, Luong TV, et al. Circulating tumor cells and EpCAM expression in neuroendocrine tumors. Clin Cancer Res. 2011;17:337-46. doi: 10.1158/1078-0432.

https://doi.org/10.1158/1078-0432.CCR-10-1776

Khan MS, Kirkwood A, Tsigani T, García-Hernández J, Hartley JA, Caplin ME, et al. Circulating tumor cells as prognostic markers in neuroendocrine tumors. J Clin Oncol. 2012;31:365-72.

https://doi.org/10.1200/JCO.2012.44.2905

Li A, Yu J, Kim H, Wolfgang CL, Canto MI, Hruban RH. MicroRNA array analysis finds elevated serum miR-1290 accurately distinguishes patients with low-stage pancreatic cancer from healthy and dis- ease controls. Clin Cancer Res. 2013;19:3600-11. doi: 10.1158/1078-0432.

https://doi.org/10.1158/1078-0432.CCR-12-3092

Thorns C, Schurmann C, Gebauer N, Wallaschofski H, Kümpers C, Bernard V, et al. Global microRNA pro- filing of pancreatic neuroendocrine neoplasias. Anti- cancer Res. 2014;34:2249-54.

Roldo C, Missiaglia E, Hagan JP, Falconi M, Capelli P, Bersani S, et al. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associ- ated with distinctive pathologic features and clinical behavior. J Clin Oncol. 2006;24:4677-84. doi: 10.1200/ JCO.2005.05.5194.

https://doi.org/10.1200/JCO.2005.05.5194

Modlin IM, Drozdov I, Kidd M. Gut neuroendocrine tumor blood qPCR fingerprint assay: Characteristics and reproducibility. Clin Chem Lab Med. 2014;52:419- 29. doi: 10.1515/cclm-2013-0496.

https://doi.org/10.1515/cclm-2013-0496

Modlin IM, Kidd M, Bodei L, Drozdov I, Aslanian H. The clinical utility of a novel blood-based multi-tran- scriptome assay for the diagnosis of neuroendocrine tumors of the gastrointestinal tract. Am J Gastroenter- ol. 2015;110:1223-32. doi: 10.1038/ajg.2015.160.

https://doi.org/10.1038/ajg.2015.160

Scarpa A, Chang D, Nones K. Whole-genome land- scape of pancreatic neuroendocrine tumours. Nature. 2017;543:65-71. doi: 10.1038/nature21063.

https://doi.org/10.1038/nature21063

Bodel L, Kidd M, Singh A, Van der Zwan W, Severi S. PRRT genomic signature in blood for prediction of 177 Lu-octreotate efficacy. Eur J Nucl Med Mol Imag- ing. 2018;15:1-15. doi: 10.1007/s00259-018-3967-6.

https://doi.org/10.1007/s00259-018-3967-6

Vargas C, Castaño R. Tumores neuroendocrinos gas- troenteropancreáticos. Rev Colomb Gastroenterol. 2010;25:165-76.

Takumi K, Fukukura Y, Higashi M. Pancreatic neu- roendocrine tumors: Correlation between the con- trast-enhanced computed tomography features and the pathological tumor grade. Eur J Radiol. 2015;84:1436-43. doi: 10.1016/j.ejrad.2015.05.005.

https://doi.org/10.1016/j.ejrad.2015.05.005

Humphrey PE, Alessandrino F, Bellizzi AM, Mor- tele KJ. Non-hyperfunctioning pancreatic endo- crine tumors: Multimodality imaging features with histopathological correlation. Abdom Imaging. 2015;40:2398-410. doi: 10.1007/s00261-015-0458-0.

https://doi.org/10.1007/s00261-015-0458-0

Antonio J, Pereira S, Rosado E, Bali M, Metens T, Chao S. Pancreatic neuroendocrine tumors: Correlation between histogram analysis of apparent diffusion coefficient maps and tumor grade. Abdom Imaging. 2015;40:3122-8. doi: 10.1007/s00261-015-0524-7.

https://doi.org/10.1007/s00261-015-0524-7

Reubi JC, Waser B. Concomitant expression of several peptide receptors in neuroendocrine tumours: Mo- lecular basis for in vivo multireceptor tumour target- ing. Eur J Nucl Med Mol Imaging. 2003;30:781-93. doi: 10.1007/s00259-003-1184-3.

