Survey
* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project
Update on Recent Developments in the Therapy of Differentiated Thyroid Cancer Marcus Middendorp, MD, and Frank Grünwald, MD In the past decade, the management of differentiated thyroid carcinoma changed significantly and thus contributed to the improvement of the already favorable prognosis of this malignant disease. Surgical treatment techniques improved and the extent of initial surgery is more individualized. Radioiodine therapy is an essential part of therapeutic regimens in almost all cases, and the use of recombinant human thyroid-stimulating hormone has established for ablation of remnant tissue, treatment of iodine-positive cancer, and sensitive thyroglobulin measurement during follow-up. Risk stratification has become more important to plan treatment and follow-up individually, particularly to evaluate the need for thyroid-stimulating hormone suppression therapy. Especially for inoperable and radioiodine-negative thyroid carcinomas, novel treatment options such as tyrosine kinase inhibitor therapy have emerged. This article deals with the current options of optimal therapy regimens in differentiated thyroid carcinoma. Semin Nucl Med 40:145-152 © 2010 Elsevier Inc. All rights reserved. D ifferentiated thyroid carcinoma (DTC) is the most common endocrine cancer, representing about 1% of all malignancies. Excellent prognosis and long-term survival with a 10-year survival of more than 90% are the main causes for the lack of relevant prospective randomized controlled trials.1 Existing guidelines and consensus reports for DTC are mainly based on large cohort studies, well-designed retrospective studies, and experience of experts in this field.2 In the last decade, several new therapeutic aspects have arisen, some of them still being controversially debated (eg, the adequate therapy of papillary thyroid microcarcinomas [PTMC]).3 This review will focus on current issues in the therapy of DTC, giving an update on the initial surgical management, the reasonable use of radioiodine therapy (RIT) for ablation of remnant thyroid tissue and for the treatment of persistent or recurrent disease, the use of recombinant human thyroid-stimulating hormone (rhTSH), the role of external beam radiation therapy (EBRT), the extent of thyrotropin-suppressive therapy, and on options in radioiodinenegative thyroid cancer. Department of Nuclear Medicine, Johann Wolfgang Goethe University, Frankfurt/Main, Germany. Address reprint requests to Frank Grünwald, MD, Department of Nuclear Medicine, Johann Wolfgang Goethe University Medical School, TheodorStern-Kai 7, 60590, Frankfurt am Main, Germany. E-mail: gruenwald@ em.uni-frankfurt.de 0001-2998/10/$-see front matter © 2010 Elsevier Inc. All rights reserved. doi:10.1053/j.semnuclmed.2009.10.006 Current Approach to Initial Surgical Therapy of DTC Primary Tumor In view of the biological heterogeneity of DTC an individualized surgical approach, also considering the postoperative quality of life, is necessary. The extent of surgery is highly dependent on an accurate risk stratification of DTC, which is still not precisely defined. Which initial surgical intervention is chosen, lobectomy, near-total thyroidectomy, or total thyroidectomy, is mainly determined by preoperative information. In suspicious solitary thyroid nodules smaller than 4 cm, lobectomy is the recommended initial approach.4,5 Completion thyroidectomy is widely accepted in DTC larger than 2 cm but controversially discussed in T1 stage (Union Internationale Contre le Cancer/American Joint Committee on Cancer 2002).6,7 In case of a preoperatively known DTC, near-total and total thyroidectomy are mostly performed with few exceptions. PTMC without metastases and invasion of the tumor capsule, with unifocal growth and favorable histology are discussed as indications for less-than-total thyroidectomy. Retrospective data could not show hitherto an advantage for total thyroidectomy with respect to overall survival.8 Some surgeons decide in favor of near-total or total thyroidectomy in case of preoperative diagnosis of PTMC, and on less-thantotal thyroidectomy in incidental finding of PTMC in case of lobectomy for benign thyroid disease. Completion thyroid145 M. Middendorp and F. Grünwald 146 ectomy and total thyroidectomy, respectively, are recommended in papillary thyroid carcinoma (PTC) larger than 1 cm, in case of multifocal disease, extrathyroidal invasion, unfavorable histology, or lymph node metastases.9 Some centers always perform near-total or total thyroidectomy arguing with frequent multifocality of PTC resulting in higher recurrence rates.10,11 In follicular thyroid carcinoma (FTC), extrathyroidal invasion and distant metastases are the most relevant prognostic factors.12 Follicular microcarcinomas are rather rare. In nonmetastatic minimally invasive FTC without extracapsular extension, less-than-total thyroidectomy is considered as an adequate therapy. However, in most cases total thyroidectomy is preferred.13 This is attributed to controversial definitions of minimal invasiveness and the suggestion, despite limited evidence that vascular invasion has a major impact on the outcome of FTC.14,15 In childhood DTC—mainly PTC—is often associated with locally aggressive disease and more frequent with distant metastases. With respect to the comparably unfavorable relation of tumor extension to thyroid volume, total thyroidectomy and lymph node dissection are suggested to be the optimal surgical strategy in most cases.16-18 Whether there is an appropriate threshold diameter, below which less-than-total