TumorDiagnostik & Therapie 2011; 32(5): 269-276
DOI: 10.1055/s-0031-1281715
Originalarbeit

© Georg Thieme Verlag KG Stuttgart · New York

Zielgerichtete Therapie für Patienten mit Schilddrüsenkarzinom

Target Therapy for Thyroid CancerP. Schütt1 , S. Müller2 , A. Matuszczyk3 , K. W. Schmid4 , A. Bockisch2 , M. Schuler1 , K. Mann3
  • 1Innere Klinik (Tumorforschung), Universitätsklinikum Essen, Universität Duisburg-Essen
  • 2Klinik für Nuklearmedizin, Universitätsklinikum Essen, Universität Duisburg-Essen
  • 3Klinik für Endokrinologie und Zentrallabor Bereich Forschung und Lehre, Universitätsklinikum Essen, Universität Duisburg-Essen
  • 4Institut für Pathologie und Neuropathologie; Westdeutsches Tumorzentrum, Universitätsklinikum Essen, Universität Duisburg-Essen
Further Information

Publication History

Publication Date:
13 October 2011 (online)

Zusammenfassung

Bislang waren die medikamentösen therapeutischen Optionen für Patienten mit metastasierten differenzierten (Radiojod-refraktären) oder medullären Schilddrüsenkarzinomen begrenzt. In den letzten Jahren wurden große Fortschritte gemacht, die genetischen Aberrationen, die für die Pathogenese dieser Tumoren ursächlich sind, zu erkennen und funktionell zu verstehen. Basierend auf diesen Erkenntnissen wurden neuere zielgerichtete Krebsmedikamente, sogenannte Kinase-Inhibitoren, für Patienten mit Schilddrüsenkarzinomen im Rahmen klinischer Studien evaluiert. Vor diesem Hintergrund wurde eine selektive Literaturrecherche der MEDLINE- und ASCO-Datenbanken durchgeführt und relevante Studien zu neueren medikamentösen Therapieansätzen bei Schilddrüsenkarzinomen in diesem Übersichtsartikel zusammengefasst. Insbesondere Inhibitoren, die die Signaltransduktion der VEGF-Rezeptorfamilie hemmen, zeigten in Phase-II-Studien bei Patienten mit differenzierten und medullären Schilddrüsenkarzinomen eine eindrucksvolle Wirksamkeit. In diesen Studien wurden objektive Remissionsraten von bis zu 35 % und Krankheitsstabilisierungsraten von weiteren 50 % beobachtet. Das mediane progressionsfreie Überleben lag zwischen 7 und 28 Monaten. Die Ergebnisse dieser Studien belegen, dass diese Kinase-Inhibitoren in der Behandlung von Schilddrüsenkarzinomen eine beachtliche Wirksamkeit aufweisen können. Dennoch sollte der klinische Nutzen dieser teilweise toxischen und kostenintensiven Therapien in Phase-III-Studien überprüft werden, bevor generelle Therapieempfehlungen ausgesprochen werden können.

Abstract

Therapeutic options for patients suffering from metastatic and radioiodine-refractory differentiated and/or medullary thyroid cancer are limited. The clinical benefit of classical chemotherapeutic agents is marginal. Recently, advantages have been made in understanding the molecular pathways that are involved in the pathogenesis of thyroid cancer. Against this background novel agents targeting oncogenic kinases involved in the pathogenesis of thyroid cancer have entered clinical trials. A selective search of Medline and ASCO databases has conducted to identify results of clinical trials using targeted agents for thyroid cancer. Promising results were reported for drugs targeting the VEGF receptor family. Significant clinical activity was observed in these trials with objective responses in 35 % and stable disease in an additional 50 % of patients. Median progression-free survival ranged from 7 to 28 months. These results demonstrate clinical activity of several targeted agents in patients with differentiated and/or medullary thyroid cancer. Further investigation of these agents in phase III trials is warranted.

Literatur

  • 1 Reiners C, Geling M, Luster M et al. Epidemiologie des Schilddrüsenkarzinoms.  Der Onkologe. 2005;  11 11-19
  • 2 Schmid K W. Pathogenese, Klassifikation und Histologie von Schilddrüsenkarzinomen.  Der Onkologe. 2010;  16 644-656
  • 3 Riemann B, Kramer J A, Schmid K W et al. Risk stratification of patients with locally aggressive differentiated thyroid cancer. Results of the MSDS trial.  Nuklearmedizin. 2010;  49 79-84
  • 4 Holzer S, Reiners C, Mann K et al. Patterns of care for patients with primary differentiated carcinoma of the thyroid gland treated in Germany during 1996. U.S. and German Thyroid Cancer Group.  Cancer. 2000;  89 192-201
  • 5 Dietlein M, Dressler J, Eschner W et al. Procedure guidelines for radioiodine therapy of differentiated thyroid cancer (version 3).  Nuklearmedizin. 2007;  46 213-219
  • 6 Mazzaferri E L, Jhiang S M. Long-term impact of initial surgical and medical therapy on papillary and follicular thyroid cancer.  Am J Med. 1994;  97 418-428
  • 7 Verburg F A, Stokkel M P, Duren C et al. No survival difference after successful (131)I ablation between patients with initially low-risk and high-risk differentiated thyroid cancer.  Eur J Nucl Med Mol Imaging. 2010;  37 276-283
  • 8 Durante C, Haddy N, Baudin E et al. Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid carcinoma: benefits and limits of radioiodine therapy.  J Clin Endocrinol Metab. 2006;  91 2892-2899
  • 9 Roman S, Lin R, Sosa J A. Prognosis of medullary thyroid carcinoma: demographic, clinical, and pathologic predictors of survival in 1252 cases.  Cancer. 2006;  107 2134-2142
  • 10 Karger S, Krause K, Fuhrer D. Molecular pathogenesis of thyroid tumors.  Dtsch Med Wochenschr. 2006;  131 1671-1674
  • 11 Viglietto G, Chiappetta G, Martinez-Tello F J et al. RET/PTC oncogene activation is an early event in thyroid carcinogenesis.  Oncogene. 1995;  11 1207-1210
  • 12 Kimura E T, Nikiforova M N, Zhu Z et al. High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma.  Cancer Res. 2003;  63 1454-1457
  • 13 Sugg S L, Zheng L, Rosen I B et al. ret/PTC-1, -2, and -3 oncogene rearrangements in human thyroid carcinomas: implications for metastatic potential?.  J Clin Endocrinol Metab. 1996;  81 3360-3365
  • 14 Tallini G, Santoro M, Helie M et al. RET/PTC oncogene activation defines a subset of papillary thyroid carcinomas lacking evidence of progression to poorly differentiated or undifferentiated tumor phenotypes.  Clin Cancer Res. 1998;  4 287-294
  • 15 Mulligan L M, Kwok J B, Healey C S et al. Germ-line mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A.  Nature. 1993;  363 458-460
  • 16 Carlson K M, Dou S, Chi D et al. Single missense mutation in the tyrosine kinase catalytic domain of the RET protooncogene is associated with multiple endocrine neoplasia type 2B.  Proc Natl Acad Sci U S A. 1994;  91 1579-1583
  • 17 Hofstra R M, Landsvater R M, Ceccherini I et al. A mutation in the RET proto-oncogene associated with multiple endocrine neoplasia type 2B and sporadic medullary thyroid carcinoma.  Nature. 1994;  367 375-376
  • 18 Frank-Raue K, Hoppner W, Frilling A et al. Mutations of the ret protooncogene in German multiple endocrine neoplasia families: relation between genotype and phenotype. German Medullary Thyroid Carcinoma Study Group.  J Clin Endocrinol Metab. 1996;  81 1780-1783
  • 19 Garcia-Rostan G, Zhao H, Camp R L et al. ras mutations are associated with aggressive tumor phenotypes and poor prognosis in thyroid cancer.  J Clin Oncol. 2003;  21 3226-3235
  • 20 Challeton C, Bounacer A, Du Villard J A et al. Pattern of ras and gsp oncogene mutations in radiation-associated human thyroid tumors.  Oncogene. 1995;  11 601-603
  • 21 Wright P A, Lemoine N R, Mayall E S et al. Papillary and follicular thyroid carcinomas show a different pattern of ras oncogene mutation.  Br J Cancer. 1989;  60 576-577
  • 22 Esapa C T, Johnson S J, Kendall-Taylor P et al. Prevalence of Ras mutations in thyroid neoplasia.  Clin Endocrinol. 1999;  50 529-535
  • 23 Vasko V, Hu S, Wu G et al. High prevalence and possible de novo formation of BRAF mutation in metastasized papillary thyroid cancer in lymph nodes.  J Clin Endocrinol Metab. 2005;  90 5265-5269
  • 24 Xing M, Vasko V, Tallini G et al. BRAF T 1796A transversion mutation in various thyroid neoplasms.  J Clin Endocrinol Metab. 2004;  89 1365-1368
  • 25 Cohen Y, Xing M, Mambo E et al. BRAF mutation in papillary thyroid carcinoma.  J Natl Cancer Inst. 2003;  95 625-627
  • 26 Nikiforova M N, Kimura E T, Gandhi M et al. BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas.  J Clin Endocrinol Metab. 2003;  88 5399-5404
  • 27 Musholt P B, Musholt T J, Morgenstern S C et al. Follicular histotypes of oncocytic thyroid carcinomas do not carry mutations of the BRAF hot-spot.  World J Surg. 2008;  32 722-728
  • 28 Xing M, Westra W H, Tufano R P et al. BRAF mutation predicts a poorer clinical prognosis for papillary thyroid cancer.  J Clin Endocrinol Metab. 2005;  90 6373-6379
  • 29 Kroll T G, Sarraf P, Pecciarini L et al. PAX8-PPARgamma1 fusion oncogene in human thyroid carcinoma (corrected).  Science. 2000;  289 1357-1360
  • 30 Nikiforova M N, Biddinger P W, Caudill C M et al. PAX8-PPARgamma rearrangement in thyroid tumors: RT-PCR and immunohistochemical analyses.  Am J Surg Pathol. 2002;  26 1016-1023
  • 31 Marques A R, Espadinha C, Catarino A L et al. Expression of PAX8-PPAR gamma 1 rearrangements in both follicular thyroid carcinomas and adenomas.  J Clin Endocrinol Metab. 2002;  87 3947-3952
  • 32 Reddi H V, McIver B, Grebe S K et al. The paired box-8 /peroxisome proliferator-activated receptor-gamma oncogene in thyroid tumorigenesis.  Endocrinology. 2007;  148 932-935
  • 33 Karger S, Berger K, Eszlinger M et al. Evaluation of peroxisome proliferator-activated receptor-gamma expression in benign and malignant thyroid pathologies.  Thyroid. 2005;  15 997-1003
  • 34 Fuhrer D. A nuclear receptor in thyroid malignancy: is PAX8 /PPARgamma the Holy Grail of follicular thyroid cancer?.  Eur J Endocrinol. 2001;  144 453-456
  • 35 Garcia-Rostan G, Costa A M, Pereira-Castro I et al. Mutation of the PIK3CA gene in anaplastic thyroid cancer.  Cancer Res. 2005;  65 10 199-10 207
  • 36 Hou P, Liu D, Shan Y et al. Genetic alterations and their relationship in the phosphatidylinositol 3-kinase/Akt pathway in thyroid cancer.  Clin Cancer Res. 2007;  13 1161-1170
  • 37 Santarpia L, El-Naggar A K, Cote G J et al. Phosphatidylinositol 3-kinase/akt and ras/raf-mitogen-activated protein kinase pathway mutations in anaplastic thyroid cancer.  J Clin Endocrinol Metab. 2008;  93 278-284
  • 38 Donghi R, Longoni A, Pilotti S et al. Gene p53 mutations are restricted to poorly differentiated and undifferentiated carcinomas of the thyroid gland.  J Clin Invest. 1993;  91 1753-1760
  • 39 Fagin J A, Matsuo K, Karmakar A et al. High prevalence of mutations of the p53 gene in poorly differentiated human thyroid carcinomas.  J Clin Invest. 1993;  91 179-184
  • 40 Ito T, Seyama T, Mizuno T et al. Unique association of p53 mutations with undifferentiated but not with differentiated carcinomas of the thyroid gland.  Cancer Res. 1992;  52 1369-1371
  • 41 Klagge A, Krause K, Schierle K et al. Somatostatin receptor subtype expression in human thyroid tumours.  Horm Metab Res. 2010;  42 237-240
  • 42 Weidinger C, Karger S, Krause K et al. Distinct regulation of intrinsic apoptosis in benign and malignant thyroid tumours.  Horm Metab Res. 2010;  42 553-556
  • 43 Viglietto G, Maglione D, Rambaldi M et al. Upregulation of vascular endothelial growth factor (VEGF) and downregulation of placenta growth factor (PlGF) associated with malignancy in human thyroid tumors and cell lines.  Oncogene. 1995;  11 1569-1579
  • 44 Bunone G, Vigneri P, Mariani L et al. Expression of angiogenesis stimulators and inhibitors in human thyroid tumors and correlation with clinical pathological features.  Am J Pathol. 1999;  155 1967-1976
  • 45 Matuszczyk A, Petersenn S, Bockisch A et al. Chemotherapy with doxorubicin in progressive medullary and thyroid carcinoma of the follicular epithelium.  Horm Metab Res. 2008;  40 210-213
  • 46 Gottlieb J A, Hill Jr C S. Chemotherapy of thyroid cancer with adriamycin. Experience with 30 patients.  N Engl J Med. 1974;  290 193-197
  • 47 Shimaoka K, Schoenfeld D A, DeWys W D et al. A randomized trial of doxorubicin versus doxorubicin plus cisplatin in patients with advanced thyroid carcinoma.  Cancer. 1985;  56 2155-2160
  • 48 Williams S D, Birch R, Einhorn L H. Phase II evaluation of doxorubicin plus cisplatin in advanced thyroid cancer: a Southeastern Cancer Study Group Trial.  Cancer Treat Rep. 1986;  70 405-407
  • 49 Matuszczyk A, Petersenn S, Voigt W et al. Chemotherapy with paclitaxel and gemcitabine in progressive medullary and thyroid carcinoma of the follicular epithelium.  Horm Metab Res. 2010;  42 61-64
  • 50 Matuszczyk A, Mann K. Neue Therapieformen beim Schilddrüsenkarzinom.  Der Onkologe. 2010;  16 690-694
  • 51 Wells Jr S A, Gosnell J E, Gagel R F et al. Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer.  J Clin Oncol. 2010;  28 767-772
  • 52 Wells S A, Robinson B G, Gagel R F et al. Vandetanib (VAN) in locally advanced or metastatic medullary thyroid cancer (MTC): A randomized, double-blind phase III trial (ZETA).  J Clin Oncol. 2010;  28 15s (suppl abstr 5503)
  • 53 Gupta-Abramson V, Troxel A B, Nellore A et al. Phase II trial of sorafenib in advanced thyroid cancer.  J Clin Oncol. 2008;  26 4714-4719
  • 54 Kloos R T, Ringel M D, Knopp M V et al. Phase II trial of sorafenib in metastatic thyroid cancer.  J Clin Oncol. 2009;  27 1675-1684
  • 55 Lam E T, Ringel M D, Kloos R T et al. Phase II clinical trial of sorafenib in metastatic medullary thyroid cancer.  J Clin Oncol. 2010;  28 2323-2330
  • 56 Cohen E E, Needles B M, Cullen K J et al. Phase 2 study of sunitinib in refractory thyroid cancer.  J Clin Oncol. 2008;  26 (20 suppl) abstr 6025
  • 57 De Souza J A, Busaidy N, Zimrin A et al. Phase II trial of sunitinib in medullary thyroid cancer (MTC).  J Clin Oncol. 2010;  28 15s (suppl abstr 5504)
  • 58 Carr L L, Mankoff D, Goulart B H et al. Phase II Study of Sunitinib in FDG-PET Positive, Differentiated Thyroid Cancer and Metastatic Medullary Carcinoma of Thyroid with Functional Imaging Correlation.  Clin Cancer Res. 2010;  16 5260-5268
  • 59 Bible K C, Suman V J, Molina J R et al. Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: results of a phase 2 consortium study.  Lancet Oncol. 2010;  11 962-972
  • 60 Kurzrock R, Sherman S I, Ball D W et al. Activity of XL 184 (Cabozantinib), an Oral Tyrosine Kinase Inhibitor, in Patients With Medullary Thyroid Cancer.  J Clin Oncol. 2011;  29 2660-2666
  • 61 Sherman S I, Jarzab B, Cabanillas M E et al. A phase II trial of the multitargeted kinase inhibitor E 7080 in advanced radioiodine (RAI)-refractory differentiated thyroid cancer (DTC).  J Clin Oncol. 2011;  29 8527 (suppl abstr 5503)
  • 62 Schlumberger M J, Elisei R, Bastholt L et al. Phase II study of safety and efficacy of motesanib in patients with progressive or symptomatic, advanced or metastatic medullary thyroid cancer.  J Clin Oncol. 2009;  27 3794-3801
  • 63 Sherman S I, Wirth L J, Droz J P et al. Motesanib diphosphate in progressive differentiated thyroid cancer.  N Engl J Med. 2008;  359 31-42
  • 64 Cohen E E, Rosen L S, Vokes E E et al. Axitinib is an active treatment for all histologic subtypes of advanced thyroid cancer: results from a phase II study.  J Clin Oncol. 2008;  26 4708-4713
  • 65 Ain K B, Lee C, Holbrook K M et al. Phase II study of lenalidomide in distantly metastatic, rapidly progressive, and radioiodine-unresponsive thyroid carcinomas: preliminary results.  J Clin Oncol. 2008;  26 (20 suppl abstr 6027)
  • 66 Ain K B, Lee C, Williams K D. Phase II trial of thalidomide for therapy of radioiodine-unresponsive and rapidly progressive thyroid carcinomas.  Thyroid. 2007;  17 663-670
  • 67 Pennell N A, Daniels G H, Haddad R I et al. A phase II study of gefitinib in patients with advanced thyroid cancer.  J Clin Oncol. 2007;  25 (20 suppl abstr 6020)
  • 68 Sherman E J, Ho A L, Haque S et al. A phase II study of VEGF trap (aflibercept) in patients with radioactive iodine-refractory, positron emission tomography (PET) positive thyroid carcinoma.  J Clin Oncol. 2010;  28 15 s (suppl abstr 5587)
  • 69 Frank-Raue K, Fabel M, Delorme S et al. Efficacy of imatinib mesylate in advanced medullary thyroid carcinoma.  Eur J Endocrinol. 2007;  157 215-220

PD Dr. med. Philipp Schütt

Innere Klinik (Tumorforschung), Universitätsklinikum Essen

Hufelandstraße 55

45122 Essen

Email: philipp.schuett@uni-due.de

    >