https://doi.org/10.1007/s00259-003-1184-3

Gouda M. Islamic constitutionalism and rule of law: A constitutional economics perspective. Const Polit Econ. 2013;24:57-85. doi: 10.1007/s10602-012-9132-5.

https://doi.org/10.1007/s10602-012-9132-5

Ahlstrom H, Eriksson B, Bergstrom M. Pancreatic neuroendocrine tumors: Diagnosis with PET. Radiol- ogy. 1995;195:333-47. doi: 10.1148/radiology.195.2.7724749.

https://doi.org/10.1148/radiology.195.2.7724749

Koopmans KP, Neels ON, Kema IP, Elsinga PH, Links TP, Vries EGE De, et al. Molecular imaging in neuroendocrine tumors: Molecular uptake mecha- nisms and clinical results. Crit Rev Oncol Hematol. 2009;71:199-213. doi: 10.1016/j.critrevonc.2009.02.009.

https://doi.org/10.1016/j.critrevonc.2009.02.009

Herder W, Lamberts S. Somatostatin and soma- tostatin analogues: Diagnostic and therapeutic uses. Curr Opin Oncol. 2002;12:53-7. doi: 10.1097/00001622- 200201000-00010.

https://doi.org/10.1097/00001622-200201000-00010

Srirajaskanthan R, Kayani I, Quigley AM, Soh J, Caplin ME. The role of 68 Ga-DOTATATE PET in patients with neuroendocrine tumors and nega- tive or equivocal findings on 111 In-DTPA-octreotide scintigraphy. J Nucl Med. 2015;51:875-82. doi: 10.2967/ jnumed.109.066134.

https://doi.org/10.2967/jnumed.109.066134

Toumpanakis C, Kim M, Rinke A. Combination of cross-sectional and molecular imaging studies in the localization of gastroenteropancreatic neuroendo- crine tumors. Neuroendocrinology. 2014;99:63-74. doi: 10.1159/000358727.

https://doi.org/10.1159/000358727

Poeppel TD, Binse I, Petersenn S, Lahner H, Schott M, Antoch G, et al. 68Ga-DOTATOC versus 68Ga-DO- TATATE PET/CT in functional imaging of neuro- endocrine tumors. J Nucl Med. 2011;52:1864-71. doi: 10.2967/jnumed.111.091165.

https://doi.org/10.2967/jnumed.111.091165

Wild D, Bomanji JB, Benkert P, Maecke H, Ell PJ, Reu- bi JC, et al. Comparison of 68 Ga-DOTANOC and 68 Ga-DOTATATE PET/CT within patients with gas- troenteropancreatic neuroendocrine tumors. J Nucl Med. 2013;54:364-72. doi: 10.2967/jnumed.112.111724.

https://doi.org/10.2967/jnumed.112.111724

Kabasakal L, Demirci E, Ocak M. Comparison of 68 Ga-DOTATATE and 68 Ga-DOTANOC PET/CT im- aging in the same patient group with neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2012;39:1271-7. doi: 10.1007/s00259-012-2123-y.

https://doi.org/10.1007/s00259-012-2123-y

Simsek DH, Kuyumcu S, Turkmen C, Sanl Y, Aykan F, Unal S, et al. Can complementary 68Ga-DO- TATATE and 18F-FDG PET/CT establish the miss- ing link between histopathology and therapeutic approach in gastroenteropancreatic neuroendocrine tumors?. J Nucl Med. 2015;55:1811-8. doi: 10.2967/ jnumed.114.142224.

https://doi.org/10.2967/jnumed.114.142224

Garin E, Jeune F Le, Devillers A, Cuggia M, Bouri- el C, Boucher E, et al. Predictive value of 18 F-FDG PET and somatostatin receptor scintigraphy in pa- tients with metastatic endocrine tumors. J Nucl Med. 2009;50:858-65. doi: 10.2967/jnumed.108.057505.

https://doi.org/10.2967/jnumed.108.057505

Binderup T, Knigge U, Loft A, Federspiel B, Kjaer A. 18F-fluorodeoxyglucose positron emission tomog- raphy predicts survival of patients with neuroendo- crine tumors. Clin Cancer Res. 2010;16:978-86. doi: 10.1158/1078-0432.