thyroidectomy is adequate, will have to be evaluated in prospective multicenter trials. What extent of surgical treatment is necessary in more aggressive variants of thyroid cancer with intermediate differentiation such as tall cell variant, columnar cell variant, diffuse sclerosing variant, insular carcinoma, and Hürthle cell carcinoma? Even though there are only limited data for these histologic subtypes, evidence-based recommendation mostly includes total thyroidectomy and neck dissection for node positivity.19 In poorly differentiated thyroid cancer, lessthan-total thyroidectomy is not recommended.20 Lymph Node Metastases As overall effect of lymph node metastases on survival is limited, there are controversial opinions concerning the indication in general (therapeutic/diagnostic/prophylactic purposes), and the extent of lymph node dissection. For PTC less than 1 cm, no data exist justifying routine lymph node dissection.21 But PTC up to 10 mm is already associated with lymph node metastases in about 16%, and therefore prophylactic lymph node dissection of bilateral central compartments is mostly recommended for all PTC inclusive of microcarcinomas. Cervical lymph node dissection of the lateral compartments is considered in case of evident lymph node metastases in preoperative clinical diagnostics or during operation.5,20,22 In FTC the threshold size for lymph node metastases is greater (⬎ 20 mm) compared to PTC (⬍ 10 mm). As lymph node metastases are infrequent in FTC, routine lymph node dissection is not needed, unless there is confirmation of lymph node metastases. Available data on poorly DTC do not support routine lymph node dissection as well except for extrathyroidal primary tumor extension.20,22 Reasonable Uses of Radioiodine Therapy Radioiodine Ablation Postoperative RIT aims at destroying of residual microscopic tumor tissue to reduce the recurrence rate, ablating remaining normal thyroid tissue to optimize follow-up conditions (eg, thyroglobulin measurement), and completing staging by a highly sensitive post-therapeutic whole-body scintigraphy (WBS). The guidelines for the management of differentiated thyroid cancer published by the American Thyroid Association (ATA), the European Association of Nuclear Medicine (EANM), and the European Thyroid Association (ETA) agree in the indication of RIT in high-risk patients and in the opinion that RIT is not necessary in unifocal thyroid microcarcinomas without metastases, capsule invasion, and lack of more aggressive histologies such as tall cell, columnar cell, and diffuse sclerosing variant, the so-called very-low-risk group.5,23,24 Due to the less precisely defined low-risk group there are controversies about the adequate management of these patients. Whatever risk stratification is applied, tumor diameter, AMES (Age, distant Metastasis, Extent, and Size of primary cancers), or MACIS (Metastasis, Age, Completeness of resection, Invasion, and Size) score, most low-risk patients have excellent prognosis, and thus there remain doubts about the benefit of RIT. Prospective controlled randomized trials with an endpoint of specific mortality are rarely realizable.25-29 As no advantage in specific mortality and recurrence rate after RIT of low-risk patients has been proved, there are significant regional differences in recommendations of postoperative RIT.27,30 ATA recommends RIT for ablation of remnants in stages III and IV, in most patients with stage II, and in selected cases of stage I (American Joint Committee on Cancer).5 ETA defines probable indications for RIT in the following circumstances: less-than-total thyroidectomy, no lymph node dissection, age below 18 years, T1 ⬎ 1 cm and T2N0M0, or more aggressive histology.23 However, RIT is often performed to enable a more comfortable follow-up and with the assumption of lower recurrence rates. Even though the effect of RIT on low-risk patients is discussed controversially, RIT is an effective therapy to eradicate small residual tumors and metastases of DTC, which already occur in microcarcinomas. Thus, RIT should be indicated routinely with only few exceptions. RIT activities are usually determined empirically and given as a single administration of 1.11 GBq (30 mCi) to 5.5 GBq (150 mCi). Up to now the most appropriate dose cannot be derived from evidence-based data.24,31 In children and adolescents, activities are set in consideration of body weight, body surface, age, or 24-h thyroid bed uptake of an 131I test activity.32 An alternative to fixed activities is an individualized activity based on pretherapeutic remnant- and lesionbased or bone marrow dosimetry. For this, the EANM Dosimetry Committee recently published a standard operation procedure. Generally, accepted, absorbed tumor Developments in therapy of differentiated thyroid cancer 147 doses should exceed 80 Gy and absorbed dose to blood should be below 2 Gy.33,34 Radioiodine Treatment of Persistent or Recurrent Disease In potentially curable patients with persistent or recurrent disease of DTC, surgical treatment is mostly considered at first. Radioiodine therapy (RIT) has proved to be an effective alternative or adjuvant therapy option for tumor eradication, deceleration of disease progress, or palliation of symptoms.35 The benefit of RIT is crucially dependent on factors such as radioiodine avidity of the tumor cells, tumor localization, histology, patient age, and individual health state.24,35 According to EANM guidelines, nonresectable lymph node metastases, pulmonary micrometastases, nonresectable or incompletely operable macrometastases of the lungs, and nonresectable soft-tissue metastases are definitive indications for RIT. Further, optional fields of application are recurrent radioiodine-positive lymph node and distant metastases (adjuvant role), nonresectable small and/or multiple lymph node metastases, inoperable bone metastases, known or assumed metastases with unknown radioiodine avidity, and anaplastic or poorly differentiated thyroid cancer with relevant well-differentiated parts and thyroglobulin (Tg) expression, respectively.24 In locoregional relapse and tumor infiltration of the upper airways, adjuvant RIT is performed after surgery in case of residual tumor tissue—if necessary, in combination with EBRT. RIT for lung metastases, mostly with empirically determined 131I activities between 3.7 GBq (100 mCi) and 11.1 GBq (300 mCi), can be repeated as often as there is objective therapy response or until complete remission (CR) or serious side effects such as lung fibrosis occur.5 Overall, if only lung metastases exist—particularly if they are small and radioiodine-positive—the prognosis is favorable and better than in any other malignant disease with lung metastases (Fig. 1).36 Therapy management of bone metastases is influenced by localization, fracture risk, pain, and radioiodine avidity. Inoperable bone metastases are treated with EBRT, RIT, intra-arterial embolization, radiofrequency ablation, bone-seeking radiopharmaceuticals, and bisphosphonates. RIT of 131I storing bone metastases with most often fixed 131I activities between 5.5 GBq (150 mCi) and 11.1 GBq (300 mCi) is associated with improved overall survival, even though CR is rarely achieved. In fracture risk and risk of neurological complications, RIT or EBRT are frequently conducted under medication with corticosteroids to minimize TSH-induced and/or radiation-associated tumor growth or swelling.37,38 Irrespective of radioiodine avidity, preferred treatment options for brain metastases are the complete surgical resection and EBRT.39 Radioiodine can also be administered in radioiodine-scan-negative cases, supposing only microscopic sites of radiopharmaceutical uptake, justified by post-therapeutic thyroglobulin decrease.40 The adequate method to determine the 131I activities remains unclear. Which method, empirically or on the basis of blood/whole-body dosimetry or tumor dosimetry, is superior to the others, cannot be assessed conclusively Figure 1 Case of a 15-year-old female patient with advanced papillary thyroid carcinoma (T3N1bM1). Initially, thyroidectomy and bilateral selective neck dissection were performed, followed by 2 radioiodine administrations. Computed tomography of the lungs before RIT (A) with 1 pulmonary nodule visible on this slice (white arrow). Anterior views of post-therapeutic whole-body scintigraphies (WBS) after the first (B) and second (C) radioiodine administration demonstrate CR of the multiple iodine-positive lung metastases. Additionally, skin contaminations can be observed in both WBS images. at present. Taking 200 cGy or rad as a basis for a maximum tolerable doses of the blood/red bone marrow, empirically set 131I activities between 3.7 GBq (100 mCi) and 11.1 GBq (300 mCi) led to an exceeding of the maximum tolerable doses in up to 22%. By contrast, most patients could have been treated with higher 131I activities.41 Apart from 148 higher 131I activities, an increase of the tumor dose (about the 2.3-fold) can be achieved by pharmacotherapy with lithium. Intake of lithium results in higher 131I retention in normal thyrocytes and thyroid carcinoma cells. As there are no trials, which have analyzed the outcome after lithium therapy, a strong recommendation for or against the use of lithium is not yet possible.42 Extended Fields of Application for rhTSH In the USA as well as in Europe, rhTSH was first licensed for the stimulated Tg measurement with or without radioiodine imaging and proved its value in the follow-up of DTC for years.43 Later on, rhTSH was approved for the postoperative radioiodine ablation in DTC (Europe 2005, USA 2007). Its use in children and adolescents is not established yet. Several trials demonstrated comparable radioiodine ablation rates after administering rhTSH or after withdrawal of thyroid hormone (WTH), but quality of life was higher after use of rhTSH. In an international randomized controlled trial ablation rates were 100% for both groups by the criterion of visible 131I uptake in the thyroid bed (3.7 GBq (100 mCi)). Following rhTSH application, 131I blood dose was one-third lower compared to WTH. Interestingly, patients treated with rhTSH had a longer effective radioisotope half-time within the remnant thyroid tissue.44-47 Retrospective data analysis of patients with DTC, who recieved postoperative RIT after either rhTSH injection or WTH, revealed similar recurrence rates after a median follow-up period of 2.5 years.48 The questions of adequate 131I activity for ablative RIT, 1.1 GBq (30 mCi), 1.85 GBq (50 mCi) or 3.7 GBq (100 mCi), and of comparable therapeutic effectivity in high-risk and low-risk patients are still controversially discussed. A recent multicenter study demonstrated that 1.85 and 3.7 GBq 131I leads to similar ablation rates. Seventy-two patients were randomly assigned to receive one of these activities after rhTSH administration. In each group, 88.9% of patients had no visible 131I uptake in the diagnostic WBS 6-8 months later.49,50 The benefit of rhTSH for RIT of persistent or metastatic disease has not been proven in prospective randomized controlled trials yet, even though several groups have dealt with this issue. Current data are still insufficient to recommend rhTSH for routine use in RIT of these patients. In certain patient collectives such as those with hypopituitarism and relevant comorbidities, in which hypothyroidism after WTH would mean an increased risk to the patient, TSH stimulation by rhTSH is reasonable and justifiable.49,51 In a retrospective study, Robbins et al52 showed that in 112 patients, who could not elevate endogenous thyrotropin or be withdrawn from thyroxine, TSH levels above 25 mU/mL could be achieved. Earlier experienced hypothyroid complications occurred less frequently. Further potential indications for rhTSH mainly regard diagnostic procedures such as the 18F-fluorodeoxyglucose-positron emission tomography for increasing sensitivity.46 M. Middendorp and F. Grünwald External Beam Radiation Therapy EBRT plays an adjuvant role in certain patients. For adequate patient selection, tumor histology, 131I WBS, and 18F-fluorodeoxyglucose-positron emission tomography have to be considered. Generally, accepted indications for EBRT are gross residual tumor masses, which cannot effectively be eradicated by surgery or RIT, older patients with microscopic residual tumor after radical resection with high-risk of recurrence and assumed low response to RIT, inoperable and radioiodine-negative relapse, inoperable brain metastases, and painful bone metastases with or without risk of neurological complications or fracture. In TNM stages pT1-3 N0M0 no advantage may be expected after EBRT. In nodal-positive patients and unfavorable prognostic factors such as poor differentiation, adjuvant EBRT can be indicated in particular cases without the proof of benefit in study literature. The use of EBRT in TNM stages pT4pN0/1/x M0 R0 is debatable with apparently low acceptance for this purpose.5,23,53-55 Schwartz et al56 presented retrospectively collected data of 131 patients with locally advanced DTC, partly with gross residual partly with microscopic tumor disease, who were treated with conformal EBRT or intensity-modulated radiotherapy. Four years after receiving a median dose of, 60 Gy (range 38-72 Gy) local relapse-free survival was 79%, disease free survival 76%, and overall survival 73%. The use of intensity-modulated radiotherapy (44%) compared to conformal EBRT was not associated with improved survival but with less frequent severe late morbidity (12% vs. 2%). Thyrotropin-Suppressive Therapy As shown in extensive preclinical and clinical studies, DTC cells express the TSH receptor on the cell membrane and react to TSH stimulation with production of specific proteins and cell proliferation. In addition, a direct triiodothyronine mediated stimulating effect on cellular growth is discussed. Herefrom derives the rationale for TSH-suppressive therapy as a therapeutic strategy of DTC. However, for low-risk patients no benefit in the outcome has been proven hitherto. Furthermore, TSH-independent proliferation of poorly DTC is hardly slowed down hereby.57-59 The indication for longterm TSH suppression should be evaluated critically, especially considering the negative impact on bone metabolism, for example, the increased risk of osteoporosis in postmenopausal women, and potential adverse cardiac events.60,61 Recommendations on adequate risk-adapted TSH ranges remarkably vary between the guidelines and authors. By the time of CR, ETA suggests TSH suppression irrespective of the risk stratification, whereas ATA recommends a TSH range of 0.1-0.5 mU/L for low-risk patients and a TSH lower than 0.1 mU/L for high-risk patients and those with persistent disease. After achievement of CR, ETA and ATA approve TSH values of 0.5-1.0 mU/L and 0.3-2.0 mU/L, respectively, for low-risk patients. In the high-risk-group, ETA guidelines schedule the continuation of TSH suppression for 3-5 years and the ATA guidelines TSH values between 0.1 and 0.5 mU/L for 5-10 years.5,23 There are several other recommendations with Developments in therapy of differentiated thyroid cancer varying TSH ranges, for example, those published by Tuttle et al62 suggesting a general TSH range of 0.1-0.4 mU/L, except for high-risk patients (⬍ 0.1 mU/L) and for the verylow-risk group (⬍ 1.0 mU/L). Finally, risk stratification has to be regarded as a dynamic process, which affects the extent of thyroxine therapy. Options in Inoperable Radioiodine-Negative DTC In particular, in the management of advanced and radioiodine-negative DTC, which may not be treatable with surgery, RIT, or EBRT, numerous novel therapeutic approaches have developed in the last decade. The spectrum includes the redifferentiation of thyroid carcinoma tissue, the inhibition of cell signaling, for example, the mitogen-activated protein kinase pathway, and of angiogenesis, gene therapy, and chemotherapy. Chemotherapy and Redifferentiation of DTC In chemotherapy of advanced and radioiodine-negative DTC, doxorubicin is mostly the substance of choice for monotherapy or in combination with other cytotoxic pharmaceuticals. The combination has not yet shown a benefit over monotherapy. Response rates are usually much below 40% with a time to progression of only a few months. Comparably, promising results with a response rate of 37% and a median overall survival of 21 months from the beginning of chemotherapy were achieved by a combination of epirubicin and carboplatinum under TSH stimulation. To what extent TSH elevation influenced the chemotherapy effect, has to be investigated in further studies.63 Chemotherapy of DTC is generally recommended in progressive disease (PD) after exhausting all surgical treatment options, RIT, and EBRT as well as considering available novel therapies within trials.5,64-66 Many groups have dealt with inducing