Skip to main content
Erschienen in: Journal of Hepato-Biliary-Pancreatic Sciences 6/2013

01.08.2013 | Original Article

Serine protease inhibitor Kazal type 1 and epidermal growth factor receptor are expressed in pancreatic tubular adenocarcinoma, intraductal papillary mucinous neoplasm, and pancreatic intraepithelial neoplasia

verfasst von: Nobuyuki Ozaki, Masaki Ohmuraya, Satoshi Ida, Daisuke Hashimoto, Yoshiaki Ikuta, Akira Chikamoto, Masahiko Hirota, Hideo Baba

Erschienen in: Journal of Hepato-Biliary-Pancreatic Sciences | Ausgabe 6/2013

Einloggen, um Zugang zu erhalten

Abstract

Background

Serine protease inhibitor Kazal type 1 (SPINK1) is expressed in normal human pancreatic acinar cells and in a variety of tumors, and binds to the epidermal growth factor receptor (EGFR), mediating cell proliferation through the mitogen-activated protein kinase cascade in pancreatic cancer cell lines. Here, we aimed to assess SPINK1 and EGFR expression in various neoplastic lesions, including tissues demonstrating precancerous changes.

Methods

Surgical specimens of pancreatic ductal adenocarcinoma (n = 23), intraductal papillary mucinous neoplasm (IPMN; n = 21), pancreatic neoplasms other than ductal adenocarcinoma (n = 8), chronic pancreatitis (n = 11), and pancreatic intraepithelial neoplasia (PanIN) lesions within the resected specimens were analyzed immunohistochemically for SPINK1 and EGFR expression.

Results

Sixty-five PanIN-1A, 32 PanIN-1B, 17 PanIN-2, and 6 PanIN-3 were identified. Both SPINK1 and EGFR were expressed in almost all PanIN lesions. All tubular ductal adenocarcinoma, IPMN, and mucinous cystadenocarcinoma samples (neoplasms of ductal origin) expressed SPINK1, whereas acinar cell carcinoma, anaplastic carcinoma, adenosquamous carcinoma, insulinoma, and islet cell carcinoma did not. EGFR was expressed in 87 % of tubular adenocarcinoma and 48 % of IPMN lesions. Among IPMN lesions, malignant lesions (IPMC) expressed EGFR more often than benign lesions (IPMA) did. Scattered expression of EGFR was observed in normal pancreatic ducts and within the tubular complex within chronic pancreatitis lesions.

Conclusions

These results indicate that SPINK1 plays a role as a growth factor, signaling through the EGFR pathway in pancreatic ductal adenocarcinoma and neoplasms, and that the EGFR is involved in the malignant transformation of IPMN.
Literatur
1.
Zurück zum Zitat Ohmuraya M, Ozaki N, Hirota M, Baba H, Yamamura K. Serine protease inhibitor Kazal type 1 (SPINK1): beyond the trypsin inhibitor. Curr Enzyme Inhib. 2009;5:110–6.CrossRef Ohmuraya M, Ozaki N, Hirota M, Baba H, Yamamura K. Serine protease inhibitor Kazal type 1 (SPINK1): beyond the trypsin inhibitor. Curr Enzyme Inhib. 2009;5:110–6.CrossRef
2.
Zurück zum Zitat Kazal LA, Spicer DS, Brahinsky RA. Isolation of a crystalline trypsin inhibitor-anticoagulant protein from the pancreas. J Am Chem Soc. 1948;70:3034–40.PubMedCrossRef Kazal LA, Spicer DS, Brahinsky RA. Isolation of a crystalline trypsin inhibitor-anticoagulant protein from the pancreas. J Am Chem Soc. 1948;70:3034–40.PubMedCrossRef
3.
Zurück zum Zitat Horii A, Kobayashi T, Tomita N, Yamamoto T, Fukushige S, Murotsu T, et al. Primary structure of human pancreatic secretory trypsin inhibitor (PSTI) gene. Biochem Biophys Res Commun. 1987;149(2):635–41. pii: 0006-291X(87)90415-3. Horii A, Kobayashi T, Tomita N, Yamamoto T, Fukushige S, Murotsu T, et al. Primary structure of human pancreatic secretory trypsin inhibitor (PSTI) gene. Biochem Biophys Res Commun. 1987;149(2):635–41. pii: 0006-291X(87)90415-3.
4.
Zurück zum Zitat Bartelt DC, Shapanka R, Greene LJ. The primary structure of the human pancreatic secretory trypsin inhibitor. Amino acid sequence of the reduced S-aminoethylated protein. Arch Biochem Biophys. 1977;179(1):189–99.PubMedCrossRef Bartelt DC, Shapanka R, Greene LJ. The primary structure of the human pancreatic secretory trypsin inhibitor. Amino acid sequence of the reduced S-aminoethylated protein. Arch Biochem Biophys. 1977;179(1):189–99.PubMedCrossRef
6.
Zurück zum Zitat Ohmuraya M, Hirota M, Araki M, Mizushima N, Matsui M, Mizumoto T, et al. Autophagic cell death of pancreatic acinar cells in serine protease inhibitor Kazal type 3-deficient mice. Gastroenterology. 2005;129(2):696–705. doi:10.1016/j.gastro.2005.05.057.PubMed Ohmuraya M, Hirota M, Araki M, Mizushima N, Matsui M, Mizumoto T, et al. Autophagic cell death of pancreatic acinar cells in serine protease inhibitor Kazal type 3-deficient mice. Gastroenterology. 2005;129(2):696–705. doi:10.​1016/​j.​gastro.​2005.​05.​057.PubMed
8.
Zurück zum Zitat Fukayama M, Hayashi Y, Koike M, Ogawa M, Kosaki G. Immunohistochemical localization of pancreatic secretory trypsin inhibitor in fetal and adult pancreatic and extrapancreatic tissues. J Histochem Cytochem. 1986;34(2):227–35.PubMedCrossRef Fukayama M, Hayashi Y, Koike M, Ogawa M, Kosaki G. Immunohistochemical localization of pancreatic secretory trypsin inhibitor in fetal and adult pancreatic and extrapancreatic tissues. J Histochem Cytochem. 1986;34(2):227–35.PubMedCrossRef
9.
Zurück zum Zitat Matsuda K, Ogawa M, Murata A, Kitahara T, Kosaki G. Elevation of serum immunoreactive pancreatic secretory trypsin inhibitor contents in various malignant diseases. Res Commun Chem Pathol Pharmacol. 1983;40(2):301–5.PubMed Matsuda K, Ogawa M, Murata A, Kitahara T, Kosaki G. Elevation of serum immunoreactive pancreatic secretory trypsin inhibitor contents in various malignant diseases. Res Commun Chem Pathol Pharmacol. 1983;40(2):301–5.PubMed
10.
Zurück zum Zitat Shibata T, Ogawa M, Takata N, Matsuda K, Niinobu T, Uda K, et al. Distribution of pancreatic secretory trypsin inhibitor in various human tissues and its inactivation in the gastric mucosa. Res Commun Chem Pathol Pharmacol. 1987;55(2):243–8.PubMed Shibata T, Ogawa M, Takata N, Matsuda K, Niinobu T, Uda K, et al. Distribution of pancreatic secretory trypsin inhibitor in various human tissues and its inactivation in the gastric mucosa. Res Commun Chem Pathol Pharmacol. 1987;55(2):243–8.PubMed
11.
Zurück zum Zitat Kitahara T, Takatsuka Y, Fujimoto KI, Tanaka S, Ogawa M, Kosaki G. Radioimmunoassay for human pancreatic secretory trypsin inhibitor: measurement of serum pancreatic secretory trypsin inhibitor in normal subjects and subjects with pancreatic diseases. Clin Chim Acta. 1980;103(2):135–43.PubMedCrossRef Kitahara T, Takatsuka Y, Fujimoto KI, Tanaka S, Ogawa M, Kosaki G. Radioimmunoassay for human pancreatic secretory trypsin inhibitor: measurement of serum pancreatic secretory trypsin inhibitor in normal subjects and subjects with pancreatic diseases. Clin Chim Acta. 1980;103(2):135–43.PubMedCrossRef
12.
Zurück zum Zitat Matsuda K, Ogawa M, Shibata T, Nishibe S, Miyauchi K, Matsuda Y, et al. Postoperative elevation of serum pancreatic secretory trypsin inhibitor. Am J Gastroenterol. 1985;80(9):694–8.PubMed Matsuda K, Ogawa M, Shibata T, Nishibe S, Miyauchi K, Matsuda Y, et al. Postoperative elevation of serum pancreatic secretory trypsin inhibitor. Am J Gastroenterol. 1985;80(9):694–8.PubMed
13.
Zurück zum Zitat Wang J, Ohmuraya M, Hirota M, Baba H, Zhao G, Takeya M, et al. Expression pattern of serine protease inhibitor kazal type 3 (Spink3) during mouse embryonic development. Histochem Cell Biol. 2008. doi:10.1007/s00418-008-0425-8. Wang J, Ohmuraya M, Hirota M, Baba H, Zhao G, Takeya M, et al. Expression pattern of serine protease inhibitor kazal type 3 (Spink3) during mouse embryonic development. Histochem Cell Biol. 2008. doi:10.​1007/​s00418-008-0425-8.
14.
Zurück zum Zitat Huhtala ML, Kahanpaa K, Seppala M, Halila H, Stenman UH. Excretion of a tumor-associated trypsin inhibitor (TATI) in urine of patients with gynecological malignancy. Int J Cancer. 1983;31(6):711–4.PubMedCrossRef Huhtala ML, Kahanpaa K, Seppala M, Halila H, Stenman UH. Excretion of a tumor-associated trypsin inhibitor (TATI) in urine of patients with gynecological malignancy. Int J Cancer. 1983;31(6):711–4.PubMedCrossRef
15.
Zurück zum Zitat Murata A, Ogawa M, Uda K, Matsuura N, Watanabe Y, Baba T, et al. Release of pancreatic secretory trypsin inhibitor from human hepatoblastoma cells on stimulation with cytokines. Life Sci. 1988;43(15):1233–40.PubMedCrossRef Murata A, Ogawa M, Uda K, Matsuura N, Watanabe Y, Baba T, et al. Release of pancreatic secretory trypsin inhibitor from human hepatoblastoma cells on stimulation with cytokines. Life Sci. 1988;43(15):1233–40.PubMedCrossRef
16.
Zurück zum Zitat Ogata N. Demonstration of pancreatic secretory trypsin inhibitor in serum-free culture medium conditioned by the human pancreatic carcinoma cell line CAPAN-1. J Biol Chem. 1988;263(26):13427–31.PubMed Ogata N. Demonstration of pancreatic secretory trypsin inhibitor in serum-free culture medium conditioned by the human pancreatic carcinoma cell line CAPAN-1. J Biol Chem. 1988;263(26):13427–31.PubMed
17.
Zurück zum Zitat Ogawa M, Matsuura N, Higashiyama K, Mori T. Expression of pancreatic secretory trypsin inhibitor in various cancer cells. Res Commun Chem Pathol Pharmacol. 1987;55(1):137–40.PubMed Ogawa M, Matsuura N, Higashiyama K, Mori T. Expression of pancreatic secretory trypsin inhibitor in various cancer cells. Res Commun Chem Pathol Pharmacol. 1987;55(1):137–40.PubMed
18.
Zurück zum Zitat Stenman UH, Koivunen E, Itkonen O. Biology and function of tumor-associated trypsin inhibitor, TATI. Scand J Clin Lab Investig Suppl. 1991;207:5–9.CrossRef Stenman UH, Koivunen E, Itkonen O. Biology and function of tumor-associated trypsin inhibitor, TATI. Scand J Clin Lab Investig Suppl. 1991;207:5–9.CrossRef
19.
Zurück zum Zitat Stenman UH, Huhtala ML, Koistinen R, Seppala M. Immunochemical demonstration of an ovarian cancer-associated urinary peptide. Int J Cancer. 1982;30(1):53–7.PubMedCrossRef Stenman UH, Huhtala ML, Koistinen R, Seppala M. Immunochemical demonstration of an ovarian cancer-associated urinary peptide. Int J Cancer. 1982;30(1):53–7.PubMedCrossRef
20.
Zurück zum Zitat Huhtala ML, Pesonen K, Kalkkinen N, Stenman UH. Purification and characterization of a tumor-associated trypsin inhibitor from the urine of a patient with ovarian cancer. J Biol Chem. 1982;257(22):13713–6.PubMed Huhtala ML, Pesonen K, Kalkkinen N, Stenman UH. Purification and characterization of a tumor-associated trypsin inhibitor from the urine of a patient with ovarian cancer. J Biol Chem. 1982;257(22):13713–6.PubMed
21.
Zurück zum Zitat Lee YC, Pan HW, Peng SY, Lai PL, Kuo WS, Ou YH, et al. Overexpression of tumour-associated trypsin inhibitor (TATI) enhances tumour growth and is associated with portal vein invasion, early recurrence and a stage-independent prognostic factor of hepatocellular carcinoma. Eur J Cancer. 2007;43(4):736–44. doi:10.1016/j.ejca.2006.11.020.PubMedCrossRef Lee YC, Pan HW, Peng SY, Lai PL, Kuo WS, Ou YH, et al. Overexpression of tumour-associated trypsin inhibitor (TATI) enhances tumour growth and is associated with portal vein invasion, early recurrence and a stage-independent prognostic factor of hepatocellular carcinoma. Eur J Cancer. 2007;43(4):736–44. doi:10.​1016/​j.​ejca.​2006.​11.​020.PubMedCrossRef
22.
Zurück zum Zitat Stenman UH. Tumor-associated trypsin inhibitor. Clin Chem. 2002;48(8):1206–9.PubMed Stenman UH. Tumor-associated trypsin inhibitor. Clin Chem. 2002;48(8):1206–9.PubMed
24.
Zurück zum Zitat Tonouchi A, Ohtsuka M, Ito H, Kimura F, Shimizu H, Kato M, et al. Relationship between pancreatic secretory trypsin inhibitor and early recurrence of intrahepatic cholangiocarcinoma following surgical resection. Am J Gastroenterol. 2006;101(7):1601–10. doi:10.1111/j.1572-0241.2006.00612.x.PubMedCrossRef Tonouchi A, Ohtsuka M, Ito H, Kimura F, Shimizu H, Kato M, et al. Relationship between pancreatic secretory trypsin inhibitor and early recurrence of intrahepatic cholangiocarcinoma following surgical resection. Am J Gastroenterol. 2006;101(7):1601–10. doi:10.​1111/​j.​1572-0241.​2006.​00612.​x.PubMedCrossRef
25.
Zurück zum Zitat Hunt LT, Barker WC, Dayhoff MO. Epidermal growth factor: internal duplication and probable relationship to pancreatic secretory trypsin inhibitor. Biochem Biophys Res Commun. 1974;60(3):1020–8. pii: 0006-291X(74)90415-X. Hunt LT, Barker WC, Dayhoff MO. Epidermal growth factor: internal duplication and probable relationship to pancreatic secretory trypsin inhibitor. Biochem Biophys Res Commun. 1974;60(3):1020–8. pii: 0006-291X(74)90415-X.
26.
Zurück zum Zitat Scheving LA. Primary amino acid sequence similarity between human epidermal growth factor-urogastrone, human pancreatic secretory trypsin inhibitor, and members of porcine secretin family. Arch Biochem Biophys. 1983;226(2):411–3. pii: 0003-9861(83)90309-0. Scheving LA. Primary amino acid sequence similarity between human epidermal growth factor-urogastrone, human pancreatic secretory trypsin inhibitor, and members of porcine secretin family. Arch Biochem Biophys. 1983;226(2):411–3. pii: 0003-9861(83)90309-0.
27.
Zurück zum Zitat Ozaki N, Ohmuraya M, Hirota M, Ida S, Wang J, Takamori H, et al. Serine protease inhibitor Kazal type 1 promotes proliferation of pancreatic cancer cells through the epidermal growth factor receptor. Mol Cancer Res MCR. 2009;7(9):1572–81. doi:10.1158/1541-7786.MCR-08-0567.CrossRef Ozaki N, Ohmuraya M, Hirota M, Ida S, Wang J, Takamori H, et al. Serine protease inhibitor Kazal type 1 promotes proliferation of pancreatic cancer cells through the epidermal growth factor receptor. Mol Cancer Res MCR. 2009;7(9):1572–81. doi:10.​1158/​1541-7786.​MCR-08-0567.CrossRef
28.
Zurück zum Zitat Hruban RH, Takaori K, Klimstra DS, Adsay NV, Albores-Saavedra J, Biankin AV, et al. An illustrated consensus on the classification of pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasms. Am J Surg Pathol. 2004;28(8):977–87. pii: 00000478-200408000-00001. Hruban RH, Takaori K, Klimstra DS, Adsay NV, Albores-Saavedra J, Biankin AV, et al. An illustrated consensus on the classification of pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasms. Am J Surg Pathol. 2004;28(8):977–87. pii: 00000478-200408000-00001.
29.
Zurück zum Zitat Oikawa T, Hitomi J, Kono A, Kaneko E, Yamaguchi K. Frequent expression of genes for receptor tyrosine kinases and their ligands in human pancreatic cancer cells. Int J Pancreatol. 1995;18(1):15–23.PubMed Oikawa T, Hitomi J, Kono A, Kaneko E, Yamaguchi K. Frequent expression of genes for receptor tyrosine kinases and their ligands in human pancreatic cancer cells. Int J Pancreatol. 1995;18(1):15–23.PubMed
30.
Zurück zum Zitat Korc M, Friess H, Yamanaka Y, Kobrin MS, Buchler M, Beger HG. Chronic pancreatitis is associated with increased concentrations of epidermal growth factor receptor, transforming growth factor alpha, and phospholipase C gamma. Gut. 1994;35(10):1468–73.PubMedCrossRef Korc M, Friess H, Yamanaka Y, Kobrin MS, Buchler M, Beger HG. Chronic pancreatitis is associated with increased concentrations of epidermal growth factor receptor, transforming growth factor alpha, and phospholipase C gamma. Gut. 1994;35(10):1468–73.PubMedCrossRef
32.
Zurück zum Zitat Friess H, Berberat P, Schilling M, Kunz J, Korc M, Buchler MW. Pancreatic cancer: the potential clinical relevance of alterations in growth factors and their receptors. J Mol Med. 1996;74(1):35–42.PubMedCrossRef Friess H, Berberat P, Schilling M, Kunz J, Korc M, Buchler MW. Pancreatic cancer: the potential clinical relevance of alterations in growth factors and their receptors. J Mol Med. 1996;74(1):35–42.PubMedCrossRef
33.
Zurück zum Zitat Korc M, Chandrasekar B, Yamanaka Y, Friess H, Buchier M, Beger HG. Overexpression of the epidermal growth factor receptor in human pancreatic cancer is associated with concomitant increases in the levels of epidermal growth factor and transforming growth factor alpha. J Clin Investig. 1992;90(4):1352–60. doi:10.1172/JCI116001.PubMedCrossRef Korc M, Chandrasekar B, Yamanaka Y, Friess H, Buchier M, Beger HG. Overexpression of the epidermal growth factor receptor in human pancreatic cancer is associated with concomitant increases in the levels of epidermal growth factor and transforming growth factor alpha. J Clin Investig. 1992;90(4):1352–60. doi:10.​1172/​JCI116001.PubMedCrossRef
36.
Zurück zum Zitat Sibilia M, Fleischmann A, Behrens A, Stingl L, Carroll J, Watt FM, et al. The EGF receptor provides an essential survival signal for SOS-dependent skin tumor development. Cell 2000;102(2):211–20. pii: S0092-8674(00)00026-X. Sibilia M, Fleischmann A, Behrens A, Stingl L, Carroll J, Watt FM, et al. The EGF receptor provides an essential survival signal for SOS-dependent skin tumor development. Cell 2000;102(2):211–20. pii: S0092-8674(00)00026-X.
37.
Zurück zum Zitat Day JD, Digiuseppe JA, Yeo C, Lai-Goldman M, Anderson SM, Goodman SN, et al. Immunohistochemical evaluation of HER-2/neu expression in pancreatic adenocarcinoma and pancreatic intraepithelial neoplasms. Hum Pathol. 1996;27(2):119–24.PubMedCrossRef Day JD, Digiuseppe JA, Yeo C, Lai-Goldman M, Anderson SM, Goodman SN, et al. Immunohistochemical evaluation of HER-2/neu expression in pancreatic adenocarcinoma and pancreatic intraepithelial neoplasms. Hum Pathol. 1996;27(2):119–24.PubMedCrossRef
38.
Zurück zum Zitat Marchbank T, Chinery R, Hanby AM, Poulsom R, Elia G, Playford RJ. Distribution and expression of pancreatic secretory trypsin inhibitor and its possible role in epithelial restitution. Am J Pathol. 1996;148(3):715–22.PubMed Marchbank T, Chinery R, Hanby AM, Poulsom R, Elia G, Playford RJ. Distribution and expression of pancreatic secretory trypsin inhibitor and its possible role in epithelial restitution. Am J Pathol. 1996;148(3):715–22.PubMed
40.
Zurück zum Zitat Bockman DE, Guo J, Buchler P, Muller MW, Bergmann F, Friess H. Origin and development of the precursor lesions in experimental pancreatic cancer in rats. Lab Investig. 2003;83(6):853–9.PubMed Bockman DE, Guo J, Buchler P, Muller MW, Bergmann F, Friess H. Origin and development of the precursor lesions in experimental pancreatic cancer in rats. Lab Investig. 2003;83(6):853–9.PubMed
41.
Zurück zum Zitat Jimenez RE, Z’Graggen K, Hartwig W, Graeme-Cook F, Warshaw AL, Fernandez-del Castillo C. Immunohistochemical characterization of pancreatic tumors induced by dimethylbenzanthracene in rats. Am J Pathol. 1999;154(4):1223–9.PubMedCrossRef Jimenez RE, Z’Graggen K, Hartwig W, Graeme-Cook F, Warshaw AL, Fernandez-del Castillo C. Immunohistochemical characterization of pancreatic tumors induced by dimethylbenzanthracene in rats. Am J Pathol. 1999;154(4):1223–9.PubMedCrossRef
43.
Zurück zum Zitat Siveke JT, Einwachter H, Sipos B, Lubeseder-Martellato C, Kloppel G, Schmid RM. Concomitant pancreatic activation of Kras(G12D) and Tgfa results in cystic papillary neoplasms reminiscent of human IPMN. Cancer Cell. 2007;12(3):266–79. doi:10.1016/j.ccr.2007.08.002.PubMedCrossRef Siveke JT, Einwachter H, Sipos B, Lubeseder-Martellato C, Kloppel G, Schmid RM. Concomitant pancreatic activation of Kras(G12D) and Tgfa results in cystic papillary neoplasms reminiscent of human IPMN. Cancer Cell. 2007;12(3):266–79. doi:10.​1016/​j.​ccr.​2007.​08.​002.PubMedCrossRef
44.
Zurück zum Zitat Yeh TS, Jan YY, Chiu CT, Ho YB, Chen TC, Lee KF, et al. Characterisation of oestrogen receptor, progesterone receptor, trefoil factor 1, and epidermal growth factor and its receptor in pancreatic cystic neoplasms and pancreatic ductal adenocarcinoma. Gut. 2002;51(5):712–6.PubMedCrossRef Yeh TS, Jan YY, Chiu CT, Ho YB, Chen TC, Lee KF, et al. Characterisation of oestrogen receptor, progesterone receptor, trefoil factor 1, and epidermal growth factor and its receptor in pancreatic cystic neoplasms and pancreatic ductal adenocarcinoma. Gut. 2002;51(5):712–6.PubMedCrossRef
Metadaten
Titel
Serine protease inhibitor Kazal type 1 and epidermal growth factor receptor are expressed in pancreatic tubular adenocarcinoma, intraductal papillary mucinous neoplasm, and pancreatic intraepithelial neoplasia
verfasst von
Nobuyuki Ozaki
Masaki Ohmuraya
Satoshi Ida
Daisuke Hashimoto
Yoshiaki Ikuta
Akira Chikamoto
Masahiko Hirota
Hideo Baba
Publikationsdatum
01.08.2013
Verlag
Springer Japan
Erschienen in
Journal of Hepato-Biliary-Pancreatic Sciences / Ausgabe 6/2013
Print ISSN: 1868-6974
Elektronische ISSN: 1868-6982
DOI
https://doi.org/10.1007/s00534-012-0587-6

Weitere Artikel der Ausgabe 6/2013

Journal of Hepato-Biliary-Pancreatic Sciences 6/2013 Zur Ausgabe

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.