https://doi.org/10.1158/1078-0432.CCR-09-1759

Koopmans KP, Neels OC, Kema IP, Elsinga PH, Sluiter WJ, Vanghillewe K, et al. Improved Staging of Patients With Carcinoid and Islet Cell Tumors With 18F-Di- hydroxy-Phenyl-Alnine and 11C-5-Hydroxy-Trypto- phan Positron Emission Tomography. J Clin Oncol. 2015;26:1489-95. doi: 10.1200/JCO.2007.15.1126.

https://doi.org/10.1200/JCO.2007.15.1126

Ambrosini V, Tomassetti P, Castellucci P, Campana D, Montini G, Rubello D, et al. Comparison between 68 Ga-DOTA-NOC and 18 F-DOPA PET for the de- tection of gastro-entero-pancreatic and lung neu- ro-endocrine tumours. Eur J Nucl Med Mol Imaging. 2008;35:1431-8. doi: 10.1007/s00259-008-0769-2.

https://doi.org/10.1007/s00259-008-0769-2

Haug A, Auernhammer CJ, Wängler B, Tiling R, Schmidt G. Intraindividual comparison of 68 Ga- DOTA-TATE and 18 F-DOPA PET in patients with well-differentiated metastatic neuroendocrine tu- mours. Eur J Nucl Med Mol Imaging. 2009;36:765-70. doi: 10.1007/s00259-008-1030-8.

https://doi.org/10.1007/s00259-008-1030-8

Christ E, Wild D, Forrer F. Glucagon-like peptide-1 re- ceptor imaging for localization of insulinomas. J Clin Endocrinol Metab. 2009;94:4398-405. doi: 10.1210/ jc.2009-1082.

https://doi.org/10.1210/jc.2009-1082

Sowa-Staszczak A, Pach D, Miko R, Mäcke H. Glu- cagon-like peptide-1 receptor imaging with [Ly- s40(Ahx-HYNIC-99mTc/EDDA)NH2]-exendin-4 for the detection of insulinoma. Eur J Nucl Med Mol Im- aging. 2013;40:524-31. doi: 10.1007/s00259-012-2299-1.

https://doi.org/10.1007/s00259-012-2299-1

Wenning AS, Kirchner P, Antwi K, Fani M, Wild D, Christ E. Preoperative glucagon-like peptide-1 receptor imaging reduces surgical trauma and pancreatic tissue loss in insulinoma patients: A report of three cases. Pa- tient Saf Surg. 2015;9:23. doi: 10.1186/s13037-015-0064-7.

https://doi.org/10.1186/s13037-015-0064-7

Antwi K, Fani M, Nicolas G, Rottenburger C, Heye T. Localization of hidden insulinomas with Ga-DO- TA-exendin-4 PET/CT: A pilot study. J Nucl Med. 2015;56:1075-8. doi: 10.2967/jnumed.115.157768.

https://doi.org/10.2967/jnumed.115.157768

Wild D, Fani M, Behe M, Brink I, Rivier JEF, Reubi JC, et al. First clinical evidence that imaging with soma- tostatin receptor antagonists is feasible. J Nucl Med. 2014;52:1412-8. doi: 10.2967/jnumed.111.088922.

https://doi.org/10.2967/jnumed.111.088922

Cescato R, Waser B, Fani M, Reubi JC. Evaluation of 177 Lu-DOTA-sst 2 antagonist versus 177 Lu-DOTA- sst agonist binding in human cancers in vitro. J Nucl Med. 2011;52:1886-91. doi: 10.2967/jnumed.111.095778.

https://doi.org/10.2967/jnumed.111.095778

Cescato R, Erchegyi J, Waser B, Maecke HR, Rivier JE, Reubi JC, et al. Design and in vitro characterization of highly sst 2-selective somatostatin antagonists suit- able for radiotargeting. J Med Chem. 2008;51:4030-7. doi: 10.1021/jm701618q.

https://doi.org/10.1021/jm701618q

Wild D, Fani M, Fischer R, Pozzo L Del, Kaul F, Krebs S, et al. Comparison of somatostatin receptor agonist and antagonist for peptide receptor radionuclide therapy: A pilot study. J Nucl Med. 2014;55:1-5. doi: 10.2967/jnumed.114.138834.

