1
|
Kunte SC, Wenter V, Toms J, Lindner S, Unterrainer M, Eilsberger F, Jurkschat K, Wängler C, Wängler B, Schirrmacher R, Tiling MW, Sheikh GT, Mehrens D, Brendel M, Rübenthaler J, Auernhammer CJ, Spitzweg C, Unterrainer LM, Holzgreve A. PET/CT imaging of differentiated and medullary thyroid carcinoma using the novel SSTR-targeting peptide [ 18F]SiTATE - first clinical experiences. Eur J Nucl Med Mol Imaging 2025; 52:900-912. [PMID: 39404789 PMCID: PMC11754387 DOI: 10.1007/s00259-024-06944-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/06/2024] [Indexed: 01/23/2025]
Abstract
PURPOSE The novel 18F-labeled somatostatin receptor (SSTR)-directed radiotracer [18F]SiTATE demonstrated promising results for the imaging of various SSTR-expressing tumor types. Although thyroid carcinomas (TC) express SSTR, data on [18F]SiTATE PET/CT imaging in TC are lacking. This study explores the use of [18F]SiTATE PET/CT in a patient cohort with histologically proven TC. METHODS As part of a prospective observational study at a single tertiary cancer center, 21 patients with TC (10 medullary (MTC) and 11 differentiated (DTC)) who underwent at least one [18F]SiTATE PET/CT were included (37 scans in total). Mean SUVmax and SUVmean of tumoral lesions, mean total-tumor-volume (TTV), and whole-body (WB)-SUVmax and WB-SUVmean on PET with their standard deviations (SDs) were determined. PET parameters were correlated to clinical parameters including tumor marker levels (thyroglobulin for DTC, calcitonin for MTC). RESULTS 89 lesions were included in the analysis. Metastases were localized in the bone, lymph nodes, lung, soft tissue, and thyroid bed. Osseous (31 lesions; SUVmax 8.6 ± 8.0; SUVmean 5.8 ± 5.4) and nodal (37 lesions; SUVmax 8.7 ± 7.8; SUVmean 5.7 ± 5.4) metastases showed the highest uptake. The MTC disease burden on PET significantly correlated with the calcitonin tumor marker level (e.g., TTV: r = 0.771, r2 = 0.594, p = 0.002). For DTC, no such correlation was present. CONCLUSION Our data demonstrate high feasibility of [18F]SiTATE PET/CT in a small cohort of patients with MTC and DTC. The use of [18F]SiTATE may overcome logistical disadvantages of 68Ga-based tracers and facilitate SSTR-targeted PET/CT imaging of thyroid carcinoma.
Collapse
Affiliation(s)
- Sophie C Kunte
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Vera Wenter
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Johannes Toms
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- DIE RADIOLOGIE, Munich, Germany
| | - Friederike Eilsberger
- Department of Nuclear Medicine, School of Medicine, Philipps University Marburg, Marburg, Germany
| | - Klaus Jurkschat
- Fakultät für Chemie und Chemische Biologie, Technische Universität Dortmund, Dortmund, Germany
| | - Carmen Wängler
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
- Research Campus M²OLIE, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Björn Wängler
- Research Campus M²OLIE, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim of Heidelberg University, Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Mannheim, Germany
| | - Ralf Schirrmacher
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB, Canada
| | - Maximilian W Tiling
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Gabriel T Sheikh
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Dirk Mehrens
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership Between DKFZ and Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | | | | | - Christine Spitzweg
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena M Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Ahmanson Translational Theranostics Division, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Bayerisches Zentrum für Krebsforschung (BZKF), Partner Site Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- Ahmanson Translational Theranostics Division, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Viering O, Rinscheid A, Holzleitner N, Dierks A, Kircher M, Wienand G, Patt M, Wester HJ, Bundschuh RA, Günther T, Lapa C, Pfob CH. Biodistribution and Radiation Dosimetry for 68 Ga-DOTA-CCK-66, a Novel CCK 2 R-Targeting Compound for Imaging of Medullary Thyroid Cancer. Clin Nucl Med 2024; 49:1091-1097. [PMID: 39093043 DOI: 10.1097/rlu.0000000000005355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
