1
|
Silindir-Gunay M, Ozolmez N. Adverse reactions to therapeutic radiopharmaceuticals. Appl Radiat Isot 2024; 214:111527. [PMID: 39332267 DOI: 10.1016/j.apradiso.2024.111527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Radiopharmaceuticals are drugs used in treatment or diagnosis that contain a radioactive part, usually a pharmaceutical part in their structure. Adverse drug reactions are harmful and unexpected responses that occur when administered at normal doses. Although radiopharmaceuticals are regarded as safe medical products, adverse reactions should not be ignored. More serious adverse reactions such as myelosuppression, pleural effusion, and death may develop in therapeutic radiopharmaceuticals due to their use at higher doses than those used in diagnosis. Therefore, monitoring adverse reactions and reporting them to health authorities is important. This review aims to provide information about adverse reactions that may be related to radiopharmaceuticals used in treatment.
Collapse
Affiliation(s)
- Mine Silindir-Gunay
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| | - Nur Ozolmez
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
2
|
Ciappuccini R, Nascimento C, Edet-Sanson A, Bardet S. Médecine nucléaire et cancers de la thyroïde en 2024 : iode 131, TEP et nouvelles approches théranostiques. Bull Cancer 2024; 111:10S64-10S72. [PMID: 39505438 DOI: 10.1016/s0007-4551(24)00409-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
NUCLEAR MEDICINE AND THYROID CANCERS IN 2024: IODINE 131, PET AND NEW THERANOSTIC APPROACHES: Nuclear medicine has long been a mainstay in the management of thyroid cancers. In patients with differentiated thyroid cancer (DTC), the most common histotype, radioiodine (RAI, 131I) has been for years a cornerstone for the treatment of RAI-avid metastases. Post-therapeutic 131I scintigraphy helps guide these treatments and contributes to the definition of refractory cancers. In these refractory patients, who represent fewer than 5% of CTDs, 18FDG PET plays a central diagnostic and prognostic role. From a therapeutic perspective, RAI uptake can be re-induced in some of these patients with the BRAF mutation by using redifferentiation protocols. In anaplastic thyroid cancer (A TC) that is rare, aggressive and undifferentiated, 18FDG PET remains the metabolic imaging of choice. In medullary thyroid cancer (MTC), PET imaging is mainly based on the use of 18F-DOPA, even if 18FDG also provides prognostic data and 68Ga-DOTATOC could allow a theranostic approach. Other radiopharmaceuticals offering new theranostic avenues in thyroid cancers are also discussed, such as prostate-specific membrane antigen (PSMA) and fibroblast activation protein (FAP). After decades of a "one-size fits all" approach in thyroid cancer management, molecular imaging is paving the way towards personalized medicine.
Collapse
Affiliation(s)
- Renaud Ciappuccini
- Service de médecine nucléaire et UCP thyroïde, Centre François-Badesse, 3, avenue du Général-Harris, 14000 Caen, France; Inserm 1086 ANTICIPE, BioTICLA, Université de Caen Normandie, 14000 Caen, France.
| | - Camila Nascimento
- Service de médecine nucléaire, IUCT-Oncopole, 1, avenue Irène-Joliot-Curie, 31000, Toulouse, France
| | - Agathe Edet-Sanson
- Service de médecine nucléaire, Centre Henri-Becquerel, 1, rue d'Amiens, 76038 Rouen, France
| | - Stéphane Bardet
- Service de médecine nucléaire et UCP thyroïde, Centre François-Badesse, 3, avenue du Général-Harris, 14000 Caen, France
| |
Collapse
|
3
|
Puranik AD, Choudhury S, Ghosh S, Dev ID, Ramchandani V, Uppal A, Bhosale V, Palsapure A, Rungta R, Pandey R, Khatri S, George G, Satamwar Y, Maske R, Agrawal A, Shah S, Purandare NC, Rangarajan V. Tata Memorial Centre Evidence Based Use of Nuclear medicine diagnostic and treatment modalities in cancer. Indian J Cancer 2024; 61:S1-S28. [PMID: 38424680 DOI: 10.4103/ijc.ijc_52_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
ABSTRACT PET/CT and radioisotope therapy are diagnostic and therapeutic arms of Nuclear Medicine, respectively. With the emergence of better technology, PET/CT has become an accessible modality. Diagnostic tracers exploring disease-specific targets has led the clinicians to look beyond FDG PET. Moreover, with the emergence of theranostic pairs of radiopharmaceuticals, radioisotope therapy is gradually making it's way into treatment algorithm of common cancers in India. We therefore would like to discuss in detail the updates in PET/CT imaging and radionuclide therapy and generate a consensus-driven evidence based document which would guide the practitioners of Oncology.
Collapse
Affiliation(s)
- Ameya D Puranik
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Duan H, Iagaru A, Aparici CM. Radiotheranostics - Precision Medicine in Nuclear Medicine and Molecular Imaging. Nanotheranostics 2022; 6:103-117. [PMID: 34976584 PMCID: PMC8671964 DOI: 10.7150/ntno.64141] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023] Open
Abstract
'See what you treat and treat what you see, at a molecular level', could be the motto of theranostics. The concept implies diagnosis (imaging) and treatment of cells (usually cancer) using the same molecule, thus guaranteeing a targeted cytotoxic approach of the imaged tumor cells while sparing healthy tissues. As the brilliant late Sam Gambhir would say, the imaging agent acts like a 'molecular spy' and reveals where the tumoral cells are located and the extent of disease burden (diagnosis). For treatment, the same 'molecular spy' docks to the same tumor cells, this time delivering cytotoxic doses of radiation (treatment). This duality represents the concept of a 'theranostic pair', which follows the scope and fundamental principles of targeted precision and personalized medicine. Although the term theranostic was noted in medical literature in the early 2000s, the principle is not at all new to nuclear medicine. The first example of theranostic dates back to 1941 when Dr. Saul Hertz first applied radioiodine for radionuclide treatment of thyroid cells in patients with hyperthyroidism. Ever since, theranostics has been an integral element of nuclear medicine and molecular imaging. The more we understand tumor biology and molecular pathology of carcinogenesis, including specific mutations and receptor expression profiles, the more specific these 'molecular spies' can be developed for diagnostic molecular imaging and subsequent radionuclide targeted therapy (radiotheranostics). The appropriate selection of the diagnostic and therapeutic radionuclide for the 'theranostic pair' is critical and takes into account not only the type of cytotoxic radiation emission, but also the linear energy transfer (LET), and the physical half-lives. Advances in radiochemistry and radiopharmacy with new radiolabeling techniques and chelators are revolutionizing the field. The landscape of cytotoxic systemic radionuclide treatments has dramatically expanded through the past decades thanks to all these advancements. This article discusses present and promising future theranostic applications for various types of diseases such as thyroid disorders, neuroendocrine tumors (NET), pediatric malignancies, and prostate cancer (PC), and provides an outlook for future perspectives.
Collapse
Affiliation(s)
| | | | - Carina Mari Aparici
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Stanford University, Stanford, CA, USA
| |
Collapse
|
5
|
Nunes RF, Zuppani RMF, Coutinho AM, Barbosa FG, Sapienza MT, Marin JFG, Buchpiguel CA. General Concepts in Theranostics. PET Clin 2021; 16:313-326. [PMID: 34053576 DOI: 10.1016/j.cpet.2021.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Theranostics describes the pairing of diagnostic biomarkers and therapeutic agents with common specific targets. Nuclear medicine is the greatest theranostics protagonist, relying on radioactive tracers for imaging biologic phenomena and delivering ionizing radiation to the tissues that take up those tracers. The concept has gained importance with the growth of personalized medicine, allowing customized management for diseases, refining patient selection, better predicting responses, reducing toxicity, and estimating prognosis. This work provides an overview of the general concepts of the theranostics approach in nuclear medicine discussing its background, features, and future directions in imaging and therapy.
Collapse
Affiliation(s)
- Rafael F Nunes
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil.
| | - Roberta M F Zuppani
- Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Artur M Coutinho
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil; Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Felipe G Barbosa
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil
| | - Marcelo T Sapienza
- Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Jose Flavio G Marin
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil; Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Carlos A Buchpiguel
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil; Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
6
|
Maghsoomi Z, Emami Z, Malboosbaf R, Malek M, Khamseh ME. Efficacy and safety of peptide receptor radionuclide therapy in advanced radioiodine-refractory differentiated thyroid cancer and metastatic medullary thyroid cancer: a systematic review. BMC Cancer 2021; 21:579. [PMID: 34016077 PMCID: PMC8139052 DOI: 10.1186/s12885-021-08257-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/26/2021] [Indexed: 12/23/2022] Open
Abstract
Background It has been shown that a subgroup of patients with differentiated thyroid cancer (DTC) and medullary thyroid carcinoma (MTC) would progress to advanced stages of thyroid cancer. Therefore, the present study was done to systematically review available evidence in order to investigate efficacy and safety of peptide receptor radionuclide therapy (PRRT) in the patients with advanced radioiodine refractory differentiated thyroid cancer (RR-DTC) and metastatic MTC. Methods For this purpose, relevant studies investigated safety and efficacy of PRRT in the patients with advanced RR-DTC and metastatic MTC were identified by searching Medline (Pubmed, Ovid, and Ebsco), Scopus, Embase, Web of Science, and Cochrane Library databases (from database inception to March 24, 2021). The review was performed according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement. Searching was done independently by two investigators. Two researchers independently extracted the data and any disagreement was adjudicated by consensus. Quality of the studies was assessed using the tool of case reports/series in systematic reviews. Results Among 2284 related papers, 41 papers met the inclusion criteria. A total of 157 patients with RR-DTC were treated with PPRT. Biochemical and objective responses (partial and complete) were observed in 25.3 and 10.5% of patients, respectively. Among 220 patients with metastatic MTC, biochemical and objective responses were observed in 37.2 and 10.6% of the patients, respectively. Forty-six deaths were reported in 95 patients with advanced RR-DTC. In addition, 63 deaths were observed in 144 patients with metastatic MTC. Major side effects were reported in 124 patients treated with 90Y -based agent. In the patients treated with 177Lu-DOTA-TATE and 111In-Octreotide, mild and transient hematologic or renal complications were reported. Conclusion Findings of the study revealed that in the absence of the established treatment for the patients with RR-DTC and metastatic MTC, PRRT could be effective with few adverse events. Trial registration PROSPERO registration number: CRD42019125245. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08257-x.
Collapse
Affiliation(s)
- Zohreh Maghsoomi
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran
| | - Zahra Emami
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran
| | - Ramin Malboosbaf
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran
| | - Mojtaba Malek
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mohammad E Khamseh
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran.
| |
Collapse
|
7
|
Filetti S, Durante C, Hartl D, Leboulleux S, Locati LD, Newbold K, Papotti MG, Berruti A. Thyroid cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Ann Oncol 2019; 30:1856-1883. [PMID: 31549998 DOI: 10.1093/annonc/mdz400] [Citation(s) in RCA: 554] [Impact Index Per Article: 110.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
MESH Headings
- Humans
- Adenocarcinoma, Follicular/diagnosis
- Adenocarcinoma, Follicular/epidemiology
- Adenocarcinoma, Follicular/pathology
- Adenocarcinoma, Follicular/therapy
- Carcinoma, Neuroendocrine/diagnosis
- Carcinoma, Neuroendocrine/epidemiology
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/therapy
- Follow-Up Studies
- Thyroid Cancer, Papillary/diagnosis
- Thyroid Cancer, Papillary/epidemiology
- Thyroid Cancer, Papillary/pathology
- Thyroid Cancer, Papillary/therapy
- Thyroid Carcinoma, Anaplastic/diagnosis
- Thyroid Carcinoma, Anaplastic/epidemiology
- Thyroid Carcinoma, Anaplastic/pathology
- Thyroid Carcinoma, Anaplastic/therapy
- Thyroid Neoplasms/diagnosis
- Thyroid Neoplasms/epidemiology
- Thyroid Neoplasms/pathology
- Thyroid Neoplasms/therapy
Collapse
Affiliation(s)
- S Filetti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - C Durante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - D Hartl
- Department of Head and Neck Oncology, Gustave Roussy, Villejuif; Université Paris Saclay, Villejuif
| | - S Leboulleux
- Université Paris Saclay, Villejuif; Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy, Villejuif, France
| | - L D Locati
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - K Newbold
- Head and Neck Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - M G Papotti
- Department of Pathology, University of Turin, Turin
| | - A Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Medical Oncology Unit, University of Brescia, ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
8
|
Lu-177-Based Peptide Receptor Radionuclide Therapy for Advanced Neuroendocrine Tumors. Nucl Med Mol Imaging 2017; 52:208-215. [PMID: 29942399 DOI: 10.1007/s13139-017-0505-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 10/18/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) is a systemic cytotoxic radiation therapy using a compound of β-emitting radionuclide chelated to a peptide for the treatment of tumor with overexpressed specific cell receptor such as somatostatin receptor subtype 2 (SSTR2) of neuroendocrine tumor (NET). Surgical resection should be performed for the curative treatment for NETs when it is feasible; however, a multi-disciplinary approach is needed when locally advanced or metastasized disease. PRRT with lutetium-177 (Lu-177)-labeled somatostatin analogues, as a new treatment modality targeting metastatic or inoperable NETs expressing the SSTR2, have been developed and successfully used for the past two decades. As Lu-177 emits both β- and γ-radiation, it has the ability as a theragnostic agent for NETs compared with only β-emitting yttrium-90 labeled PRRT. Several recent studies reported that Lu-177 gave an overall positive response and improved the patients' quality of life. To fully exploit its potential, large comparative studies are needed for the assessment of distinct efficacies of Lu-177 labeled PRRT. Additionally, for extending the indications and developing new regimens of Lu-177-based PRRT, more dedicated clinical research is required.
Collapse
|
9
|
Abstract
Theranostics labeled with Y-90 or Lu-177 are highly efficient therapeutic approaches for the systemic treatment of various cancers including neuroendocrine tumors and prostate cancer. Peptide receptor radionuclide therapy (PRRT) has been used for many years for metastatic or inoperable neuroendocrine tumors. However, renal and hematopoietic toxicities are the major limitations for this therapeutic approach. Kidneys have been considered as the "critical organ" because of the predominant glomerular filtration, tubular reabsorption by the proximal tubules, and interstitial retention of the tracers. Severe nephrotoxity, which has been classified as grade 4-5 based on the "Common Terminology Criteria on Adverse Events," was reported in the range from 0%-14%. There are several risk factors for renal toxicity; patient-related risk factors include older age, preexisting renal disease, hypertension, diabetes mellitus, previous nephrotoxic chemotherapy, metastatic lesions close to renal parenchyma, and single kidney. There are also treatment-related issues, such as choice of radionuclide, cumulative radiation dose to kidneys, renal radiation dose per cycle, activity administered, number of cycles, and time interval between cycles. In the literature, nephrotoxicity caused by PRRT was documented using different criteria and renal function tests, from serum creatinine level to more accurate and sophisticated methods. Generally, serum creatinine level was used as a measure of kidney function. Glomerular filtration rate (GFR) estimation based on serum creatinine was preferred by several authors. Most commonly used formulas for estimation of GFR are "Modifications of Diet in Renal Disease" (MDRD) equation and "Cockcroft-Gault" formulas. However, more precise methods than creatinine or creatinine clearance are recommended to assess renal function, such as GFR measurements using Tc-99m-diethylenetriaminepentaacetic acid (DTPA), Cr-51-ethylenediaminetetraacetic acid (EDTA), or measurement of Tc-99m-MAG3 clearance, particularly in patients with preexisting risk factors for long-term nephrotoxicity. Proximal tubular reabsorption and interstitial retention of tracers result in excessive renal irradiation. Coinfusion of positively charged amino acids, such as l-lysine and l-arginine, is recommended to decrease the renal retention of the tracers by inhibiting the proximal tubular reabsorption. Furthermore, nephrotoxicity may be reduced by dose fractionation. Patient-specific dosimetric studies showed that renal biological effective dose of <0Gy was safe for patients without any risk factors. A renal threshold value <28Gy was recommended for patients with risk factors. Despite kidney protection, renal function impairment can occur after PRRT, especially in patients with risk factors and high single or cumulative renal absorbed dose. Therefore, patient-specific dosimetry may be helpful in minimizing the renal absorbed dose while maximizing the tumor dose. In addition, close and accurate renal function monitoring using more precise methods, rather than plasma creatinine levels, is essential to diagnose the early renal functional changes and to follow-up the renal function during the treatment.
Collapse
Affiliation(s)
- Belkis Erbas
- Department of Nuclear Medicine, Hacettepe University, Medical School, Ankara, Turkey.
| | - Murat Tuncel
- Department of Nuclear Medicine, Hacettepe University, Medical School, Ankara, Turkey
| |
Collapse
|
10
|
Estorch M. Tratamiento con 177Lu-DOTATATE: pasado, presente y futuro. Rev Esp Med Nucl Imagen Mol 2017; 36:69-71. [DOI: 10.1016/j.remn.2017.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 11/30/2022]
|
11
|
Use of 177 Lu-dotatate in the treatment of iodine refractory thyroid carcinomas. Rev Esp Med Nucl Imagen Mol 2017. [DOI: 10.1016/j.remnie.2016.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
12
|
Oliván-Sasot P, Falgás-Lacueva M, García-Sánchez J, Vera-Pinto V, Olivas-Arroyo C, Bello-Arques P. Use of 177Lu-dotatate in the treatment of iodine refractory thyroid carcinomas. Rev Esp Med Nucl Imagen Mol 2016; 36:116-119. [PMID: 27793633 DOI: 10.1016/j.remn.2016.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/19/2016] [Accepted: 08/29/2016] [Indexed: 11/29/2022]
Abstract
In a patient with a differentiated thyroid cancer the standard treatment protocol to be followed is surgery, ablation of thyroid remnants with 131Iodine (131I), and TSH suppression. However, the treatment with 131I is not effective in some cases, and it no longer becomes a therapeutic option due to cell de-differentiation with loss of 131I uptake. Systemic treatment can be used as other options, although patients are not always responsive; thus, the disease may progress and therapeutic options may run out. Endocrine tumours may express somatostatin receptors,and this characteristic has been used, not only for diagnosis, but also for their treatment through somatostatin analogue labelling with radioactive isotopes. This was the case of a patient suffering from iodine-refractory follicular thyroid carcinoma, with somatostatin receptors expression, treated with 177Lu-DOTATATE, showing an excellent clinical and analytical response.
Collapse
Affiliation(s)
- P Oliván-Sasot
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, España.
| | - M Falgás-Lacueva
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - J García-Sánchez
- Servicio de Oncología Médica, Hospital Arnau de Vilanova, Valencia, España
| | - V Vera-Pinto
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - C Olivas-Arroyo
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - P Bello-Arques
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, España
| |
Collapse
|
13
|
Brunner P, Jörg AC, Glatz K, Bubendorf L, Radojewski P, Umlauft M, Marincek N, Spanjol PM, Krause T, Dumont RA, Maecke HR, Müller-Brand J, Briel M, Schmitt A, Perren A, Walter MA. The prognostic and predictive value of sstr2-immunohistochemistry and sstr2-targeted imaging in neuroendocrine tumors. Eur J Nucl Med Mol Imaging 2016; 44:468-475. [DOI: 10.1007/s00259-016-3486-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/05/2016] [Indexed: 11/29/2022]
|
14
|
Ahlstedt J, Tran TA, Strand SE, Gram M, Åkerström B. Human Anti-Oxidation Protein A1M--A Potential Kidney Protection Agent in Peptide Receptor Radionuclide Therapy. Int J Mol Sci 2015; 16:30309-20. [PMID: 26694383 PMCID: PMC4691176 DOI: 10.3390/ijms161226234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 11/28/2015] [Accepted: 12/11/2015] [Indexed: 11/16/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has been in clinical use for 15 years to treat metastatic neuroendocrine tumors. PRRT is limited by reabsorption and retention of the administered radiolabeled somatostatin analogues in the proximal tubule. Consequently, it is essential to develop and employ methods to protect the kidneys during PRRT. Today, infusion of positively charged amino acids is the standard method of kidney protection. Other methods, such as administration of amifostine, are still under evaluation and show promising results. α1-microglobulin (A1M) is a reductase and radical scavenging protein ubiquitously present in plasma and extravascular tissue. Human A1M has antioxidation properties and has been shown to prevent radiation-induced in vitro cell damage and protect non-irradiated surrounding cells. It has recently been shown in mice that exogenously infused A1M and the somatostatin analogue octreotide are co-localized in proximal tubules of the kidney after intravenous infusion. In this review we describe the current situation of kidney protection during PRRT, discuss the necessity and implications of more precise dosimetry and present A1M as a new, potential candidate for renal protection during PRRT and related targeted radionuclide therapies.
Collapse
Affiliation(s)
- Jonas Ahlstedt
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund 221 84, Sweden.
| | - Thuy A Tran
- Lund University Bioimaging Center, Lund University, Lund 221 84, Sweden.
| | - Sven-Erik Strand
- Section of Medical Radiation Physics, Department of Clinical Sciences in Lund, Lund University, Lund 221 84, Sweden.
| | - Magnus Gram
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund 221 84, Sweden.
| | - Bo Åkerström
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund 221 84, Sweden.
| |
Collapse
|
15
|
Traub-Weidinger T, Putzer D, von Guggenberg E, Dobrozemsky G, Nilica B, Kendler D, Bale R, Virgolini IJ. Multiparametric PET imaging in thyroid malignancy characterizing tumour heterogeneity: somatostatin receptors and glucose metabolism. Eur J Nucl Med Mol Imaging 2015; 42:1995-2001. [DOI: 10.1007/s00259-015-3114-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 06/10/2015] [Indexed: 01/22/2023]
|
16
|
Defining a rational step-care algorithm for managing thyroid carcinoma patients with elevated thyroglobulin and negative on radioiodine scintigraphy (TENIS): considerations and challenges towards developing an appropriate roadmap. Eur J Nucl Med Mol Imaging 2015; 42:1167-71. [DOI: 10.1007/s00259-015-3058-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 03/31/2015] [Indexed: 11/26/2022]
|
17
|
Marincek N, Radojewski P, Dumont RA, Brunner P, Müller-Brand J, Maecke HR, Briel M, Walter MA. Somatostatin Receptor–Targeted Radiopeptide Therapy with 90Y-DOTATOC and 177Lu-DOTATOC in Progressive Meningioma: Long-Term Results of a Phase II Clinical Trial. J Nucl Med 2015; 56:171-6. [DOI: 10.2967/jnumed.114.147256] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
18
|
Lapa C, Werner RA, Schmid JS, Papp L, Zsótér N, Biko J, Reiners C, Herrmann K, Buck AK, Bundschuh RA. Prognostic value of positron emission tomography-assessed tumor heterogeneity in patients with thyroid cancer undergoing treatment with radiopeptide therapy. Nucl Med Biol 2014; 42:349-54. [PMID: 25595135 DOI: 10.1016/j.nucmedbio.2014.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 12/02/2014] [Accepted: 12/04/2014] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Peptide receptor radionuclide therapy (PRRT) is a treatment option for both iodine-refractory differentiated and advanced medullary thyroid cancer (TC). It requires over-expression of somatostatin receptor subtype II (SSTR) that can be non-invasively assessed by positron emission tomography (PET). Assessment of tumor heterogeneity is increasingly used as a tool for prognostication prediction. We investigated the potential of SSTR-PET to assess intraindividual tumor heterogeneity and thereby treatment response prior to PRRT. METHODS 12 patients with progressive radioiodine-refractory differentiated (1 papillary, 1 oxyphilic, 2 oncocytic, 4 follicular) or medullary (n=4) TC were enrolled. SSTR-PET was performed at baseline. Conventional PET parameters and heterogeneity parameters were analyzed regarding their potential to predict progression-free (PFS, mean, 221 days) and overall survival (OS, mean, 450 days). Parameters of a subgroup of lesions (n=23) were also correlated with morphological response according to modified RECIST criteria. RESULTS In patient-based analysis, all conventional parameters failed to predict PFS. Several textural parameters showed a significant capability to assess PFS. Thereby, "Grey level non uniformity" had the highest area under the curve (AUC, 0.93) in Receiver operating characteristics analysis followed by "Contrast" (AUC, 0.89). In lesion-based analysis, only "Entropy" revealed potential to evaluate disease progression. OS could not be assessed by any parameter investigated. CONCLUSIONS Tumor heterogeneity seems to be a predictor of response to PRRT in patients with iodine-refractory differentiated/advanced medullary thyroid cancer and outperforms conventional PET parameters like standardized uptake value. In a "theranostic" approach, assessment of textural parameters may help in selecting patients who might benefit from PRRT.
Collapse
Affiliation(s)
- Constantin Lapa
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany.
| | - Rudolf A Werner
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany.
| | - Jan-Stefan Schmid
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany.
| | - Laszló Papp
- Mediso Medical Imaging Systems Ltd., Budapest, Hungary.
| | | | - Johannes Biko
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany.
| | - Christoph Reiners
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany.
| | - Ken Herrmann
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany.
| | - Andreas K Buck
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany.
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany; Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Bonn, Bonn, Germany.
| |
Collapse
|
19
|
Dumont RA, Seiler D, Marincek N, Brunner P, Radojewski P, Rochlitz C, Müller-Brand J, Maecke HR, Briel M, Walter MA. Survival after somatostatin based radiopeptide therapy with (90)Y-DOTATOC vs. (90)Y-DOTATOC plus (177)Lu-DOTATOC in metastasized gastrinoma. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2014. [PMID: 25625026 DOI: 10.7892/boris.67183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We aimed to explore the effects of (90)Y-DOTATOC and (90)Y-DOTATOC plus (177)Lu-DOTATOC on survival of patients with metastasized gastrinoma. Patients with progressive metastasized gastrinoma were treated with repeated cycles of (90)Y-DOTATOC or with cycles alternating between (90)Y-DOTATOC and (177)Lu-DOTATOC until tumor progression or permanent toxicity. Multivariable Cox regression analyses were used to study predictors of survival. A total of 36 patients were enrolled; 30 patients received (90)Y-DOTATOC (median activity per patient 11.8GBq; range: 6.1-62.2GBq) and 6 patients received (90)Y-DOTATOC plus (177)Lu-DOTATOC (median activity per patient: 14.8GBq; range: 7.4-14.8GBq). Response was found in 26 patients (72.2%), including morphological (n=12, 33.3%), biochemical (n=14, 38.9%) and/or clinical response (n=6, 16.2%). A total of 21 patients (58.3%) experienced hematotoxicity grade 1/2, while 1 patient (2.8%) experienced hematotoxicity grade 3; no grade 4 hematotoxicity occurred. Furthermore, 2 patients (5.6%) developed grade 4 renal toxicity; no grade 5 renal toxicity occurred. Responders had a significantly longer median survival from time of enrollment than non-responders (45.1 months, range: 37.1-53.1 months vs. 12.6 months, range: 11.0-14.2, hazard ratio: 0.12 (0.027-0.52), p=0.005). Additionally, there was a trend towards longer median survival with (90)Y-DOTATOC plus (177)Lu-DOTATOC as compared to (90)Y-DOTATOC alone (60.2 months, range: 19.8-100.6 months vs. 27.0 months, range: 4.0-50.0, hazard ratio: 0.21 (0.01-3.98), p=0.16). Response to (90)Y-DOTATOC and (90)Y-DOTATOC plus (177)Lu-DOTATOC therapy is associated with a longer survival in patients with metastasized gastrinoma. Both treatment regimens are promising tools for management of progressive gastrinoma.
Collapse
Affiliation(s)
- Rebecca A Dumont
- Institute of Nuclear Medicine, University Hospital Basel CH ; Department of Radiology, David Geffen School of Medicine, UCLA Los Angeles, USA
| | - Daniela Seiler
- Institute of Nuclear Medicine, University Hospital Basel CH
| | - Nicolas Marincek
- Institute of Nuclear Medicine, University Hospital Basel CH ; Institute of Nuclear Medicine, University Hospital Bern CH
| | | | | | | | | | - Helmut R Maecke
- Division of Radiological Chemistry, University Hospital Basel CH
| | - Matthias Briel
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel CH ; Department of Clinical Epidemiology and Biostatistics, McMaster University Hamilton, CA
| | - Martin A Walter
- Institute of Nuclear Medicine, University Hospital Basel CH ; Institute of Nuclear Medicine, University Hospital Bern CH
| |
Collapse
|
20
|
Lindner C, Dierneder J, Pall G, Pirich C, Hoffmann M, Raderer M, Becherer A, Niederle B, Lipp R, Lind P, Gallowitsch H, Romeder F, Virgolini I. [Treatment of patients with radioiodine refractory, differentiated thyroid carcinoma. A Consensus Statement]. Nuklearmedizin 2014; 54:125-30. [PMID: 25421138 DOI: 10.3413/nukmed-0688-14-07] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/11/2014] [Indexed: 02/01/2023]
Abstract
There is no clear standard therapy for patients with radioactive iodine (131I)-refractory locally advanced or metastatic differentiated thyroid cancer. The therapeutic options for this indication have expanded with the recently approved multiple kinase inhibitor sorafenib. Recommendations for the definition and the management of iodine refractory patients were worked up by an interdisciplinary expert panel, consisting of endocrine surgeons, medical oncologists and nuclear medicine specialists.
Collapse
Affiliation(s)
- Christina Lindner
- Christina Lindner, MSc, Department Life Sciences, Medizinische und Pharmazeutische Biotechnologie, IMC FH Krems, E-Mail:
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Czepczyński R, Matysiak-Grześ M, Gryczyńska M, Bączyk M, Wyszomirska A, Stajgis M, Ruchała M. Peptide receptor radionuclide therapy of differentiated thyroid cancer: efficacy and toxicity. Arch Immunol Ther Exp (Warsz) 2014; 63:147-54. [PMID: 25403743 PMCID: PMC4359293 DOI: 10.1007/s00005-014-0318-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 08/01/2014] [Indexed: 11/24/2022]
Abstract
In rare cases of differentiated thyroid carcinoma (DTC), radioiodine treatment is no longer effective due to cell dedifferentiation. Targeting somatostatin receptors in DTC cells by radiolabelled somatostatin analogues could provide an alternative therapy option. The aim of this study was to evaluate safety and efficacy of peptide receptor radionuclide therapy (PRRT) in patients with advanced, non-iodine avid DTC. Eleven patients aged 47–81 years (median: 65 years) with a history of several courses of radioiodine therapy, increasing thyroglobulin (Tg) and negative whole body scan, were qualified to the study. After confirming receptor expression by somatostatin receptor scintigraphy, PRRT with yttrium-90 labelled analogue was initiated. Fractionated treatment protocol was used with four doses of 90Y-DOTA-TOC in 12-week intervals. Activity of each dose was 3.7 GBq (100 mCi). Of 11 patients, 5 died before receiving the fourth course of PRRT. In the remaining six patients, morphological response, evaluated 3 months after the last course using RECIST criteria showed partial remission (PR) in one patient, stable disease (SD) in two patients and progressive disease (PD) in three patients. Biochemical response based on Tg measurements before and after PRRT showed PR in one patient, SD in four patients and PD in one patient. Median survival was 21 months from the first course of PRRT. Only minor and transient hematological toxicity was observed in some patients. We conclude that PRRT is generally well-tolerated and may be a valuable option for some patients with radioiodine-refractory DTC.
Collapse
Affiliation(s)
- Rafał Czepczyński
- Department of Endocrinology, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznań, Poland,
| | | | | | | | | | | | | |
Collapse
|
22
|
Woelfl S, Bogner S, Huber H, Salaheddin-Nassr S, Hatzl M, Decristoforo C, Virgolini I, Gabriel M. Expression of somatostatin receptor subtype 2 and subtype 5 in thyroid malignancies. Nuklearmedizin 2014; 53:179-85. [PMID: 24967740 DOI: 10.3413/nukmed-0646-14-02] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 06/12/2014] [Indexed: 11/20/2022]
Abstract
AIM To retrospectively analyse the expression of somatostatin receptor subtypes 2 (SSTR 2) and 5 (SSTR 5) in thyroid malignancies, possibly the most relevant subtypes for targeted therapy with somatostatin peptide radioligands. In addition, findings were also correlated with the course of disease. PATIENTS, METHODS 87 consecutive patients (59 women, 28 men) with thyroid malignancy were included; 52 had papillary carcinoma, 24 follicular carcinoma, six medullary carcinoma, two poorly differentiated carcinoma and three anaplastic carcinoma. After initial therapy 70 (80.5%) patients showed complete remission, 11 (12.6%) patients partial remission with clinical and biochemical signs of residual disease and six (6.9%) patients progressive disease. The immunohistochemical staining results of the primary malignancy for SSTR 2 and SSTR 5 were semiquantitatively assessed and correlated with various outcome parameters. RESULTS In 10 of 87 (11.49%) thyroid cancer samples SSTR 2 showed positive immunohistochemical expression as compared to 75 of 87 (86.20%) for SSTR 5. All SSTR 2-positive cases expressed SSTR 5. Persistent or recurrent disease was found in 17 of 87 cases (19.54%). Fifty percent (6 /12) of SSTR 5-negative patients showed persistent disease as compared to 14.7 % (11 / 75) of SSTR 5-positive patients: seven of these were exclusively SSTR 5-positive, 4 showed dual expression of SSTR 5 and SSTR 2 (p = 0.01). No case showed only SSTR 2 expression. CONCLUSIONS SSTR 5 was shown to be the main receptor subtype in the analysed differentiated or anaplastic thyroid malignancies, whereas SSTR 2 was found only in a small percentage. Deficient SSTR expression may indicate higher risk for persistent or recurrent disease after initial therapy. For this reason immunohistochemistry can be considered a prognostic marker which should be further validated in prospective studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - M Gabriel
- Prof. Dr. Michael Gabriel, Department of Nuclear Medicine and Endocrinology General Hospital, Krankenhausstr. 9, 4021 Linz, Austria, Tel. +43/(0)732/78 06 61-41, Fax -65, E-mail: ,
| |
Collapse
|
23
|
Versari A, Sollini M, Frasoldati A, Fraternali A, Filice A, Froio A, Asti M, Fioroni F, Cremonini N, Putzer D, Erba PA. Differentiated thyroid cancer: a new perspective with radiolabeled somatostatin analogues for imaging and treatment of patients. Thyroid 2014; 24:715-26. [PMID: 24102584 DOI: 10.1089/thy.2013.0225] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND The expression of somatostatin receptors (SSTR) in thyroid cells may offer the possibility to identify metastatic lesions and to select patients for peptide receptor radionuclide therapy (PRRT). We investigated (68)Ga-DOTATOC positron emission tomography/computed tomography (PET/CT) to select patients with progressive differentiated thyroid cancer (DTC) for PRRT as well as treatment response and toxicity in treated patients. METHODS We enrolled 41 patients with progressive radioiodine-negative DTC (24 women and 17 men; mean age=54.3 years, median=59 years, range=19-78 years). In all patients, [(18)F]FDG-PET/CT was performed to determine recurrent disease with enhanced glucose metabolism, and (68)Ga-DOTATOC PET/CT was used to identify SSTR expression. Dosimetric evaluation was performed with (111)In-DOTATOC scintigraphy. Eleven patients were treated with PRRT receiving a fractionated injection of 1.5-3.7 GBq (90)Y-DOTATOC/administration. Serial (68)Ga-DOTATOC PET/CT scans were performed in all treated patients to evaluate treatment response. Parameters provided by (68)Ga-DOTATOC PET/CT were analyzed as potential therapeutic predictors to differentiate responding from nonresponding. In all treated patients, adverse events and toxicity were recorded. RESULTS (68)Ga-DOTATOC PET/CT were positive in 24/41 of radioiodine-negative DTC patients. Based on the high expression of SSTR detected by (68)Ga-DOTATOC PET/CT, 13 patients were suitable for PRRT. Two out of 13 patients were not treated due to the lack of fulfillment of other study inclusion criteria. PRRT induced disease control in 7/11 patients (two partial response and five stabilization) with a duration of response of 3.5-11.5 months. Objective response was associated with symptoms relief. Functional volume (FV) over time obtained by PET/CT was the only parameter demonstrating a significant difference between lesions responding and nonresponding to PRRT (p=0.001). Main PRRT adverse events were nausea, asthenia, and transient hematologic toxicity. One patient experienced permanent renal toxicity. CONCLUSIONS In our series, SSTR imaging provided positive results in more than half of the cases with radioiodine-negative DTC, and about one third of patients were eligible for PRRT. (68)Ga-DOTATOC PET/CT seems a reliable tool both for patient selection and evaluation of treatment response. In our experience, FV determination over time seems to represent a reliable parameter to determine tumor response to PRRT, although further investigations are needed to better define its role.
Collapse
Affiliation(s)
- Annibale Versari
- 1 Nuclear Medicine, Arcispedale Santa Maria Nuova, Clinical Cancer Research Institute (IRCCS) , Reggio Emilia, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Somatostatin-based radiopeptide therapy with [177Lu-DOTA]-TOC versus [90Y-DOTA]-TOC in neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2013; 41:214-22. [DOI: 10.1007/s00259-013-2559-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 08/27/2013] [Indexed: 11/29/2022]
|
25
|
Zaknun JJ, Bodei L, Mueller-Brand J, Pavel ME, Baum RP, Hörsch D, O’Dorisio MS, O’Dorisiol TM, Howe JR, Cremonesi M, Kwekkeboom DJ. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2013; 40:800-16. [PMID: 23389427 PMCID: PMC3622744 DOI: 10.1007/s00259-012-2330-6] [Citation(s) in RCA: 495] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Peptide receptor radionuclide therapy (PRRNT) is a molecularly targeted radiation therapy involving the systemic administration of a radiolabelled peptide designed to target with high affinity and specificity receptors overexpressed on tumours. PRRNT employing the radiotagged somatostatin receptor agonists (90)Y-DOTATOC ([(90)Y-DOTA(0),Tyr(3)]-octreotide) or (177)Lu-DOTATATE ([(177)Lu-DOTA(0),Tyr(3),Thr(8)]-octreotide or [(177)Lu-DOTA(0),Tyr(3)]-octreotate) have been successfully used for the past 15 years to target metastatic or inoperable neuroendocrine tumours expressing the somatostatin receptor subtype 2. Accumulated evidence from clinical experience indicates that these tumours can be subjected to a high absorbed dose which leads to partial or complete objective responses in up to 30 % of treated patients. Survival analyses indicate that patients presenting with high tumour receptor expression at study entry and receiving (177)Lu-DOTATATE or (90)Y-DOTATOC treatment show significantly higher objective responses, leading to longer survival and improved quality of life. Side effects of PRRNT are typically seen in the kidneys and bone marrow. These, however, are usually mild provided adequate protective measures are undertaken. Despite the large body of evidence regarding efficacy and clinical safety, PRRNT is still considered an investigational treatment and its implementation must comply with national legislation, and ethical guidelines concerning human therapeutic investigations. This guidance was formulated based on recent literature and leading experts' opinions. It covers the rationale, indications and contraindications for PRRNT, assessment of treatment response and patient follow-up. This document is aimed at guiding nuclear medicine specialists in selecting likely candidates to receive PRRNT and to deliver the treatment in a safe and effective manner. This document is largely based on the book published through a joint international effort under the auspices of the Nuclear Medicine Section of the International Atomic Energy Agency.
Collapse
Affiliation(s)
- John J. Zaknun
- Nuclear Medicine Section, Division of Human Health, International Atomic Energy Agency, IAEA, Vienna, Austria
- Zentralklinik Bad Berka, Center for Molecular Radiotherapy and Molecular Imaging, ENETS Center of Excellence, Bad Berka, Germany
| | - L. Bodei
- Division of Nuclear Medicine, European Institute of Oncology, Milan, Italy
| | - J. Mueller-Brand
- Klinik und Institut für Nuklearmedizin, Universitätsspital Basel, Basel, Switzerland
| | - M. E. Pavel
- Campus Virchow Klinikum, Klinik für Gastroenterologie, Hepatologie, Endokrinologie, Diabetes und Stoffwechsel-erkrankungen, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - R. P. Baum
- Zentralklinik Bad Berka, Department of Internal Medicine, Gastroenterology and Endocrinology, ENETS Center of Excellence, Bad Berka, Germany
| | - D. Hörsch
- Zentralklinik Bad Berka, Department of Internal Medicine, Gastroenterology and Endocrinology, ENETS Center of Excellence, Bad Berka, Germany
| | - M. S. O’Dorisio
- RJ and LA Carver College of Medicine, Department of Pediatrics, University of Iowa, Iowa City, IA USA
| | - T. M. O’Dorisiol
- RJ and LA Carver College of Medicine, Department of Internal Medicine, University of Iowa, Iowa City, IA USA
| | - J. R. Howe
- RJ and LA Carver College of Medicine, Department of Surgical Oncology, University of Iowa, Iowa City, IA USA
| | - M. Cremonesi
- Service of Health Physics, European Institute of Oncology, Milan, Italy
| | - D. J. Kwekkeboom
- Department of Nuclear Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Marincek N, Jörg AC, Brunner P, Schindler C, Koller MT, Rochlitz C, Müller-Brand J, Maecke HR, Briel M, Walter MA. Somatostatin-based radiotherapy with [90Y-DOTA]-TOC in neuroendocrine tumors: long-term outcome of a phase I dose escalation study. J Transl Med 2013; 11:17. [PMID: 23320604 PMCID: PMC3561188 DOI: 10.1186/1479-5876-11-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 01/09/2013] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND We describe the long-term outcome after clinical introduction and dose escalation of somatostatin receptor targeted therapy with [90Y-DOTA]-TOC in patients with metastasized neuroendocrine tumors. METHODS In a clinical phase I dose escalation study we treated patients with increasing [90Y-DOTA]-TOC activities. Multivariable Cox regression and competing risk regression were used to compare efficacy and toxicities of the different dosage protocols. RESULTS Overall, 359 patients were recruited; 60 patients were enrolled for low dose (median: 2.4 GBq/cycle, range 0.9-7.8 GBq/cycle), 77 patients were enrolled for intermediate dose (median: 3.3 GBq/cycle, range: 2.0-7.4 GBq/cycle) and 222 patients were enrolled for high dose (median: 6.7 GBq/cycle, range: 3.7-8.1 GBq/cycle) [90Y-DOTA]-TOC treatment. The incidences of hematotoxicities grade 1-4 were 65.0%, 64.9% and 74.8%; the incidences of grade 4/5 kidney toxicities were 8.4%, 6.5% and 14.0%, and the median survival was 39 (range: 1-158) months, 34 (range: 1-118) months and 29 (range: 1-113) months. The high dose protocol was associated with an increased risk of kidney toxicity (Hazard Ratio: 3.12 (1.13-8.59) vs. intermediate dose, p = 0.03) and a shorter overall survival (Hazard Ratio: 2.50 (1.08-5.79) vs. low dose, p = 0.03). CONCLUSIONS Increasing [90Y-DOTA]-TOC activities may be associated with increasing hematological toxicities. The dose related hematotoxicity profile of [90Y-DOTA]-TOC could facilitate tailoring [90Y-DOTA]-TOC in patients with preexisting hematotoxicities. The results of the long-term outcome suggest that fractionated [90Y-DOTA]-TOC treatment might allow to reduce renal toxicity and to improve overall survival. (ClinicalTrials.gov number NCT00978211).
Collapse
|
27
|
Villard L, Romer A, Marincek N, Brunner P, Koller MT, Schindler C, Ng QKT, Mäcke HR, Müller-Brand J, Rochlitz C, Briel M, Walter MA. Cohort study of somatostatin-based radiopeptide therapy with [(90)Y-DOTA]-TOC versus [(90)Y-DOTA]-TOC plus [(177)Lu-DOTA]-TOC in neuroendocrine cancers. J Clin Oncol 2012; 30:1100-6. [PMID: 22393097 DOI: 10.1200/jco.2011.37.2151] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Radiopeptide therapy is commonly performed with a single radioisotope. We aimed to compare the effectiveness of somatostatin-based radiopeptide therapy with a single versus a combination of radioisotopes. PATIENTS AND METHODS In a cohort study, patients with metastasized neuroendocrine cancer were treated with repeated cycles of (90)yttrium-labeled tetraazacyclododecane-tetraacetic acid modified Tyr-octreotide ([(90)Y-DOTA]-TOC) or with cycles alternating between [(90)Y-DOTA]-TOC and (177)lutetium-labeled DOTA-TOC ([(177)Lu-DOTA]-TOC) until tumor progression or permanent toxicity. Multivariable Cox regression and competing risk regression were used to study predictors of survival and renal toxicity in patients completing three or more treatment cycles. RESULTS A total of 486 patients completed three or more treatment cycles; 237 patients received [(90)Y-DOTA]-TOC and 249 patients received [(90)Y-DOTA]-TOC + [(177)Lu-DOTA]-TOC. Patients receiving [(90)Y-DOTA]-TOC + [(177)Lu-DOTA]-TOC had a significantly longer survival than patients receiving [(90)Y-DOTA]-TOC alone (5.51 v 3.96 years; hazard ratio, 0.64; 95% CI, 0.47 to 0.88; P = .006). The rates of severe hematologic toxicities (6.3% v 4.4%; P = .25) and severe renal toxicity (8.9% v 11.2%; P = .47) were comparable in both groups. CONCLUSION [(90)Y-DOTA]-TOC + [(177)Lu-DOTA]-TOC was associated with improved overall survival compared with [(90)Y-DOTA]-TOC alone in patients completing three or more cycles of treatment. Contrary to the current practice in radiopeptide therapy, our results suggest an advantage of using a combination of radioisotopes.
Collapse
Affiliation(s)
- Linda Villard
- University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Therapeutic options for advanced, unresectable radioiodine-resistant thyroid cancers have historically been limited. Recent progress in understanding the pathogenesis of the various subtypes of thyroid cancer has led to increased interest in the development of targeted therapies, with potential strategies including angiogenesis inhibition, inhibition of aberrant intracellular signaling in the MAPK and PI3K/AKT/mTOR pathways, radioimmunotherapy, and redifferentiation agents. On the basis of a recent positive phase III clinical trial, the RET, vascular endothelial growth factor receptor (VEGFR), and epidermal growth factor receptor (EGFR) inhibitor vandetanib has received FDA approval as of April 2011 for use in the treatment of advanced medullary thyroid cancer. Several other recent phase II clinical trials in advanced thyroid cancer have demonstrated significant activity, and multiple other promising therapeutic strategies are in earlier phases of clinical development. The recent progress in targeted therapy is already revolutionizing management paradigms for advanced thyroid cancer, and will likely continue to dramatically expand treatment options in the coming years.
Collapse
Affiliation(s)
- David A. Liebner
- Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus, OH, USA
| | - Manisha H. Shah
- A438 Starling-Loving Hall, 320 W 10th Ave, Columbus, OH 43210, USA
| |
Collapse
|
29
|
Imhof A, Brunner P, Marincek N, Briel M, Schindler C, Rasch H, Mäcke HR, Rochlitz C, Müller-Brand J, Walter MA. Response, survival, and long-term toxicity after therapy with the radiolabeled somatostatin analogue [90Y-DOTA]-TOC in metastasized neuroendocrine cancers. J Clin Oncol 2011; 29:2416-23. [PMID: 21555692 DOI: 10.1200/jco.2010.33.7873] [Citation(s) in RCA: 444] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE To investigate response, survival, and safety profile of the somatostatin-based radiopeptide (90)yttrium-labeled tetraazacyclododecane-tetraacetic acid modified Tyr-octreotide ([(90)Y-DOTA]-TOC) in neuroendocrine cancers. PATIENTS AND METHODS In a clinical phase II single-center open-label trial, patients with neuroendocrine cancers were treated with repeated cycles of [(90)Y-DOTA]-TOC. Each cycle consisted of a single intravenous injection of 3.7GBq/m(2) body-surface [(90)Y-DOTA]-TOC. Additional cycles were withheld in case of tumor progression and/or permanent toxicity. RESULTS Overall, 1,109 patients received 2,472 cycles of [(90)Y-DOTA]-TOC (median, two; range, one to 10 cycles per patient). Of the 1,109 patients, 378 (34.1%) experienced morphologic response; 172 (15.5%), biochemical response; and 329 (29.7%), clinical response. During a median follow-up of 23 months, 491 patients (44.3%) died. Longer survival was correlated with each: morphologic (hazard ratio [HR], 0.46; 95% CI, 0.38 to 0.56; median survival, 44.7 v 18.3 months; P < .001), biochemical (HR, 0.75; 95% CI, 0.59 to 0.96; 35.3 v 25.7 months; P = .023), and clinical response (HR, 0.68; 95% CI, 0.56 to 0.82; 36.8 v 23.5 months; P < .001). Overall, 142 patients (12.8%) developed grade 3 to 4 transient hematologic toxicities, and 103 patients (9.2%) experienced grade 4 to 5 permanent renal toxicity. Multivariable regression revealed that tumoral uptake in the initial imaging study was predictive for overall survival (HR, 0.45; 95% CI, 0.29 to 0.69; P < .001), whereas the initial kidney uptake was predictive for severe renal toxicity (HR, 1.59; 95% CI, 1.17 to 2.17; P = .003). CONCLUSION This study documents the long-term outcome of [(90)Y-DOTA]-TOC treatment in a large cohort. Response to [(90)Y-DOTA]-TOC is associated with longer survival. Somatostatin receptor imaging is predictive for both survival after [(90)Y-DOTA]-TOC treatment and occurrence of renal toxicity.
Collapse
Affiliation(s)
- Anna Imhof
- Institute of Nuclear Medicine, University Hospital, Petersgraben 4, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Goffredo V, Paradiso A, Ranieri G, Gadaleta CD. Yttrium-90 (90Y) in the principal radionuclide therapies: an efficacy correlation between peptide receptor radionuclide therapy, radioimmunotherapy and transarterial radioembolization therapy. Ten years of experience (1999-2009). Crit Rev Oncol Hematol 2011; 80:393-410. [PMID: 21388824 DOI: 10.1016/j.critrevonc.2011.01.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 01/12/2011] [Accepted: 01/27/2011] [Indexed: 01/17/2023] Open
Abstract
The clinical application of the pure beta emitter (90)Y constitutes a fundamental advancement in non-invasive medicine. Nowadays, mainly three oncological therapies exploit the intrinsic emissive characteristic of (90)Y. Radionuclide therapies include peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumour (NET) treatment, radioimmunotherapy (RIT) in non-Hodgkin's lymphoma (NHL) treatment and transarterial radioembolization therapy (TARET) in unresectable hepatocellular carcinoma (HCC) and liver metastatic colorectal cancer (mCRC) treatment. The last ten years of clinical experience from E-PubMed research have been reviewed and an efficacy correlation between (90)Y-therapies has shown a better objective response rate for RIT (ORR 80±15%; range 53-100) compared to PRRT (ORR 23.5±14%; range 9-50), and TARET (ORR for mCRC, 40±25%; range 19-91, and ORR for HCC, 42±20%; range 20-82). This review reports on the state of the art of the efficacy of (90)Y-therapies from the last decade and discusses new perspectives of therapeutic development.
Collapse
Affiliation(s)
- Veronica Goffredo
- Interventional Radiology Unit with Integrated Section of Medical Oncology, National Cancer Institute Giovanni Paolo II of Bari, Via Hahnemann 10, Bari, Italy.
| | | | | | | |
Collapse
|
31
|
Silberstein EB. The Problem of the Patient with Thyroglobulin Elevation but Negative Iodine Scintigraphy: The TENIS Syndrome. Semin Nucl Med 2011; 41:113-20. [DOI: 10.1053/j.semnuclmed.2010.10.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
32
|
Grozinsky-Glasberg S, Barak D, Fraenkel M, Walter MA, Müeller-Brand J, Eckstein J, Applebaum L, Shimon I, Gross DJ. Peptide receptor radioligand therapy is an effective treatment for the long-term stabilization of malignant gastrinomas. Cancer 2010; 117:1377-85. [DOI: 10.1002/cncr.25646] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 07/30/2010] [Accepted: 08/16/2010] [Indexed: 11/08/2022]
|
33
|
Saha S, Majumdar R, Dighe RR, Chakravarty AR. Enhanced photodynamic effect of cobalt(III) dipyridophenazine complex on thyrotropin receptor expressing HEK293 cells. Metallomics 2010; 2:754-65. [PMID: 21072367 DOI: 10.1039/c0mt00028k] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ternary cobalt(III) complexes [CoL(B)] (1-3) of a trianionic tetradentate phenolate-based ligand (L) and phenanthroline bases (B), viz. 1,10-phenanthroline (phen in 1), dipyridoquinoxaline (dpq in 2) and dipyridophenazine (dppz in 3) are synthesized, characterized from X-ray crystallographic, analytical and spectral techniques, and their utility in photodynamic therapy (PDT) of thyroid diseases caused by TSH receptor dysfunction is probed. The complexes display a visible spectral band within the PDT spectral window at ~690 nm. Photodynamic potential was estimated through DNA cleavage activity of the dpq and dppz complexes in UV-A light of 365 nm and red light of 676 nm. The reactions proceed via the hydroxyl radical pathway. The complexes retain their DNA photocleavage activity in red light under anaerobic conditions, a situation normally prevails in hypoxic tumor core. Investigation into the photocytotoxic potential of these complexes showed that the dppz complex 3 is approximately 4-fold more active in the HEK293 cells expressing human thyrotropin receptor (HEK293-hTSHR) than in the parental cell line and has an insignificant effect on an unrelated human cervical carcinoma cell line (HeLa). Photoexcitation of complex 3 in HEK293-hTSHR cells leads to damage hTSHR as evidenced from the decrease in cAMP formation both in absence and presence of hTSH and decrease in the TSHR immunofluorescence with a concomitant cytoplasmic translocation of the membrane protein, cadherin. The involvement of hTSHR is evidenced from the ability of complex 3 to bind to the extracellular domain of hTSHR (hTSHR-ECD) with a K(d) value of 81 nM and from the photocleavage of hTSHR-ECD.