https://doi.org/10.2967/jnumed.114.138834

Puli SR, Kalva N, Bechtold ML, Pamulaparthy SR, Cashman MD, Estes NC, et al. Diagnostic accuracy of endoscopic ultrasound in pancreatic neuroendo- crine tumors: A systematic review and meta analysis. World J Gastroenterol. 2013;19:3678-84. doi: 10.3748/ wjg.v19.i23.3678.

https://doi.org/10.3748/wjg.v19.i23.3678

Wiesli P, Brändle M, Schmid C, Krähenbühl L, Furrer J. Selective arterial calcium stimulation and hepatic venous sampling in the evaluation of hyperinsu- linemic hypoglycemia: Potential and limitations. J Vasc Interv Radiol. 2004;15:1251-6. doi: 10.1097/01. RVI.0000140638.55375.1E.

https://doi.org/10.1097/01

Francis JM, Kiezun A, Ramos AH, Serra S, Pedamallu CS, Qian ZR, et al. Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nat Genet. 2013;45:1483-7. doi: 10.1038/ng.2821.

https://doi.org/10.1038/ng.2821

Georgitsi M, Raitila A, Karhu A, Luijt RB, Aalfs C, Sane T, et al. Germline CDKN1B/p27Kip1 mutation in multiple endocrine neoplasia. J Clin Endocrinol Me- tab. 2015;92:3321-5. doi: 10.1210/jc.2006-2843.

https://doi.org/10.1210/jc.2006-2843

Lubensky IA, Pack S, Ault D, Vortmeyer AO, Libutti SK, Choyke PL, et al. Multiple neuroendocrine tu- mors of the pancreas in von Hippel-Lindau disease patients histopathological and molecular genetic analysis. Am J Pathol. 1998;153:223-31. doi: 10.1016/ S0002-9440(10)65563-0.

https://doi.org/10.1016/S0002-9440(10)65563-0

Vogt S, Jones N, Christian D, Engel C, Nielsen M, Kaufmann A, et al. Expanded extracolonic tumor spectrum in MUTYH-associated polyposis. Gas- troenterology. 2009;137:1976-85. doi: 10.1053/j.gas- tro.2009.08.052.

https://doi.org/10.1053/j.gastro.2009.08.052

Dong X, Wang L, Taniguchi K, Wang X, Cunningham JM, Mcdonnell SK, et al. Mutations in CHEK2 asso- ciated with prostate cancer risk. Am J Hum Genet. 2003;72:270-80. doi: 10.1086/346094.

https://doi.org/10.1086/346094

Uccella S, Blank A, Maragliano R, Sessa F, Perren A, La Rosa S. Calcitonin-producing neuroendocrine neoplasms of the pancreas: Clinicopathological study of 25 cases and review of the literature. Endocr Pathol. 2017;28:1-11. doi: 10.1007/s12022-017-9505-4.

https://doi.org/10.1007/s12022-017-9505-4

Eriksson O, Velikyan I, Selvaraju RK, Kandeel F, Jo- hansson L, Antoni G, et al. Detection of metastatic insulinoma by positron emission tomography with [68Ga]exendin-4-A case report. J Clin Endocrinol Metab. 2014;99:1519-24. doi: 10.1210/jc.2013-3541.

https://doi.org/10.1210/jc.2013-3541

Pfeifer A, Knigge U, Binderup T, Mortensen J, Oturai P, Loft A, et al. 64Cu - DOTATATE PET for neuroen- docrine tumors: A prospective head to head compari- son with 111 In-DTPA-octreotide in 112 patients. J Nucl Med. 2015;56:847-54. doi: 10.2967/jnumed.115.156539.

https://doi.org/10.2967/jnumed.115.156539

Descargas

Publicado

2018-01-01

Cómo citar

(1)
Guzmán, Y. F.; López, R. del P.; Vera, A.; González Devia D. Herramientas Para El Abordaje diagnóstico De Los Tumores Neuroendocrinos De páncreas. Rev Colomb Cir 2018, 33, 79-99.

Número

Sección

Artículo de Revisión

Métricas

QR Code

Algunos artículos similares: