Emerging therapies for thyroid carcinoma

Emerging therapies for thyroid carcinoma

t h e s u r g e o n 1 0 ( 2 0 1 2 ) 5 3 e5 8 available at www.sciencedirect.com The Surgeon, Journal of the Royal Colleges of Surgeons of Edinburgh ...

151KB Sizes 0 Downloads 128 Views

t h e s u r g e o n 1 0 ( 2 0 1 2 ) 5 3 e5 8

available at www.sciencedirect.com

The Surgeon, Journal of the Royal Colleges of Surgeons of Edinburgh and Ireland www.thesurgeon.net

Review

Emerging therapies for thyroid carcinoma S. Walsh*, R. Prichard, A.D.K. Hill Department of Surgery, RCSI Smurfitt Building, Beaumont Hospital, Dublin 9, Ireland

article info

abstract

Article history:

Thyroid carcinoma is the most commonly diagnosed endocrine malignancy. Its incidence

Received 20 March 2011

is currently rising worldwide. The discovery of genetic mutations associated with the

Received in revised form

development of thyroid cancer, such as BRAF and RET, has lead to the development of new

1 August 2011

drugs which target the pathways which they influence. Despite recent advances, the

Accepted 17 August 2011

prognosis of anaplastic thyroid carcinoma is still unfavourable. In this review we look at

Available online 9 September 2011

emerging novel therapies for the treatment of well-differentiated and medullary thyroid carcinoma, and advances and future directions in the management of anaplastic thyroid

Keywords: Thyroid carcinoma Tyrosine kinase inhibitors

carcinoma. ª 2011 Royal College of Surgeons of Edinburgh (Scottish charity number SC005317) and Royal College of Surgeons in Ireland. Published by Elsevier Ltd. All rights reserved.

Papillary Medullary Anaplastic BRAF RET Vandetanib Sunitinib Sorafenib Imatinib Motesanib

Introduction Thyroid carcinoma is the commonest endocrine malignancy, with an overall estimated incidence of 7.7 per 100,000.1 The last two decades have seen a worldwide rise in the incidence,2 with papillary carcinoma showing the greatest proportional increase over time.3 Thyroid carcinomas comprise a heterogenous group of tumours with different clinical and pathological characteristics and can be loosely divided into three main

subgroups. Differentiated thyroid carcinomas include papillary, follicular and Hu¨rthle cell carcinomas. These tumours arise from follicular thyroid cells and have similarities in their favourable clinical course and the principles of their management.4 Medullary thyroid carcinoma originates from parafollicular C-cells. However, one quarter of medullary carcinomas are caused by point mutations in the RET oncogene and are associated with familial syndromes, including multiple endocrine neoplasia type 2A and type 2B.5 Anaplastic

* Corresponding author. E-mail address: [email protected] (S. Walsh). 1479-666X/$ e see front matter ª 2011 Royal College of Surgeons of Edinburgh (Scottish charity number SC005317) and Royal College of Surgeons in Ireland. Published by Elsevier Ltd. All rights reserved. doi:10.1016/j.surge.2011.08.004

54

t h e s u r g e o n 1 0 ( 2 0 1 2 ) 5 3 e5 8

carcinoma, the majority of which arise de novo with a small proportion arising from well-differentiated thyroid cancers, responds disappointingly to conventional therapies and has an extremely poor overall prognosis.6 In general the treatment of thyroid tumours comprises surgical resection, radioactive iodine and thyroid hormone suppressive therapy.7 Overall prognosis is excellent with a 98% 10-year survival for differentiated thyroid carcinoma.8 However when patients develop metastatic disease, systemic chemotherapy is of limited benefit with poor overall response rates and little increase in overall survival rates.9 For patients with poorly responsive differentiated thyroid carcinoma or metastatic medullary carcinoma novel therapeutic options are desperately needed to improve disease outcomes. Multiple novel therapies targeting angiogenesis have been recently developed and are currently in clinical trials assessing their efficacy. Anaplastic carcinoma almost forms a separate entity with an overall 1 year survival rate of 20%.6 Surgical intervention is largely aimed at providing local control and has little impact on overall survival, while the majority are insensitive to radioactive iodine.10 Therefore, new developments are predominantly aimed at neoadjuvant treatment with a combination of systemic chemotherapy and radiotherapy. The aim of this review is discuss the biological mutations underpinning advanced thyroid carcinoma and to focus on the emerging novel therapies for its treatment. Secondly, an update on the current advances in the management of patients with anaplastic thyroid carcinoma will be provided.

Targeting cell signalling pathways Tyrosine kinase inhibitors Protein kinases are amongst the most promising new targets for the treatment of cancer. They act by regulating cellular signal transduction pathways, thereby influencing cell growth, survival and differentiation. The identification of the dysregulation and upregulation of various protein kinases, and the pathways in which they are involved, has led to the development of tyrosine kinase inhibitors. In 2002, imatinib (Gleevec) was introduced into routine clinical practice for the treatment of gastrointestinal stromal tumours. Imatinib was also found to be highly effective in the treatment of chronic myeloid leukaemia.11 The addition the tyrosine kinase inhibitor lapatinib to the treatment of breast carcinoma which overexpresses the HER2 oncogene has significantly improved the prognosis of these patients.12 The success of these tyrosine kinase inhibitors has led to increased research into the effect on tyrosine kinase inhibition on a vast range of solid tumours, including thyroid carcinoma.

Thyroid carcinoma mutations Recent endeavours to elicit the pathogenesis of thyroid carcinoma have revealed several genetic mutations which lead to its development including aberrations of BRAF and RET.

BRAF Several intracellular signalling pathways, including the PI3K/ AKT cell signalling pathway and the MAPK (mitogen activated protein kinase) cell signalling cascade have been shown to be integral to the development and progression of thyroid carcinoma.13 Among the three forms of Raf kinases, BRAF (the gene for the B-type Raf kinase), with its gene located on chromosome 7, is the most potent activator of the MAPK signalling pathway. BRAF activating point mutations are clustered in exons 11 and 15 of the gene.14 The most common genetic alteration in thyroid carcinoma is the oncogenic T1799A transversion mutation of BRAF.14 This aberrancy, which was discovered in 2008, is observed in approximately 44% of papillary thyroid carcinoma and 24% of anaplastic thyroid carcinoma.14 Raf is a key component of the MAPK signalling cascade, of which there are several inhibitors. The MEK inhibitor, CI-1040, has been shown to inhibit the proliferation and growth of thyroid cancer cells in vitro.15 This effect seemed to be BRAF mutation or RAS mutation selective. Thyroid cancer cell lines containing either RET or RAF mutations were found to be highly sensitive to two RAF kinase inhibitors, AAL-881 and LBT-613.16 AZD6244 is an inhibitor of MAPK ERK, which is down stream of RAF, inhibited the MEKERK pathway across a spectrum of thyroid cancer cells, and was also most effective in BRAF mutated cell lines.17

RET The RET (REarranged during Transcription) receptor protein is a tyrosine kinase influencing cell survival.18 The RET oncogene is located on chromosome 10q11.2.19 After dimerization, RET activates several signal transduction pathways, including the MAPK signalling cascade and the PI3K/AKT cell signalling pathway, leading to increased cell growth, survival and proliferation.20 Point mutations in the RET proto-oncogene are responsible for almost all inherited and almost half of all sporadic medullary thyroid carcinomas.21 Exposure to radiation is a major risk factor for the development of RET associated papillary thyroid carcinoma.19 Currently there is no tyrosine kinase inhibitor specific to RET available, but there are several multi-targeted tyrosine kinase inhibitors which have demonstrated significant activity against RET, including vandetanib (ZD6474), sorafenib (BAY 43-9006), motesanib, imatenib and sunitinib.

Clinical evidence for tyrosine kinase inhibitors in thyroid carcinoma Vandetanib Vandetanib (ZD6474) is an orally available tyrosine kinase inhibitor, which targets vascular endothelial growth factor receptor (VEGR), epidermal growth factor receptor (EGFR) and RET signalling.22 Genetic alterations of RET have been implicated in the pathophysiology of medullary thyroid carcinoma. Upregulation of VEGF in thyroid carcinoma in comparison with benign thyroid tissue was first observed in 1997.23 Increased expression of EGFR has been observed in follicular, medullary and anaplastic thyroid cancer.24e26 The initial phase II clinical trials performed in thirty patients with advanced or unresectable hereditary medullary thyroid

t h e s u r g e o n 1 0 ( 2 0 1 2 ) 5 3 e5 8

cancer demonstrated good response to 300 mg vandetanib daily, with minimal adverse events.27 Further clinical trials showed the over half of patients with advanced hereditary MTC achieved disease stability after receiving only 100 mg vandetanib daily.28 The results of ZETA, the first phase III multi-centre study evaluating oral once-daily vandetanib 300 mg in 331 patients with unresectable, locally advanced or metastatic hereditary or sporadic medullary thyroid cancer, have recently been released.29 The investigators looked at progression free survival, objective response rate and disease control rate. Over half of the patients enrolled were positive for RET mutation. A response rate of 45% was reported as well as a significant decrease in calcitonin and CEA biomarkers. These results are promising although limited by the advanced disease of the patients enrolled.

Sorafenib Sorafenib (BAY 43-9006) is a multi-kinase inhibitor which targets BRAF, VEGFR, KIT (a stem cell growth factor protooncogene involved in cell survival and differentiation) and platelet-derived growth factor receptor (PDGFR), and has been demonstrated to strongly inhibit RET kinase in vitro.30 The original phase II clinical trial, in which 400 mg of sorafenib was administered twice daily to 21 patients with metastatic medullary carcinoma showed that 87% of the patients with sporadic MTC achieved stable disease.31 In a similar trial, all 5 patients treated with sorafenib exhibited partial response.32 In both of these trials the authors reported side-effects which included hand and foot syndrome (HFS), fatigue and diarrhea. Hand and foot syndrome, characterized by a painful palmarplantar rash, is a cutaneous side-effect associated with various chemotherapy agents. Sorafenib has emerged as a leading cause of HFS, with 10e62% of patients treated with it reporting the side-effect.33 A more recent study has shown that patients with metastatic papillary thyroid carcinoma also respond to treatment with sorafenib, suggesting that the therapeutic use of sorafenib may not be limited to the treatment of medullary thyroid carcinoma.34 In this study, the partial response rate was 33% and the stable disease rate, 44%.

Imatinib Imatinib (Gleevec) is a tyrosine kinase inhibitor which inhibits RET, PDGFR and KIT, and has revolutionized the treatment of chronic myeloid leukaemia. Early cell-line studies showed that treatment with imatinib significantly inhibited growth in anaplastic p-53 mutated thyroid carcinoma cell lines, but not in p-53 mutated papillary thyroid carcinoma cell lines.35 Further in vivo studies demonstrated additive effects between imatinib and gefitinib, an inhibitor of HER1, or endothelial growth factor receptor, when used to treat anaplastic thyroid carcinoma xenografts in nude mice.36 However a phase II clinical trial testing the use of imatinib in medullary thyroid carcinoma yielded disappointing results.37 Less than a quarter of the patients achieved stable disease and there was no evidence of partial response. Furthermore, patients with hypothyroidism who were being treated with imatinib developed increased levothyroxine requirements.38 In contrast, another trial testing imatinib in a similar cohort of patients reported that the drug was well tolerated, although disease stability was only transient.39

55

Sunitinib Sunitinib is a small molecule inhibitor targeting similar protein kinases to sorafenibdVEGFR, PDGFR, KIT and RET. It has been shown to downregulate the MEK/ERK and SAPK/JNK cell signalling pathways in papillary thyroid carcinoma.40 Phase I clinical trials showed acceptable safety profiles and partial response to sunitinib in conjunction with capecitabine in patients with advanced thyroid carcinoma.41 Subsequently a phase II clinical trial treated 33 patients with either welldifferentiated thyroid carcinoma, or medullary thyroid carcinoma with sunitinib.42 One patient had complete response, 28% had a partial response and 46% exhibited disease stability. The side-effect profile was similar to that of sorafenib, and included hand and foot syndrome, neutropaenia and fatigue. The first results of the THYSU trial were recently released.43 Patients with iodine-refractive advanced thyroid cancer were treated with 50 mg daily for 4 weeks, every 6 weeks. Of the 15 patients eligible for evaluation, one had a partial response and twelve demonstrated disease stability. The final results are yet to be released. These papers suggest that sunitinib is a promising treatment for thyroid carcinoma. However, it is important to bear in mind that sunitinb causes an increase in levothyrroxine requirements.44 This warrants caution when treating a cohort of patients who may have undergone thyroid surgery as part of their cancer treatment.

Motesanib Motesanib, like sorafenib and sunitinib, is a tyrosine kinase inhibitor targeting RET, VEGFR, PDGFR and KIT. It is orally available, facilitating easy administration. Motesanib was tested in a cohort of 93 patients with advanced thyroid carcinoma, comprising 61% papillary thyroid carcinoma.45 A response rate of 14% and disease stability rate of 67% was observed. A phase II clinical trial in which motesanib was given to patients with advanced, progressive or metastatic medullary carcinoma reported an impressive disease stability rate of 81%, and significant decreases in tumour markers.46 Changes in serum placental growth factor and soluble VEGF two weeks after commencement of treatment have been identified as potential predictors of response to motesanib.47 In view of the impressive response rates, favourable safety profile and availability of potential predictors of response motesanib may in the future play a role in the treatment of both differentiated and medullary thyroid carcinoma.

Other emerging therapies There are several other therapeutic agents undergoing investigation for their use in thyroid carcinoma. Src is a protooncogene involved in signalling and is thus involved in the control of cell proliferation, cell differentiation, migration, angiogenesis, and survival. Src is thus thought to play a key role in tumour formation and progression.48e50 Four out of five thyroid cancer cell lines showed good sensitivity to inhibition of Src with AZD0530.51 Further investigation is warranted before progression to clinical trials. Bortezomib effectively inhibited thyroid tumour growth in vivo and in vitro.52 This finding prompted the commencement of two clinical trials, one testing its use in iodine refractory metastatic thyroid cancer (NCT00104871), and the other testing its use in

56

t h e s u r g e o n 1 0 ( 2 0 1 2 ) 5 3 e5 8

conjunction with vandetanib in the treatment of metastatic medullary thyroid carcinoma (NCT00923247). A key down stream component in the PI3K pathway is the serine/threonine kinase known as mTOR, the human target of rapamycin. mTOR is thus a critical regulator of tumour formation and progression. Consistent with its role in tumour formation and progression, targeted therapy against mTOR has been shown to decrease tumour growth in model systems.53e55 Indeed, several agents that inhibit mTor are currently in clinical trials for the treatment of multiple cancer types.56e58 In vitro studies have shown that anaplastic and medullary thyroid cancer cell lines are sensitive to treatment with everolimus (RAD001), a novel inhibitor of mTor.59,60 In vitro studies revealed that while everolimus blocked tumour growth, it did not slow disease progression.61 Thus the recommendation was to combine everolimus with other therapies which will inhibit metastasis. There are currently several clinical trials recruiting patients with advanced thyroid carcinoma, to test everolimus either alone, or in combination with sorafenib (NCT01263951, NCT01118065, NCT01164176).

Anaplastic thyroid carcinoma Anaplastic thyroid carcinoma accounts for merely 2e5% of all thyroid cancer.62 Due to their aggressive nature, they are classified as T4 and Stage IV, regardless of the actual tumour burden. Research into the treatment of anaplastic thyroid carcinoma is deficient due to the rarity of the tumour and the rapid progression of the disease. As a result survival rates are low (20% at one year) and this has remained virtually static in the past fifty years.63 However a recent study of 75 patients with anaplastic thyroid carcinoma showed remission in 89% of those who received surgical therapy and chemoradiation. These patients also had a longer median survival.64 A retrospective analysis of 5 patients demonstrated that a combination of radiation and docetaxel was effective for the treatment of anaplastic thyroid cancer.65 In this cohort, two patients achieved complete response, and two patients partial response. A study by Foote et al also reported improved survival rates of 70% at 1 year and 60% at 2 years following aggressive multimodal therapy for stage IV anaplastic thyroid carcinoma.66 Aggressive multimodal therapy constituted surgery, if feasible, intensity-modulated radiation therapy and radiosensitising adjuvant chemotherapy involving four cycles of docetaxel and doxorubicin. A Serbian study reported a clinical benefit of 50% from treatment with radiotherapy followed by doxorubicin and cisplatin.67 A series of 50 patients treated in Hong Kong further demonstrated that patients with anaplastic thyroid carcinoma benefit from an aggressive multidisciplinary approach, combining surgery, radiotherapy and chemotherapy.68 A recent study examining the effect of imatinib on patients with advanced anaplastic thyroid carcinoma reported that of the 8 patients included for analysis, 2 exhibited partial response and 4 achieved disease stability.69 Preclinical data indicates that the main role for this drug lies in the treatment of anaplastic thyroid carcinoma. The main challenges in the search for new treatments for anaplastic thyroid carcinoma

are the rarity of this tumour and its aggressive nature, leading to difficulty in recruiting patients for clinical trials. There is clearly a need for a large multi-centred clinical trial testing the use of imatinib in metastatic anaplastic thyroid carcinoma. There are two phase II clinical trials in the recruitment phase, testing the use of sorafenib alone, and bevacizimab with doxorubicin, in advanced anaplastic thyroid carcinoma (NCT00126568, NCT00804830).

Conclusion The treatment of well-differentiated and medullary thyroid carcinomas has recently seen many exciting advances, with new targeted drugs achieving promising results in phase III clinical trials. However these advances have yet to become applicable to anaplastic thyroid carcinoma, which still carries a dismal prognosis in contrast with other subtypes of thyroid carcinoma. Preclinical studies have yielded promising results in the response of anaplastic thyroid cancer to a number of targeted therapies. However, the difficulty in recruiting these patients, who frequently have advanced disease at presentation, to participate in clinical trials has resulted in these drugs failing to be adequately tested clinically. Collaboration between cancer centres worldwide is imperative in recruiting sufficient cases of anaplastic thyroid cancer to prove the efficacy of new drugs in the treatment of this rare, but lethal cancer.

references

1. Aschebrook-Kilfoy B, Ward MH, Sabra MM, Devesa SS. Thyroid cancer incidence patterns in the United States by histologic type, 1992-2006. Thyroid 2011;21:125e34. 2. Sipos JA, Mazzaferri EL. Thyroid cancer epidemiology and prognostic variables. Clin Oncol (R Coll Radiol) 2010;22:395e404. 3. Enewold L, Zhu K, Ron E, Marrogi AJ, Stojadinovic A, Peoples GE, et al. Rising thyroid cancer incidence in the United States by demographic and tumor characteristics, 1980-2005. Cancer Epidemiol Biomarkers Prev 2009;18:784e91. 4. Hundahl SA, Cady B, Cunningham MP, Mazzaferri E, McKee RF, Rosai J, et al. Initial results from a prospective cohort study of 5583 cases of thyroid carcinoma treated in the united states during 1996. U.S. and German Thyroid Cancer Study Group. An American College of Surgeons Commission on Cancer Patient Care Evaluation study. Cancer 2000;89: 202e17. 5. Pacini F, Castagna MG, Cipri C, Schlumberger M. Medullary thyroid carcinoma. Clin Oncol (R Coll Radiol) 2010;22:475e85. 6. Kebebew E, Greenspan FS, Clark OH, Woeber KA, McMillan A. Anaplastic thyroid carcinoma. Treatment outcome and prognostic factors. Cancer 2005;103:1330e5. 7. Sherman SI. Thyroid carcinoma. Lancet 2003;361:501e11. 8. Cushing SL, Palme CE, Audet N, Eski S, Walfish PG, Freeman JL. Prognostic factors in well-differentiated thyroid carcinoma. Laryngoscope 2004;114:2110e5. 9. Haugen BR. Management of the patient with progressive radioiodine non-responsive disease. Semin Surg Oncol 1999;16: 34e41. 10. Namba H, Hara T, Tukazaki T, Migita K, Ishikawa N, Ito K, et al. Radiation-induced G1 arrest is selectively mediated by the p53-WAF1/Cip1 pathway in human thyroid cells. Cancer Res. 1995;55:2075e80.

t h e s u r g e o n 1 0 ( 2 0 1 2 ) 5 3 e5 8

11. Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med 2001;344:1031e7. 12. Paul B, Trovato JA, Thompson J. Lapatinib: a dual tyrosine kinase inhibitor for metastatic breast cancer. Am J Health Syst Pharm 2008;65:1703e10. 13. Xing M. Genetic alterations in the phosphatidylinositol-3 kinase/Akt pathway in thyroid cancer. Thyroid 2010;20: 697e706. 14. Xing M. BRAF mutation in thyroid cancer. Endocr Relat Cancer 2005;12:245e62. 15. Liu D, Liu Z, Jiang D, Dackiw AP, Xing M. Inhibitory effects of the mitogen-activated protein kinase kinase inhibitor CI-1040 on the proliferation and tumor growth of thyroid cancer cells with BRAF or RAS mutations. J Clin Endocrinol Metab 2007;92: 4686e95. 16. Ouyang B, Knauf JA, Smith EP, Zhang L, Ramsey T, Yusuff N, et al. Inhibitors of Raf kinase activity block growth of thyroid cancer cells with RET/PTC or BRAF mutations in vitro and in vivo. Clin Cancer Res. 2006;12:1785e93. 17. Ball DW, Jin N, Rosen DM, Dackiw A, Sidransky D, Xing M, et al. Selective growth inhibition in BRAF mutant thyroid cancer by the mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244. J Clin Endocrinol Metab 2007;92:4712e8. 18. Carlomagno F, Santoro M. Identification of RET kinase inhibitors as potential new treatment for sporadic and inherited thyroid cancer. J Chemother 2004;16(Suppl. 4):49e51. 19. de Groot JW, Links TP, Plukker JT, Lips CJ, Hofstra RM. RET as a diagnostic and therapeutic target in sporadic and hereditary endocrine tumors. Endocr Rev. 2006;27:535e60. 20. De Vita G, Melillo RM, Carlomagno F, Visconti R, Castellone MD, Bellacosa A, et al. Tyrosine 1062 of RETMEN2A mediates activation of Akt (protein kinase B) and mitogen-activated protein kinase pathways leading to PC12 cell survival. Cancer Res. 2000;60:3727e31. 21. Phay JE, Shah MH. Targeting RET receptor tyrosine kinase activation in cancer. Clin Cancer Res. 2010;16:5936e41. 22. Carlomagno F, Vitagliano D, Guida T, Ciardiello F, Tortora G, Vecchio G, et al. ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Res. 2002;62:7284e90. 23. Soh EY, Duh QY, Sobhi SA, Young DM, Epstein HD, Wong MG, et al. Vascular endothelial growth factor expression is higher in differentiated thyroid cancer than in normal or benign thyroid. J Clin Endocrinol Metab 1997;82:3741e7. 24. Ensinger C, Spizzo G, Moser P, Tschoerner I, Prommegger R, Gabriel M, et al. Epidermal growth factor receptor as a novel therapeutic target in anaplastic thyroid carcinomas. Ann N Y Acad Sci. 2004;1030:69e77. 25. Westermark K, Lundqvist M, Wallin G, Dahlman T, Hacker GW, Heldin NE, et al. EGF-receptors in human normal and pathological thyroid tissue. Histopathology 1996;28:221e7. 26. Di Carlo A, Mariano A, Pisano G, Parmeggiani U, Beguinot L, Macchia V. Epidermal growth factor receptor and thyrotropin response in human thyroid tissues. J Endocrinol Invest 1990;13: 293e9. 27. Wells Jr SA, Gosnell JE, Gagel RF, Moley J, Pfister D, Sosa JA, et al. Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Oncol 2010;28:767e72. 28. Robinson BG, Paz-Ares L, Krebs A, Vasselli J, Haddad R. Vandetanib (100 mg) in patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Endocrinol Metab 2010;95:2664e71. 29. Wells SA, BGR, Gagel RF, Dralle H, Fagin JA, Santoro M, Baudin E, Vasselli JR, Read J, Schlumberger M. Vandetanib (VAN) in locally advanced or metastatic medullary thyroid

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

57

cancer (MTC): A randomized, double-blind phase III trial (ZETA). J Clin Oncol 2010;28. abst 5508. Carlomagno F, Anaganti S, Guida T, Salvatore G, Troncone G, Wilhelm SM, et al. BAY 43-9006 inhibition of oncogenic RET mutants. J Natl Cancer Inst 2006;98:326e34. Lam ET, Ringel MD, Kloos RT, Prior TW, Knopp MV, Liang J, et al. Phase II clinical trial of sorafenib in metastatic medullary thyroid cancer. J Clin Oncol 2010;28:2323e30. Frank-Raue K, Ganten M, Kreissl MC, Raue F. Rapid response to sorafenib in metastatic medullary thyroid carcinoma. Exp Clin Endocrinol Diabetes 2011;119:151e5. Lipworth AD, Robert C, Zhu AX. Hand-foot syndrome (handfoot skin reaction, palmar-plantar erythrodysesthesia): focus on sorafenib and sunitinib. Oncology 2009;77:257e71. Chen L, Shen Y, Luo Q, Yu Y, Lu H, Zhu R. Response to sorafenib at a low dose in patients with radioiodinerefractory pulmonary metastases from papillary thyroid carcinoma. Thyroid 2011;21:119e24. Podtcheko A, Ohtsuru A, Tsuda S, Namba H, Saenko V, Nakashima M, et al. The selective tyrosine kinase inhibitor, STI571, inhibits growth of anaplastic thyroid cancer cells. J Clin Endocrinol Metab 2003;88:1889e96. Kurebayashi J, Okubo S, Yamamoto Y, Ikeda M, Tanaka K, Otsuki T, et al. Additive antitumor effects of gefitinib and imatinib on anaplastic thyroid cancer cells. Cancer Chemother Pharmacol 2006;58:460e70. de Groot JW, Zonnenberg BA, van Ufford-Mannesse PQ, de Vries MM, Links TP, Lips CJ, et al. A phase II trial of imatinib therapy for metastatic medullary thyroid carcinoma. J Clin Endocrinol Metab 2007;92:3466e9. de Groot JW, Zonnenberg BA, Plukker JT, van Der Graaf WT, Links TP. Imatinib induces hypothyroidism in patients receiving levothyroxine. Clin Pharmacol Ther 2005;78:433e8. Frank-Raue K, Fabel M, Delorme S, Haberkorn U, Raue F. Efficacy of imatinib mesylate in advanced medullary thyroid carcinoma. Eur J Endocrinol 2007;157:215e20. Fenton MS, Marion KM, Salem AK, Hogen R, Naeim F, Hershman JM. Sunitinib inhibits MEK/ERK and SAPK/JNK pathways and increases sodium/iodide symporter expression in papillary thyroid cancer. Thyroid 2010;20:965e74. Sweeney CJ, Chiorean EG, Verschraegen CF, Lee FC, Jones S, Royce M, et al. A phase I study of sunitinib plus capecitabine in patients with advanced solid tumors. J Clin Oncol 2010;28: 4513e20. Carr LL, Mankoff DA, Goulart BH, Eaton KD, Capell PT, Kell EM, et al. Phase II study of daily sunitinib in FDG-PETpositive, iodine-refractory differentiated thyroid cancer and metastatic medullary carcinoma of the thyroid with functional imaging correlation. Clin Cancer Res. 2010;16: 5260e8. Ravaud A, de la Fouchardiere C, Asselineau J, Delord JP, Do Cao C, Niccoli P, et al. Efficacy of sunitinib in advanced medullary thyroid carcinoma: intermediate results of phase II THYSU. Oncologist 2010;15:212e3. author reply 4. Torino F, Corsello SM, Longo R, Barnabei A, Gasparini G. Hypothyroidism related to tyrosine kinase inhibitors: an emerging toxic effect of targeted therapy. Nat Rev Clin Oncol 2009;6:219e28. Sherman SI, Wirth LJ, Droz JP, Hofmann M, Bastholt L, Martins RG, et al. Motesanib diphosphate in progressive differentiated thyroid cancer. N Engl J Med 2008;359:31e42. Schlumberger MJ, Elisei R, Bastholt L, Wirth LJ, Martins RG, Locati LD, 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;27:3794e801. Bass MB, Sherman SI, Schlumberger MJ, Davis MT, Kivman L, Khoo HM, et al. Biomarkers as predictors of response to

58

48. 49. 50. 51.

52.

53.

54.

55.

56. 57.

58. 59.

t h e s u r g e o n 1 0 ( 2 0 1 2 ) 5 3 e5 8

treatment with motesanib in patients with progressive advanced thyroid cancer. J Clin Endocrinol Metab 2010;95: 5018e27. Kim LC, Song L, Haura EB. Src kinases as therapeutic targets for cancer. Nat Rev Clin Oncol 2009;6:587e95. Wheeler DL, Iida M, Dunn EF. The role of Src in solid tumors. Oncologist 2009;14:667e78. Finn RS. Targeting Src in breast cancer. Ann Oncol 2008;19: 1379e86. Schweppe RE, Kerege AA, French JD, Sharma V, Grzywa RL, Haugen BR. Inhibition of Src with AZD0530 reveals the SrcFocal Adhesion kinase complex as a novel therapeutic target in papillary and anaplastic thyroid cancer. J Clin Endocrinol Metab 2009;94:2199e203. Stenner F, Liewen H, Zweifel M, Weber A, Tchinda J, Bode B, et al. Targeted therapeutic approach for an anaplastic thyroid cancer in vitro and in vivo. Cancer Sci. 2008;99:1847e52. Mabuchi S, Kawase C, Altomare DA, Morishige K, Sawada K, Hayashi M, et al. mTOR is a promising therapeutic target both in cisplatin-sensitive and cisplatin-resistant clear cell carcinoma of the ovary. Clin Cancer Res. 2009;15:5404e13. Lu CH, Wyszomierski SL, Tseng LM, Sun MH, Lan KH, Neal CL, et al. Preclinical testing of clinically applicable strategies for overcoming trastuzumab resistance caused by PTEN deficiency. Clin Cancer Res. 2007;13:5883e8. Chresta CM, Davies BR, Hickson I, Harding T, Cosulich S, Critchlow SE, et al. AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res.70: 288e298. Courtney KD, Corcoran RB, Engelman JA. The PI3K pathway as drug target in human cancer. J Clin Oncol.28: 1075e1083. Bose S, Chandran S, Mirocha JM, Bose N. The Akt pathway in human breast cancer: a tissue-array-based analysis. Mod Pathol 2006;19:238e45. Alvarez RH, Valero V, Hortobagyi GN. Emerging targeted therapies for breast cancer. J Clin Oncol.28: 3366e3379. Papewalis C, Wuttke M, Schinner S, Willenberg HS, Baran AM, Scherbaum WA, et al. Role of the novel mTOR inhibitor RAD001 (everolimus) in anaplastic thyroid cancer. Horm Metab Res. 2009;41:752e6.

60. Grozinsky-Glasberg S, Rubinfeld H, Nordenberg Y, Gorshtein A, Praiss M, Kendler E, et al. The rapamycinderivative RAD001 (everolimus) inhibits cell viability and interacts with the Akt-mTOR-p70S6K pathway in human medullary thyroid carcinoma cells. Mol Cell Endocrinol 2010; 315:87e94. 61. Guigon CJ, Fozzatti L, Lu C, Willingham MC, Cheng SY. Inhibition of mTORC1 signaling reduces tumor growth but does not prevent cancer progression in a mouse model of thyroid cancer. Carcinogenesis 2010;31:1284e91. 62. Chiacchio S, Lorenzoni A, Boni G, Rubello D, Elisei R, Mariani G. Anaplastic thyroid cancer: prevalence, diagnosis and treatment. Minerva Endocrinol 2008;33:341e57. 63. Smallridge RC, Copland JA. Anaplastic thyroid carcinoma: pathogenesis and emerging therapies. Clin Oncol (R Coll Radiol) 2010;22:486e97. 64. Swaak-Kragten AT, de Wilt JH, Schmitz PI, Bontenbal M, Levendag PC. Multimodality treatment for anaplastic thyroid carcinomaetreatment outcome in 75 patients. Radiother Oncol 2009;92:100e4. 65. Troch M, Koperek O, Scheuba C, Dieckmann K, Hoffmann M, Niederle B, et al. High efficacy of concomitant treatment of undifferentiated (anaplastic) thyroid cancer with radiation and docetaxel. J Clin Endocrinol Metab 2010;95: E54e7. 66. Foote RL, Molina JR, Kasperbauer JL, Lloyd RV, McIver B, Morris JC, et al. Enhanced survival in locoregionally confined anaplastic thyroid carcinoma: a single-institution experience using aggressive multimodal therapy. Thyroid 2011;21:25e30. 67. Vrbic S, Pejcic I, Vrbic M, Filipovic S. Therapy of stage IV B anaplastic thyroid carcinoma: single institution experience. J BUON 2009;14:41e4. 68. Yau T, Lo CY, Epstein RJ, Lam AK, Wan KY, Lang BH. Treatment outcomes in anaplastic thyroid carcinoma: survival improvement in young patients with localized disease reated by combination of surgery and radiotherapy. Ann Surg Oncol 2008;15:2500e5. 69. Ha HT, Lee JS, Urba S, Koenig RJ, Sisson J, Giordano T, et al. A phase II study of imatinib in patients with advanced anaplastic thyroid cancer. Thyroid 2010;20:975e80.