radioiodine uptake in radioiodine-negative DTC. For this, glitazones and retinoid derivates are used. In the latter, signals are transduced by specific nuclear receptors, the retinoid-X receptors, which act as transcription factors modulating the activity of retinoic acid-responsive genes.67 After intake of 13-cis-retinoic acid (1.5 mg/kg body weight) for 6 weeks, Simon et al68 attained a moderate or intensive radioiodine uptake in 23 of 50 patients with PTC, FTC, or Hürthle cell tumors and with afore predominantly negative posttherapeutic 131I WBS. In addition to an indirect antitumor effect by increased radioiodine uptake, a direct antiproliferative effect is discussed. With regard to radioiodine uptake and tumor diameter, objective therapy response was seen in 10 of 50 patients (20%), stable disease (SD) in 9 of 50 (18%), and PD in 31 of 50 (62%) patients. In a small series of 11 patients69 with progressive, metastatic PTC and FTC increasing 131I uptake was visible in only 2 cases after 8 weeks of medication with 13-cis-retinoic acid. Authors concluded that redifferentiation therapy has no benefit in aggressive advanced PTC and FTC. Preliminary data with comparable results exist for redifferentiation 149 mediated by all-trans-retinoic acid.70 Another study dealt with the induction of radioiodine uptake by a peroxisome proliferator-activated receptor-gamma (PPAR-␥) agonist. Taking rosiglitazone for 6 weeks (8 mg/d), new tumorassociated 131I storage was detected by posttherapeutic and/or diagnostic 131I WBS in 6 of 23 patients (26%) with elevated thyroglobulin (⬎ 10 ng/ml). Five of these showed intense immunohistochemical staining for PPAR-␥.71 Inhibition of Cellular Signaling and Angiogenesis In the development of PTC rearrangements of the rearranged during transfection (RET) oncogene (approximately 30%) and v-raf murine sarcoma viral oncogene homolog B1 (BRAF) point mutations (approximately 40%) are particularly involved. In contrast, genetic alterations of FTC are primarily made up of PAX8-PPAR␥ translocations (25%-60%) and RAS mutations (40%-50%).72 Further, abnormalities of DTC cells regard the overexpression of tyrosine kinase receptors, including those for the vascular endothelial growth factor (VEGF), the epidermal growth factor (EGF), and the insulin growth factor-1. The basis for molecular targeted therapy of the DTC is the inhibition of the mitogen-activated protein kinase and as well the phosphatidylinositol 3-kinase (PI3K) pathway, and of angiogenesis by kinase inhibitors, monoclonal antibodies, antisense oligonucleotides, and the farnesyl transferase inhibitor. Small-molecule kinase inhibitors can be divided in those, which interfere with the adenosine triphosphate-binding domain, from those, which have targets outside this area, for example, at the substrate- or ligand-binding domain. Such therapies are only reasonable for subgroup of tumors, which highly express the corresponding target. Thus, pretherapeutic immunohistochemical screening of the specific target will become more and more important. It should be expected that therapy considering several molecular targets and combination with other treatment options will increase response rates.73,74 Following phase II trials exemplarily represent the molecular targeted therapy of the advanced, inoperable, and radioiodine-negative thyroid carcinoma, mainly PTC and FTC. The first study examined the outcome after therapy with the multikinase inhibitor sorafenib (molecular targets: BRAF, vraf-1 murine leukemia viral oncogene homolog 1 [CRAF], vascular endothelial growth factor receptor [VEGFR], RET, and platelet-derived growth factor receptor [PDGFR]) in 30 patients (27 with PTC and FTC), of whom 25 were evaluable. Comprising all histologies, partial response (PR) was achieved in 23% of patients (7/30), SD in 53% of patients (16/30), and PD occurred in 7% of patients (2/30). Median progression free survival (PFS) was 84 weeks for PTC and FTC. There were only grade 1 and 2 adverse events. In one case liver failure resulted in death. Significantly improved overall response rate and PFS were shown compared to chemotherapy.75 Sherman et al76 treated 93 patients with thyroid cancer (82 analyzable) with the multikinase inhibitor motesanib diphosphate (molecular targets: VEGFR, PDGFR, and cellular homolog of the feline sarcoma viral oncogene v-kit [C-KIT]) M. Middendorp and F. Grünwald 150 and demonstrated the following results: PR in 13 of 93 patients (14%), SD in 62 of 93 (67%), and PD in 7 of 93 (8%) cases. Stable disease outlasted 24 weeks in 33 of 93 patients (35%). The estimated PFS was 40 weeks. In a third study on 60 patients (36 with PTC and FTC evaluable) treated with the selective VEGFR inhibitor axitinib, PR was observed in 30% (18/60), SD in 38% (23/60), PD in 7% (4/60)—the median PFS was 18.1 months.77 Pennell et al78 evaluated the therapy effect of the EGFR inhibitor gefitinib in 27 patients with thyroid cancer, 17 of them with PTC and FTC (63%). Neither CR nor PR was observed. The rates for SD after 3, 6, and 12 months were 48%, 24%, and 12%, respectively, for all histologies. For DTC, a median PFS of 3.9 months and an overall survival of 27.4 months were determined. Gene Therapy Another future treatment option is gene therapy, which can be summarized in corrective (eg, p53 restoration), cytoreductive (eg, sodium-iodide symporter [NIS] gene transfer followed by 131I therapy), and immunomodulatory gene therapies (eg, IL-2 and IL-12 gene transfer). Especially NIS gene therapy proved as a promising approach in preclinical studies. However, due to lack of integration into thyroid hormone synthesis, NIS