ABSTRACT Cholecystokinin 2 receptor (CCK 2 R) is a promising target for imaging and treatment of medullary thyroid cancer due to its overexpression in over 90% of tumor cells. 68 Ga-DOTA-CCK-66 is a recently introduced PET tracer selective for CCK 2 R, which has shown favorable pharmacokinetics in vivo in preclinical experiments. In order to further investigate safety and suitability of this tracer in the human setting, whole-body distribution and radiation dosimetry were evaluated. PATIENTS AND METHODS Six patients with a history of medullary thyroid cancer were injected intravenously with 169 ± 19 MBq of 68 Ga-DOTA-CCK-66. Whole-body PET/CT scans were acquired at 10 minutes, 1 hour, 2 hours, and 4 hours after tracer injection. Time-activity curves per organ were determined, and mean organ-absorbed doses and effective doses were calculated using OLINDA/EXM. RESULTS Injection of a standard activity of 150 MBq of 68 Ga-DOTA-CCK-66 results in an effective dose of 4.5 ± 0.9 mSv. The highest absorbed organ doses were observed in the urinary bladder wall (40 mGy) and the stomach (15 mGy), followed by the kidneys (6 mGy), as well as the liver and the spleen (3 mGy each). CCK 2 R-expressing tumor manifestations could be detected in 2 of the 6 patients, including lymph node, bone, and liver metastases. CONCLUSIONS 68 Ga-DOTA-CCK-66 exhibits a favorable dosimetry. Beyond physiologic receptor expression of the stomach, no other relevant tracer accumulation could be observed, rendering this organ at risk in case of subsequent radioligand therapy using 177 Lu-DOTA-CCK-66.
Collapse
Affiliation(s)
- Oliver Viering
- From the Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Andreas Rinscheid
- Medical Physics and Radiation Protection, University Hospital Augsburg, Augsburg, Germany
| | - Nadine Holzleitner
- TUM School of Natural Sciences, Department of Chemistry, Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Alexander Dierks
- From the Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Malte Kircher
- From the Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Georgine Wienand
- From the Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Marianne Patt
- From the Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Hans-Jürgen Wester
- TUM School of Natural Sciences, Department of Chemistry, Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Ralph A Bundschuh
- From the Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | | | | | - Christian H Pfob
- From the Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| |
Collapse
|
3
|
de Andrade FA, Bulzico D, Corbo R, Vaisman F. Is peptide receptor radionuclide therapy still a promising option for medullary thyroid carcinoma? Endocrine 2024:10.1007/s12020-024-04114-6. [PMID: 39609369 DOI: 10.1007/s12020-024-04114-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/13/2024] [Indexed: 11/30/2024]
Abstract
Medullary thyroid carcinoma (MTC) is a rare cancer that originates from germline RET proto-oncogene mutations in all hereditary forms and from somatic RET mutations in most sporadic cases. Currently, highly selective RET inhibitors have been approved for clinical use in patients with RET mutations with persistent, recurrent or metastatic disease. This therapy has proven efficacy, low toxicity, and a limited impact on patients' quality of life. However, for recurrent or metastatic RET-negative disease, few systemic therapies are available. Multikinase inhibitors are used; however, tumour cells frequently develop resistance mechanisms, or treatment must be discontinued due to the high incidence of side effects. In this context, peptide receptor radionuclide therapy (PRRT) may be a treatment option, but its clinical utility remains under investigation. The aim of this review is to evaluate the evidence of PRRT in MTC and discuss its limitations in the RET inhibitor era.
Collapse
Affiliation(s)
| | - Daniel Bulzico
- Endocrine Oncology Unit, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil
- Nuclear Medicine Section, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil
| | - Rossana Corbo
- Endocrine Oncology Unit, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil
- Nuclear Medicine Section, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil
| | - Fernanda Vaisman
- Endocrine Oncology Unit, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil.
- Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.
| |
Collapse
|
4
|
Luvhengo TE, Moeng MS, Sishuba NT, Makgoka M, Jonas L, Mamathuntsha TG, Mbambo T, Kagodora SB, Dlamini Z. Holomics and Artificial Intelligence-Driven Precision Oncology for Medullary Thyroid Carcinoma: Addressing Challenges of a Rare and Aggressive Disease. Cancers (Basel) 2024; 16:3469. [PMID: 39456563 PMCID: PMC11505703 DOI: 10.3390/cancers16203469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objective: Medullary thyroid carcinoma (MTC) is a rare yet aggressive form of thyroid cancer comprising a disproportionate share of thyroid cancer-related mortalities, despite its low prevalence. MTC differs from other differentiated thyroid malignancies due to its heterogeneous nature, presenting complexities in both hereditary and sporadic cases. Traditional management guidelines, which are designed primarily for papillary thyroid carcinoma (PTC), fall short in providing the individualized care required for patients with MTC. In recent years, the sheer volume of data generated from clinical evaluations, radiological imaging, pathological assessments, genetic mutations, and immunological profiles has made it humanly impossible for clinicians to simultaneously analyze and integrate these diverse data streams effectively. This data deluge necessitates the adoption of advanced technologies to assist in decision-making processes. Holomics, which is an integrated approach that combines various omics technologies, along with artificial intelligence (AI), emerges as a powerful solution to address these challenges. Methods: This article reviews how AI-driven precision oncology can enhance the diagnostic workup, staging, risk stratification, management, and follow-up care of patients with MTC by processing vast amounts of complex data quickly and accurately. Articles published in English language and indexed in Pubmed were searched. Results: AI algorithms can identify patterns and correlations that may not be apparent to human clinicians, thereby improving the precision of personalized treatment plans. Moreover, the implementation of AI in the management of MTC enables the collation and synthesis of clinical experiences from across the globe, facilitating a more comprehensive understanding of the disease and its treatment outcomes. Conclusions: The integration of holomics and AI in the management of patients with MTC represents a significant advancement in precision oncology. This innovative approach not only addresses the complexities of a rare and aggressive disease but also paves the way for global collaboration and equitable healthcare solutions, ultimately transforming the landscape of treatment and care of patients with MTC. By leveraging AI and holomics, we can strive toward making personalized healthcare accessible to every individual, regardless of their economic status, thereby improving overall survival rates and quality of life for MTC patients worldwide. This global approach aligns with the United Nations Sustainable Development Goal 3, which aims to ensure healthy lives and promote well-being at all ages.
Collapse
Affiliation(s)
| | - Maeyane Stephens Moeng
- Department of Surgery, University of the Witwatersrand, Johannesburg 2193, South Africa; (M.S.M.); (N.T.S.)
| | - Nosisa Thabile Sishuba
- Department of Surgery, University of the Witwatersrand, Johannesburg 2193, South Africa; (M.S.M.); (N.T.S.)
| | - Malose Makgoka
- Department of Surgery, University of Pretoria, Pretoria 0002, South Africa;
| | - Lusanda Jonas
- Department of Surgery, University of Limpopo, Mankweng 4062, South Africa; (L.J.); (T.G.M.)
| | | | - Thandanani Mbambo
- Department of Surgery, University of KwaZulu-Natal, Durban 2025, South Africa;
| | | | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI, Precision Oncology and Cancer Prevention (POCP), University of Pretoria, Pretoria 0028, South Africa;
| |
Collapse
|
5
|
Zhang Y, Zheng WH, Zhou SH, Gu JL, Yu Q, Zhu YZ, Yan YJ, Zhu Z, Shang JB. Molecular genetics, therapeutics and RET inhibitor resistance for medullary thyroid carcinoma and future perspectives. Cell Commun Signal 2024; 22:460. [PMID: 39342195 PMCID: PMC11439284 DOI: 10.1186/s12964-024-01837-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Medullary thyroid carcinoma (MTC) is a rare type of thyroid malignancy that accounts for approximately 1-2% of all thyroid cancers (TCs). MTC include hereditary and sporadic cases, the former derived from a germline mutation of rearrangement during transfection (RET) proto-oncogene, whereas somatic RET mutations are frequently present in the latter. Surgery is the standard treatment for early stage MTC, and the 10-year survival rate of early MTC is over 80%. While for metastatic MTC, chemotherapy showing low response rate, and there was a lack of effective systemic therapies in the past. Due to the high risk (ca. 15-20%) of distant metastasis and limited systemic therapies, the 10-year survival rate of patients with advanced MTC was only 10-40% from the time of first metastasis. Over the past decade, targeted therapy for RET has developed rapidly, bringing hopes to patients with advanced and progressive MTC. Two multi-kinase inhibitors (MKIs) including Cabozantinib and Vandetanib have been shown to increase progression-free survival (PFS) for patients with metastatic MTC and have been approved as choices of first-line treatment. However, these MKIs have not prolonged overall survival (OS) and their utility is limited due to high rates of off-target toxicities. Recently, new generation TKIs, including Selpercatinib and Pralsetinib, have demonstrated highly selective efficacy against RET and more favorable side effect profiles, and gained approval as second-line treatment options. Despite the ongoing development of RET inhibitors, the management of advanced and progressive MTC remains challenging, drug resistance remains the main reason for treatment failure, and the mechanisms are still unclear. Besides, new promising therapeutic approaches, such as novel drug combinations and next generation RET inhibitors are under development. Herein, we overview the pathogenesis, molecular genetics and current management approaches of MTC, and focus on the recent advances of RET inhibitors, summarize the current situation and unmet needs of these RET inhibitors in MTC, and provide an overview of novel strategies for optimizing therapeutic effects.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wei-Hui Zheng
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shi-Hong Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jia-Lei Gu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, Zhejiang, China
| | - Qing Yu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yi-Zhou Zhu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yu-Jie Yan
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Zhi Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Jin-Biao Shang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China.
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Giovanella L, Tuncel M, Aghaee A, Campenni A, De Virgilio A, Petranović Ovčariček P. Theranostics of Thyroid Cancer. Semin Nucl Med 2024; 54:470-487. [PMID: 38503602 DOI: 10.1053/j.semnuclmed.2024.01.011] [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: 01/13/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/21/2024]
Abstract
Molecular imaging is pivotal in evaluating and managing patients with different thyroid cancer histotypes. The existing, pathology-based, risk stratification systems can be usefully refined, by incorporating tumor-specific molecular and molecular imaging biomarkers with theranostic value, allowing patient-specific treatment decisions. Molecular imaging with different radioactive iodine isotopes (ie, I131, I123, I124) is a central component of differentiated carcinoma (DTC)'s risk stratification while [18F]F-fluorodeoxyglucose ([18F]FDG) PET/CT is interrogated about disease aggressiveness and presence of distant metastases. Moreover, it is particularly useful to assess and risk-stratify patients with radioiodine-refractory DTC, poorly differentiated, and anaplastic thyroid cancers. [18F]F-dihydroxyphenylalanine (6-[18F]FDOPA) PET/CT is the most specific and accurate molecular imaging procedure for patients with medullary thyroid cancer (MTC), a neuroendocrine tumor derived from thyroid C-cells. In addition, [18F]FDG PET/CT can be used in patients with more aggressive clinical or biochemical (ie, serum markers levels and kinetics) MTC phenotypes. In addition to conventional radioiodine therapy for DTC, new redifferentiation strategies are now available to restore uptake in radioiodine-refractory DTC. Moreover, peptide receptor theranostics showed promising results in patients with advanced and metastatic radioiodine-refractory DTC and MTC, respectively. The current appropriate role and future perspectives of molecular imaging and theranostics in thyroid cancer are discussed in our present review.
Collapse
Affiliation(s)
- Luca Giovanella
- Department of Nuclear Medicine, Gruppo Ospedaliero Moncucco, Lugano, Switzerland; Clinic for Nuclear Medicine, University Hospital Zürich, Zürich, Switzerland.
| | - Murat Tuncel
- Department of Nuclear Medicine, Hacettepe University, Ankara, Turkey
| | - Atena Aghaee
- Department of Nuclear Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alfredo Campenni
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| | - Armando De Virgilio
- Department of Head and Neck Surgery Humanitas Research Hospital, Rozzano, Italy
| | - Petra Petranović Ovčariček
- Department of Oncology and Nuclear Medicine, University Hospital Center Sestre Milosrdnice, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
7
|
Hassan M, Bokhari TH, Lodhi NA, Khosa MK, Usman M. A review of recent advancements in Actinium-225 labeled compounds and biomolecules for therapeutic purposes. Chem Biol Drug Des 2023; 102:1276-1292. [PMID: 37715360 DOI: 10.1111/cbdd.14311] [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: 03/16/2022] [Revised: 06/03/2023] [Accepted: 07/17/2023] [Indexed: 09/17/2023]
Abstract
In nuclear medicine, cancers that cannot be cured or can only be treated partially by traditional techniques like surgery or chemotherapy are killed by ionizing radiation as a form of therapeutic treatment. Actinium-225 is an alpha-emitting radionuclide that is highly encouraging as a therapeutic approach and more promising for targeted alpha therapy (TAT). Actinium-225 is the best candidate for tumor cells treatment and has physical characteristics such as high (LET) linear energy transfer (150 keV per μm), half-life (t1/2 = 9.92d), and short ranges (400-100 μm) which prevent the damage of normal healthy tissues. The introduction of various new radiopharmaceuticals and radioisotopes has significantly assisted the advancement of nuclear medicine. Ac-225 radiopharmaceuticals continuously demonstrate their potential as targeted alpha therapeutics. 225 Ac-labeled radiopharmaceuticals have confirmed their importance in medical and clinical areas by introducing [225 Ac]Ac-PSMA-617, [225 Ac]Ac-DOTATOC, [225 Ac]Ac-DOTA-substance-P, reported significantly improved response in patients with prostate cancer, neuroendocrine, and glioma, respectively. The development of these radiopharmaceuticals required a suitable buffer, incubation time, optimal pH, and reaction temperature. There is a growing need to standardize quality control (QC) testing techniques such as radiochemical purity (RCP). This review aims to summarize the development of the Ac-225 labeled compounds and biomolecules. The current state of their reported resulting clinical applications is also summarized as well.
Collapse
Affiliation(s)
- Maria Hassan
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | | | - Nadeem Ahmed Lodhi
- Isotope Production Division, Pakistan institute of Nuclear Science & Technology (PINSTECH), Islamabad, Pakistan
| | | | - Muhammad Usman
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| |
Collapse
|
8
|
Liu Q, Kulkarni HR, Zhao T, Schuchardt C, Chen X, Zhu Z, Zhang J, Baum RP. Peptide Receptor Radionuclide Therapy in Patients With Advanced Progressive Medullary Thyroid Cancer: Efficacy, Safety, and Survival Predictors. Clin Nucl Med 2023; 48:221-227. [PMID: 36723881 DOI: 10.1097/rlu.0000000000004539] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE Progressive metastatic medullary thyroid carcinoma (MTC) is often characterized by rapid disease progression and poor prognosis, with only few therapeutic options available. Peptide receptor radionuclide therapy (PRRT) has demonstrated remarkable success in the management of gastroenteropancreatic neuroendocrine tumors and has also been suggested to treat MTC. However, evidence on its effectiveness and long-term outcome for this indication is still limited. The objective of this study was to assess the safety and efficacy of PRRT in patients with advanced, progressive MTC and to determine survival. Potential predictors of survival were also evaluated. METHODS From September 2003 to June 2019, 28 patients (15 men and 13 women; mean age, 49 ± 14 years) with progressive, somatostatin receptor-positive advanced MTC received PRRT with 177Lu- or 90Y-labeled somatostatin analogs at Zentralklinik Bad Berka, Germany. Toxicity was graded according to Common Terminology Criteria for Adverse Events version 5.0. Treatment response was evaluated according to RECIST (Response Evaluation Criteria in Solid Tumors) 1.1, as well as molecular imaging criteria (European Organisation for Research and Treatment of Cancer). Kaplan-Meier analysis was used to calculate progression-free survival (PFS) and overall survival (OS), defined from the start of PRRT. Univariate and multivariate Cox regression analyses were performed to identify parameters associated with PFS and OS. RESULTS Seventy-seven cycles of PRRT were administered (mean cumulative administered activity, 16.0 ± 7.8 GBq). No acute or long-term grade 3/4 toxicity was recorded with a follow-up of 3 to 140 months, except for 1 patient (4%) who suffered from grade 3 anemia (possibly related to disease progression). According to the RECIST criteria, the disease control rate after 3 to 4 months of PRRT was 56% (partial remission, 12%; stable disease, 44%). The disease control rate (72%) was higher by molecular response evaluation. Median OS and PFS were 63.7 and 10.1 months, respectively. The annual OS rates were 84% at 1 year, 65% at 3 years, 57% at 5 years, and 18% at 10 years. The annual PFS rates were 42% at 1 year, 21% at 2 years, and 13% at 5 years. Patients with bone metastases had poorer OS and PFS than those without metastases (median OS, 58.7 vs 92.3 months [P = 0.035; hazard ratio, 2.7; 95% confidence interval, 0.92-7.84]; median PFS, 8.5 vs 12.8 months [P = 0.592; hazard ratio, 1.2; 95% confidence interval, 0.56-2.76]). CONCLUSIONS Peptide receptor radionuclide therapy was well tolerated and effective in patients with advanced, aggressive MTC. Bone metastasis was an independent adverse prognostic factor for OS.
Collapse
Affiliation(s)
| | - Harshad R Kulkarni
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| | | | - Christiane Schuchardt
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| | | | - Zhaohui Zhu
- From the Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing
| | | | - Richard P Baum
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Wiesbaden, Germany
| |
Collapse
|
9
|
Lezaic L, Erba PA, Decristoforo C, Zaletel K, Mikolajczak R, Maecke H, Maina T, Konijnenberg M, Kolenc P, Trofimiuk-Müldner M, Przybylik-Mazurek E, Virgolini I, de Jong M, Fröberg AC, Rangger C, Di Santo G, Skorkiewicz K, Garnuszek P, Solnica B, Nock BA, Fedak D, Gaweda P, Hubalewska-Dydejczyk A. [ 111In]In-CP04 as a novel cholecystokinin-2 receptor ligand with theranostic potential in patients with progressive or metastatic medullary thyroid cancer: final results of a GRAN-T-MTC Phase I clinical trial. Eur J Nucl Med Mol Imaging 2023; 50:892-907. [PMID: 36334104 PMCID: PMC9852173 DOI: 10.1007/s00259-022-05992-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Medullary thyroid cancer (MTC) is a rare malignant tumour of the parafollicular C-cells with an unpredictable clinical course and currently suboptimal diagnostic and therapeutic options, in particular in advanced disease. Overexpression of cholecystokinin-2 receptors (CCK2R) represents a promising avenue to diagnostic imaging and targeted therapy, ideally through a theranostic approach. MATERIALS AND METHODS A translational study (GRAN-T-MTC) conducted through a Phase I multicentre clinical trial of the indium-111 labelled CP04 ([111In]In-CP04), a CCK2R-seeking ligand was initiated with the goal of developing a theranostic compound. Patients with proven advanced/metastatic MTC or short calcitonin doubling time were enrolled. A two-step concept was developed through the use of low- and high-peptide mass (10 and 50 μg, respectively) for safety assessment, with the higher peptide mass considered appropriate for therapeutic application. Gelofusine was co-infused in a randomized fashion in the second step for the evaluation of potential reduction of the absorbed dose to the kidneys. Imaging for the purpose of biodistribution, dosimetry evaluation, and diagnostic assessment were performed as well as pre-, peri-, and postprocedural clinical and biochemical assessment. RESULTS Sixteen patients were enrolled. No serious adverse events after application of the compound at both peptide amounts were