Collapse
Affiliation(s)
- Sounik Saha
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | | | | | | |
Collapse
|
34
|
Bible KC, Suman VJ, Molina JR, Smallridge RC, Maples WJ, Menefee ME, Rubin J, Sideras K, Morris JC, McIver B, Burton JK, Webster KP, Bieber C, Traynor AM, Flynn PJ, Goh BC, Tang H, Ivy SP, Erlichman C. Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: results of a phase 2 consortium study. Lancet Oncol 2010; 11:962-72. [PMID: 20851682 PMCID: PMC3107731 DOI: 10.1016/s1470-2045(10)70203-5] [Citation(s) in RCA: 313] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chemotherapy has historically proven ineffective in advanced differentiated thyroid cancers, but the realisation that various tyrosine kinases are activated in the disease suggested a potential therapeutic role for tyrosine-kinase inhibitors. We investigated the safety and efficacy of pazopanib. METHODS This phase 2 trial was done from Feb 22, 2008, to Jan 31, 2009, in patients with metastatic, rapidly progressive, radioiodine-refractory differentiated thyroid cancers. Each patient received 800 mg continuous pazopanib daily in 4-week cycles until disease progression, drug intolerance, or both occurred. Up to two previous therapies were allowed, and measurable disease with radiographic progression in the 6-month period before enrolment was a requirement for inclusion. The primary endpoint was any tumour response, according to the Response Evaluation Criteria in Solid Tumors 1.0. This study is registered with ClinicalTrials.gov, number NCT00625846. FINDINGS 39 patients were enrolled. One patient had received no previous radioiodine therapy and another withdrew consent before treatment. Clinical outcomes could, therefore, be assessed in 37 patients (19 [51%] men, median age 63 years). The study is closed to accrual of new patients, but several enrolled patients are still being treated. Patients received a median of 12 cycles (range 1 to >23, total >383). Confirmed partial responses were recorded in 18 patients (response rate 49%, 95% CI 35-68), with likelihood of response lasting longer than 1 year calculated to be 66%. Maximum concentration of pazopanib in plasma during cycle one was significantly correlated with radiographic response (r=-0·40, p=0·021). 16 (43%) patients required dose reductions owing to adverse events, the most frequent of which (any grade) were fatigue (29 patients), skin and hair hypopigmentation (28), diarrhoea (27), and nausea (27). Two patients who died during treatment had pre-existing contributory disorders. INTERPRETATION Pazopanib seems to represent a promising therapeutic option for patients with advanced differentiated thyroid cancers. The correlation of the patient's response and pazopanib concentration during the first cycle might indicate that treatment can be individualised to achieve optimum outcomes. Assessment of pazopanib in an expanded cohort of patients with differentiated thyroid cancer, as well as in cohorts of patients with medullary and anaplastic thyroid cancers, is presently being done. FUNDING National Cancer Institute, supported in part by NCI CA15083 and CM62205.
Collapse
Affiliation(s)
- Keith C Bible
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Martinelli J, Balali-Mood B, Panizzo R, Lythgoe MF, White AJP, Ferretti P, Steinke JHG, Vilar R. Coordination chemistry of amide-functionalised tetraazamacrocycles: structural, relaxometric and cytotoxicity studies. Dalton Trans 2010; 39:10056-67. [PMID: 20877892 DOI: 10.1039/c0dt00815j] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Three different tetraazamacrocyclic ligands containing four amide substituents that feature groups (namely allyl, styryl and propargyl groups) suitable for polymerisation have been synthesised. Gadolinium(III) complexes of these three ligands have been prepared as potential monomers for the synthesis of polymeric MRI contrast agents. To assess the potential of these monomers as MRI contrast agents, their relaxation enhancement properties and cytotoxicity have been determined. A europium(III) complex of one of these ligands (with propargyl substituents) is also presented together with its PARACEST properties. In addition, to gain further insight into the coordination chemistry of the tetra-propargyl substituted ligand, the corresponding zinc(II) and cadmium(II) complexes have been prepared. The X-ray crystal structures of the tetra-propargyl ligand and its corresponding gadolinium(III), zinc(II) and cadmium(II) complexes are also presented.
Collapse
|
36
|
Middendorp M, Grünwald F. Update on recent developments in the therapy of differentiated thyroid cancer. Semin Nucl Med 2010; 40:145-52. [PMID: 20113682 DOI: 10.1053/j.semnuclmed.2009.10.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
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.
Collapse
Affiliation(s)
- Marcus Middendorp
- Department of Nuclear Medicine, Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | | |
Collapse
|
37
|
Sughrue ME, Kane AJ, Shangari G, Parsa AT, Berger MS, McDermott MW. Prevalence of previous extracranial malignancies in a series of 1228 patients presenting with meningioma. J Neurosurg 2010; 113:1115-21. [PMID: 20433279 DOI: 10.3171/2010.3.jns091975] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECT The study of patients with multiple neoplasms can yield valuable insight into the common pathogenesis of both diseases, as well as identify more subtle risk factors that might not be as readily apparent otherwise. The authors present an analysis of the prevalence of previously diagnosed extracranial malignancies at the time of meningioma diagnosis in 1228 patients evaluated at a single institution. METHODS All patients who underwent evaluation and/or treatment for meningioma between 1991 and 2007 at the authors' institution were identified. The intake history and physical were assessed for any history of extracranial malignancy. Using the National Cancer Institute data, the authors calculated an expected cancer prevalence for their meningioma patient population, and compared this derived value to the observed rate of these cancers in this population. RESULTS There were 1228 patients included in this study. A total of 50 patients (4.1%) with newly diagnosed meningioma had a history of an extracranial malignant tumor at the time of their initial meningioma diagnosis. In general, most malignancies did not differ in prevalence from their expected frequency in the population in the present study. Notable exceptions were acute leukemia (p < 0.0001), and papillary thyroid carcinoma, which had a prevalence 2.5 times that expected in this population (p < 0.05). CONCLUSIONS The data support a growing body of evidence suggesting an epidemiological link between papillary carcinoma of the thyroid and meningioma. Although the link between these tumors is not immediately apparent, it is possible that further exploration will yield interesting insight into the pathogenesis of both diseases.
Collapse
Affiliation(s)
- Michael E Sughrue
- Brain Tumor Research Center, Department of Neurological Surgery, University of California, San Francisco, California 94143, USA
| | | | | | | | | | | |
Collapse
|
38
|
Pinto AE, Leite V, Soares J. Clinical implications of molecular markers in follicular cell-derived thyroid cancer. Expert Rev Mol Diagn 2009; 9:679-94. [PMID: 19817553 DOI: 10.1586/erm.09.54] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The increasing use/applications of molecular biology techniques have provided new insights on the genetic changes that underlie carcinogenesis and tumor progression in thyroid cancer. Molecular analysis may improve the histopathologic evaluation of follicular cell-derived thyroid carcinoma, not only elucidating some unresolved problems related to the diagnosis and disease prognosis, but also by improving patient management. Besides increasing our comprehension of cancer biology, either genetic alterations or gene expression profiles implicated in thyroid carcinogenesis shed new light on innovative diagnostic procedures as well as on targeted therapies.
Collapse
Affiliation(s)
- António E Pinto
- Serviço de Anatomia Patológica, Instituto Português de Oncologia de Lisboa, EPE, Rua Professor Lima Basto, 1099-023 Lisbon, Portugal.
| | | | | |
Collapse
|