gene transfer led to an only temporarily increased radioiodine uptake of infected tumor cells (in vitro), and xenografted tumors (in vivo) with consequently low tumor dose due to a short residence time despite initial high uptake values. Up to now one major problem has been the inefficiency and lack of safety of the available vector systems, limited by factors such as a high first-pass effect in the liver, neutralization by host immunity, and low infectivity. In the future, replication-selective viral vectors and biodegradable polymers could be suitable vector systems. Existing data reveal that gene therapy will only be used in a multimodality treatment approach for the foreseeable future.73,79 Other Treatment Options Medication or targeted radiotherapy with somatostatin analogues are other potential treatment options. Intramuscular injection of long-acting octreotide for 6 months in 8 patients with progressive, advanced, and radioiodine-negative but somatostatin receptor-positive thyroid cancer did not result in any objective therapy response, neither PR nor SD.80 For peptide receptor radionuclide therapy (PRRT) of radioiodine-negative DTC there are numerous studies with small case numbers, which have been summarized by Teunissen et al81 in this review objective therapy response was reported in 5 of 62 of patients (8%) and SD in 42% with a time to progression of less than 12 months (not available for all patients). In particular, for the often somatostatin receptor-overexpressing Hürthle cell tumors, PRRT is an option that has to be evaluated in prospective trials. However, treatment effect seems to be worse in DTC compared to gastroenteropancreatic neuroendocrine tumors. In a monocenter trial on yttrium-90 DOTA-TOC ((DOTA(0)-Phe(1)-Tyr(3))octreotid) therapy in 24 patients with radioiodine-negative DTC, 7 patients had a decrease in thyroglobulin of 48.9% (range, 0.3%-54.5%) and 17 had an increase of 36.8% (range, 2.6%-4912.8%). Me- dian overall survival from the beginning of PRRT was 42.4 months for responders and 13.7 months for nonresponders.82 Cyclo-oxygenase 2 is often overexpressed in malignant tumors as in DTC. In a prospective phase II trial, treatment of progressive metastatic DTC with the selective Cyclo-oxygenase 2 inhibitor celecoxib resulted in an objective response in only 1 of 32 patients (3%). Applying RECIST criteria, 12 of 32 cases achieved an SD. Twenty-three of 32 patients (38%) stopped participation in this study due to PD or toxicity. Thus, Mrozek et al83 assessed that celecoxib has no benefit in most patients with advanced DTC. References 1. Stokkel MP, Duchateau CS, Dragoiescu C: The value of FDG-PET in the follow-up of differentiated thyroid cancer: A review of the literature. Q J Nucl Med Mol Imaging 50 78-87, 2006 2. Mallick UK, Charalambous H: Current issues in the management of differentiated thyroid cancer. Nucl Med Commun 25:873-881, 2004 3. Biersack HJ, Grünwald F: Thyroid Cancer (ed 2). New York, Springer, 2005 4. Sclabas GM, Staerkel GA, Shapiro SE, et al: Fine-needle aspiration of the thyroid and correlation with histopathology in a contemporary series of 240 patients. Am J Surg 186:702-709, 2003 5. Cooper DS, Doherty GM, Haugen BR, et al: Management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 16:109-142, 2006 6. Passler C, Scheuba C, Asari R, et al: Importance of tumour size in papillary and follicular thyroid cancer. Br J Surg 92:184-189, 2005 7. Shaha A: Selective surgical management of well-differentiated thyroid cancer. Ann NY Acad Sci 1138:58-64, 2008 8. Bilimoria KY, Bentrem DJ, Ko CY, et al: Extent of surgery affects survival for papillary thyroid cancer. Ann Surg 246:375-381, 2007 9. Pazaitou-Panayiotou K, Capezzone M, Pacini F: Clinical features and therapeutic implication of papillary thyroid microcarcinoma. Thyroid 17:1085-1092, 2007 10. Sakorafas GH, Giotakis J, Stafyla V: Papillary thyroid microcarcinoma: A surgical perspective. Cancer Treat Rev 31:423-438, 2005 11. Hay ID, Hutchinson ME, Gonzalez-Losada T, et al: Papillary thyroid microcarcinoma: A study of 900 cases observed in a 60-year period. Surgery 144:980-987, 2008 12. D’Avanzo A, Treseler P, Ituarte PH, et al: Follicular thyroid carcinoma: Histology and prognosis. Cancer 100:1123-1129, 2004 13. Delbridge L, Parkyn R, Philips J, et al: Minimally invasive follicular thyroid carcinoma: Completion thyroidectomy or not? ANZ J Surg 72:844-845, 2002 14. Brennan MD, Bergstralh EJ, van Heerden JA, et al: Follicular thyroid cancer treated at the Mayo Clinic, 1946 through 1970: Initial manifestations, pathologic findings, therapy, and outcome. Mayo Clin Proc 66:11-22, 1991 15. Furlan JC, Bedard YC, Rosen IB: Clinicopathologic significance of histologic vascular invasion in papillary and follicular thyroid carcinomas. J Am Coll Surg 198:341-348, 2004 16. Demidchik YE, Demidchik EP, Reiners C, et al: Comprehensive clinical assessment of 740 cases of surgically treated thyroid cancer in children of Belarus. Ann Surg 243:525-532, 2006 17. Thompson GB, Hay ID: Current strategies for surgical management and adjuvant treatment of childhood papillary thyroid carcinoma. World J Surg 28:1187-1198, 2004 18. Gingalewski CA, Newman KD: Seminars: Controversies in the management of pediatric thyroid malignancy. J Surg Oncol 94:748-752, 2006 19. Carling T, Ocal IT, Udelsman R: Special variants of differentiated thyroid cancer: Does it alter the extent of surgery versus well-differentiated thyroid cancer? World J Surg 31:916-923, 2007 20. Dralle H, Machens A: Surgical approaches in thyroid cancer and lymph-node metastases. Best Pract Res Clin Endocrinol Metab 22:971987, 