witnessed; transient tachycardia and flushing were observed in two patients. No changes in biochemistry and clinical status were observed on follow-up. Preliminary dosimetry assessment revealed the highest dose to urinary bladder, followed by the kidneys and stomach wall. The effective dose for 200 MBq of [111In]In-CP04 was estimated at 7±3 mSv and 7±1 mSv for 10 μg and 50 μg CP04, respectively. Administration of Gelofusine reduced the dose to the kidneys by 53%, resulting in the organ absorbed dose of 0.044±0.019 mSv/MBq. Projected absorbed dose to the kidneys with the use of [177Lu]Lu-CP04 was estimated at 0.9±0.4 Gy/7.4 GBq. [111In]In-CP04 scintigraphy was positive in 13 patients (detection rate of 81%) with superior diagnostic performance over conventional imaging. CONCLUSION In the present study, [111In]In-CP04 was shown to be a safe and effective radiopharmaceutical with promising theranostic characteristics for patients with advanced MTC.
Collapse
Affiliation(s)
- Luka Lezaic
- University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Paola Anna Erba
- Regional Center of Nuclear Medicine, Department of Translational Research and New Technology in Medicine, University of Pisa, and Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Katja Zaletel
- University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Renata Mikolajczak
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, Otwock-Świerk, Poland
| | | | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR Demokritos, Athens, Greece
| | | | - Petra Kolenc
- University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Malgorzata Trofimiuk-Müldner
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Jakubowskiego str. 2, 30-688, Krakow, Poland
| | - Elwira Przybylik-Mazurek
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Jakubowskiego str. 2, 30-688, Krakow, Poland
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | | | | | - Christine Rangger
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Gianpaolo Di Santo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | | | - Piotr Garnuszek
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, Otwock-Świerk, Poland
| | - Bogdan Solnica
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Berthold A Nock
- Molecular Radiopharmacy, INRASTES, NCSR Demokritos, Athens, Greece
| | - Danuta Fedak
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Paulina Gaweda
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Alicja Hubalewska-Dydejczyk
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Jakubowskiego str. 2, 30-688, Krakow, Poland.
| |
Collapse
|
10
|
Update on the Diagnosis and Management of Medullary Thyroid Cancer: What Has Changed in Recent Years? Cancers (Basel) 2022; 14:cancers14153643. [PMID: 35892901 PMCID: PMC9332800 DOI: 10.3390/cancers14153643] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Medullary thyroid carcinoma (MTC) is a neoplasm originating from parafollicular C cells. MTC is a rare disease, but its prognosis is less favorable than that of well-differentiated thyroid cancers. To improve the prognosis of patients with MTC, early diagnosis and prompt therapeutic management are crucial. In the following paper, recent advances in laboratory and imaging diagnostics and also pharmacological and surgical therapies of MTC are discussed. Currently, a thriving direction of development for laboratory diagnostics is immunohistochemistry. The primary imaging modality in the diagnosis of MTC is the ultrasound, but opportunities for development are seen primarily in nuclear medicine techniques. Surgical management is the primary method of treating MTCs. There are numerous publications concerning the stratification of particular lymph node compartments for removal. With the introduction of more effective methods of intraoperative parathyroid identification, the complication rate of surgical treatment may be reduced. The currently used pharmacotherapy is characterized by high toxicity. Moreover, the main limitation of current pharmacotherapy is the development of drug resistance. Currently, there is ongoing research on the use of tyrosine kinase inhibitors (TKIs), highly specific RET inhibitors, radiotherapy and immunotherapy. These new therapies may improve the prognosis of patients with MTCs.