2008 Developments in therapy of differentiated thyroid cancer 21. Ito Y, Jikuzono T, Higashiyama T, et al: Clinical significance of lymph node metastasis of thyroid papillary carcinoma located in one lobe. World J Surg 30:1821-1828, 2006 22. Vorländer C, Lienenlüke RH, Wahl RA: Lymph node dissection in papillary and follicular thyroid cancer. Chirurg 79:564-570, 2008 23. Pacini F, Schlumberger M, Dralle H, et al: European consensus for the management of patients with differentiated thyroid carcinoma of the follicular epithelium. Eur J Endocrinol 154:787-803, 2006 24. Luster M, Clarke SE, Dietlein M, et al: Guidelines for radioiodine therapy of differentiated thyroid cancer. Eur J Nucl Med Mol Imaging 35:1941-1959, 2008 25. Sawka AM, Thephamongkhol K, Brouwers M, et al: Clinical review 170: A systematic review and metaanalysis of the effectiveness of radioactive iodine remnant ablation for well-differentiated thyroid cancer. J Clin Endocrinol Metab 89:3668-3676, 2004 26. Hay ID, Grant CS, Bergstralh EJ, et al: Unilateral total lobectomy: Is it sufficient surgical treatment for patients with AMES low-risk papillary thyroid carcinoma? Surgery 124:958-964, 1998 27. Hay ID, McConahey WM, Goellner JR: Managing patients with papillary thyroid carcinoma: Insights gained from the Mayo Clinic’s experience of treating 2,512 consecutive patients during 1940 through 2000. Trans Am Clin Climatol Assoc 113:241-260, 2002 28. Hay ID: Selective use of radioactive iodine in the postoperative management of patients with papillary and follicular thyroid carcinoma. J Surg Oncol 94:692-700, 2006 29. Mazzaferri EL: A randomized trial of remnant ablation—In search of an impossible dream? J Clin Endocrinol Metab 89:3662-3664, 2004 30. Sawka AM, Rotstein L, Brierley JD, et al: Regional differences in opinions on adjuvant radioactive iodine treatment of thyroid carcinoma within Canada and the United States. Thyroid 17:1235-1242, 2007 31. Hackshaw A, Harmer C, Mallick U, et al: 131I activity for remnant ablation in patients with differentiated thyroid cancer: A systematic review. J Clin Endocrinol Metab 92:28-38, 2007 32. Jarzab B, Handkiewicz-Junak D, Wloch J, et al: Juvenile differentiated thyroid carcinoma and the role of radioiodine in its treatment: A qualitative review. Endocr Relat Cancer 12:773-803, 2005 33. Maxon HR 3rd, Englaro EE, Thomas SR, et al: Radioiodine-131 therapy for well-differentiated thyroid cancer—A quantitative radiation dosimetric approach: Outcome and validation in 85 patients. J Nucl Med 33:1132-1136, 1992 34. Lassmann M, Hänscheid H, Chiesa C, et al: EANM Dosimetry Committee: Series on standard operational procedures for pre-therapeutic dosimetry I: Blood and bone marrow dosimetry in differentiated thyroid cancer therapy. Eur J Nucl Med Mol Imaging 35:1405-1412, 2008 35. 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 91:2892-2899, 2006 36. Menzel C, Grünwald F, Schomburg A, et al: “High-dose” radioiodine therapy in advanced differentiated thyroid carcinoma. J Nucl Med 37: 1496-1503, 1996 37. Bernier MO, Leenhardt L, Hoang C, et al: Survival and therapeutic modalities in patients with bone metastases of differentiated thyroid carcinomas. J Clin Endocrinol Metab 86:1568-1573, 2001 38. Muresan MM, Olivier P, Leclère J, et al: Bone metastases from differentiated thyroid carcinoma. Endocr Relat Cancer 15:37-49, 2008 39. McWilliams RR, Giannini C, Hay ID, et al: Management of brain metastases from thyroid carcinoma: A study of 16 pathologically confirmed cases over 25 years. Cancer 98:356-362, 2003 40. Menzel C, Zaplatnikov K, Diehl M, et al: The influence of thyroglobulin on functional imaging in differentiated thyroid cancer. Nucl Med Commun 25:239-243, 2004 41. Kulkarni K, Van Nostrand D, Atkins F, et al: The relative frequency in which empiric dosages of radioiodine would potentially overtreat or undertreat patients who have metastatic well-differentiated thyroid cancer. Thyroid 16:1019-1023, 2006 42. Koong SS, Reynolds JC, Movius EG, et al: Lithium as a potential adjuvant to 131I therapy of metastatic, well differentiated thyroid carcinoma. J Clin Endocrinol Metab 84:912-916, 1999 151 43. Sugino K, Ito K, Takami H: Management of differentiated thyroid carcinoma with radioiodine and recombinant human TSH. Endocr J 53: 723-728, 2006 44. Pacini F, Ladenson PW, Schlumberger M, et al: Radioiodine ablation of thyroid remnants after preparation with recombinant human thyrotropin in differentiated thyroid carcinoma: Results of an international, randomized, controlled study. J Clin Endocrinol Metab 91:926-932, 2006 45. Hänscheid H, Lassmann M, Luster M, et al: Iodine biokinetics and dosimetry in radioiodine therapy of thyroid cancer: Procedures and results of a prospective international controlled study of ablation after rhTSH or hormone withdrawal. J Nucl Med 47:648-654, 2006 46. Duntas LH, Cooper DS: Review on the occasion of a decade of recombinant human TSH: Prospects and novel uses. Thyroid 18:509-516, 2008 47. Intenzo CM, Jabbour S, Dam HQ, et al: Changing concepts in the management of differentiated thyroid cancer. Semin Nucl Med 35:257265, 2005 48. Tuttle RM, Brokhin M, Omry G, et al: Recombinant human TSH-assisted radioactive iodine remnant ablation achieves short-term clinical recurrence rates similar to those of traditional thyroid hormone withdrawal. J Nucl Med 49:764-770, 2008 49. Haugen BR, Cooper DS, Emerson CH, et al: Expanding indications for recombinant human TSH in thyroid cancer. Thyroid 18:687-694, 2008 50. Pilli T, Brianzoni E, Capoccetti F, et al: A comparison of 1850 (50 mCi) and 3700 MBq (100 mCi) 131I administered doses for recombinant thyrotropin-stimulated postoperative thyroid remnant ablation in differentiated thyroid cancer. J Clin Endocrinol Metab 92:3542-3546, 2007 51. Risse JH, Grünwald F, Bender H, et al: Recombinant human thyrotropin in thyroid cancer and hypopituitarism due to sella metastasis. Thyroid 9:1253-1256, 1999 52. Robbins RJ, Driedger A, Magner J: Recombinant human thyrotropinassisted radioiodine therapy for patients with metastatic thyroid cancer who could not elevate endogenous thyrotropin or be withdrawn from thyroxine. Thyroid 16:1121-1130, 2006 53. Lee N, Tuttle M: The role of external beam radiotherapy in the treatment of papillary thyroid cancer. Endocr Relat Cancer 13:971-977, 2006 54. Schuck A, Biermann M, Schober O, et al: Strahlentherapie des Schilddrüsenkarzinoms. [German]. Onkologe 11:87-92, 2005 55. Biermann M, Pixberg MK, Schuck A, et al: Multicenter study differentiated thyroid carcinoma (MSDS). Diminished acceptance of adjuvant external beam radiotherapy. Nuklearmedizin 42:244-250, 2003 56. Schwartz DL, Lobo MJ, Ang KK, et al: Postoperative external beam radiotherapy for differentiated thyroid cancer: Outcomes and morbidity with conformal treatment. Int J Radiat Oncol Biol Phys 74:10831091, 2009 57. Biondi B, Filetti S, Schlumberger M: Thyroid-hormone therapy and thyroid cancer: A reassessment. Nat Clin Pract Endocrinol Metab 1:3240, 2005 58. Brabant G: Thyrotropin suppressive therapy in thyroid carcinoma: What are the targets? J Clin Endocrinol Metab 93:1167-1169, 2008 59. Cooper DS, Specker B, Ho M, et al: Thyrotropin suppression and disease progression in patients with differentiated thyroid cancer: Results from the National Thyroid Cancer Treatment Cooperative Registry. Thyroid 8:737-744, 1998 60. Heemstra KA, Hamdy NA, Romijn JA, et al: The effects of thyrotropinsuppressive therapy on bone metabolism in patients with well-differentiated thyroid carcinoma. Thyroid 16:583-591, 2006 61. Biondi B, Cooper DS: The clinical significance of subclinical thyroid dysfunction. Endocr Rev 29:76-131, 2008 62. Tuttle RM, Leboeuf R, Shaha AR: Medical management of thyroid cancer: A risk adapted approach. J Surg Oncol 97:712-716, 2008 63. Santini F, Bottici V, Elisei R, et al: Cytotoxic effects of carboplatinum and epirubicin in the setting of an elevated serum thyrotropin for advanced poorly differentiated thyroid cancer. J Clin Endocrinol Metab 87:4160-4165, 2002 64. Williams SD, Birch R, Einhorn LH: Phase II evaluation of doxorubicin M. Middendorp and F. Grünwald 152 65. 66. 67. 68. 69. 70. 71. 72. 73. plus cisplatin in advanced thyroid cancer: A Southeastern Cancer Study Group trial. Cancer Treat Rep 70:405-407, 1986 Raue F: Chemotherapy in thyroid cancer: Indications and results [German]. Onkologe 3:55-58, 1997 Haugen BR: Management of the patient with progressive radioiodine non-responsive disease. Semin Surg Oncol 16:34-41, 1999 Grünwald F, Menzel C, Bender H, et al: Redifferentiation therapyinduced radioiodine uptake in thyroid cancer. J Nucl Med 39:19031906, 1998 Simon D, Körber C, Krausch M, et al: Clinical impact of retinoids in redifferentiation therapy of advanced thyroid cancer: Final results of a pilot study. Eur J Nucl Med Mol Imaging 29:775-782, 2002 Courbon F, Zerdoud S, Bastie D, et al: Defective efficacy of retinoic acid treatment in patients with metastatic thyroid carcinoma. Thyroid 16: 1025-1031, 2006 Zhang Y, Jia S, Liu Y, et al: A clinical study of all-trans-retinoid-induced differentiation therapy of advanced thyroid cancer. Nucl Med Commun 28:251-255, 2007 Tepmongkol S, Keelawat S, Honsawek S, et al: Rosiglitazone effect on radioiodine uptake in thyroid carcinoma patients with high thyroglobulin but negative total body scan: A correlation with the expression of peroxisome proliferator-activated receptor-gamma. Thyroid 18:697704, 2008 DeLellis RA: Pathology and genetics of thyroid carcinoma. J Surg Oncol 94:662-669, 2006 Baudin E, Schlumberger M: New therapeutic approaches for metastatic thyroid carcinoma. Lancet Oncol 8:148-156, 2007 74. Coelho SM, Carvalho DP, Vaisman M: New perspectives on the treatment of differentiated thyroid cancer. Arq Bras Endocrinol Metabol 51:612-624, 2007 75. Gupta-Abramson V, Troxel AB, Nellore A, et al: Phase II trial of sorafenib in advanced thyroid cancer. J Clin Oncol 26:4714-4719, 2008 76. Sherman SI, Wirth LJ, Droz JP, et al: Motesanib diphosphate in progressive differentiated thyroid cancer. N Engl J Med 359:31-42, 2008 77. Cohen EE, Rosen LS, Vokes EE, et al: Axitinib is an active treatment for all histologic subtypes of advanced thyroid cancer: Results from a phase II study. J Clin Oncol 26:4708-4713, 2008 78. Pennell NA, Daniels GH, Haddad RI, et al: A phase II study of gefitinib in patients with advanced thyroid cancer. Thyroid 18:317-323, 2008 79. Spitzweg C: Gene therapy in thyroid cancer. Horm Metab Res 41:500509, 2009 80. Kohlfuerst S, Igerc I, Gallowitsch HJ, et al: Is there a role for sandostatin treatment in patients with progressive thyroid cancer and iodine-negative but somatostatin-receptor-positive metastases? Thyroid 16:11131119, 2006 81. Teunissen JJ, Kwekkeboom DJ, Krenning EP: Staging and treatment of differentiated thyroid carcinoma with radiolabeled somatostatin analogs. Trends Endocrinol Metab 17:19-25, 2006 82. Iten F, Muller B, Schindler C, et al: [(90)Yttrium-DOTA]-TOC response is associated with survival benefit in iodine-refractory thyroid cancer: Long-term results of a phase 2 clinical trial. Cancer 115:2052-2062, 2009 83. Mrozek E, Kloos RT, Ringel MD, et al: Phase II study of celecoxib in metastatic differentiated thyroid carcinoma. J Clin Endocrinol Metab 91:2201-2204, 2006