Collapse
|
11
|
Angelousi A, Hayes AR, Chatzellis E, Kaltsas GA, Grossman AB. Metastatic medullary thyroid carcinoma: a new way forward. Endocr Relat Cancer 2022; 29:R85-R103. [PMID: 35521769 PMCID: PMC9175549 DOI: 10.1530/erc-21-0368] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/26/2022] [Indexed: 11/21/2022]
Abstract
Medullary thyroid carcinoma (MTC) is a rare malignancy comprising 1-2% of all thyroid cancers in the United States. Approximately 20% of cases are familial, secondary to a germline RET mutation, while the remaining 80% are sporadic and also harbour a somatic RET mutation in more than half of all cases. Up to 15-20% of patients will present with distant metastatic disease, and retrospective series report a 10-year survival of 10-40% from time of first metastasis. Historically, systemic therapies for metastatic MTC have been limited, and cytotoxic chemotherapy has demonstrated poor objective response rates. However, in the last decade, targeted therapies, particularly multitargeted tyrosine kinase inhibitors (TKIs), have demonstrated prolonged progression-free survival in advanced and progressive MTC. Both cabozantinib and vandetanib have been approved as first-line treatment options in many countries; nevertheless, their use is limited by high toxicity rates and dose reductions are often necessary. New generation TKIs, such as selpercatinib or pralsetinib, that exhibit selective activity against RET, have recently been approved as a second-line treatment option, and they exhibit a more favourable side-effect profile. Peptide receptor radionuclide therapy or immune checkpoint inhibitors may also constitute potential therapeutic options in specific clinical settings. In this review, we aim to present all current therapeutic options available for patients with progressive MTC, as well as new or as yet experimental treatments.
Collapse
Affiliation(s)
- Anna Angelousi
- Unit of Endocrinology, First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
- Correspondence should be addressed to A Angelousi or A B Grossman: or
| | - Aimee R Hayes
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, UK
| | - Eleftherios Chatzellis
- Endocrinology Diabetes and Metabolism Department, 251 Hellenic Air Force and VA General Hospital, Athens, Greece
| | - Gregory A Kaltsas
- First Department of Propaedeutic Internal Medicine, Laiko Hospital, National & Kapodistrian University of Athens, Athens, Greece
| | - Ashley B Grossman
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, UK
- Green Templeton College, University of Oxford, Oxford, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
- Correspondence should be addressed to A Angelousi or A B Grossman: or
| |
Collapse
|
12
|
Giovanella L, Deandreis D, Vrachimis A, Campenni A, Petranovic Ovcaricek P. Molecular Imaging and Theragnostics of Thyroid Cancers. Cancers (Basel) 2022; 14:1272. [PMID: 35267580 PMCID: PMC8909041 DOI: 10.3390/cancers14051272] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Molecular imaging plays an important role in the evaluation and management of different thyroid cancer histotypes. The existing risk stratification models can be refined, by incorporation of tumor-specific molecular markers that have theranostic power, to optimize patient-specific (individualized) treatment decisions. Molecular imaging with varying radioisotopes of iodine (i.e., 131I, 123I, 124I) is an indispensable component of dynamic and theragnostic risk stratification of differentiated carcinoma (DTC) while [18F]F-fluorodeoxyglucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) helps in addressing disease aggressiveness, detects distant metastases, and risk-stratifies patients with radioiodine-refractory DTC, poorly differentiated and anaplastic thyroid cancers. For medullary thyroid cancer (MTC), a neuroendocrine tumor derived from thyroid C-cells, [18F]F-dihydroxyphenylalanine (6-[18F]FDOPA) PET/CT and/or [18F]FDG PET/CT can be used dependent on serum markers levels and kinetics. In addition to radioiodine therapy for DTC, some theragnostic approaches are promising for metastatic MTC as well. Moreover, new redifferentiation strategies are now available to restore uptake in radioiodine-refractory DTC while new theragnostic approaches showed promising preliminary results for advanced and aggressive forms of follicular-cell derived thyroid cancers (i.e., peptide receptor radiotherapy). In order to help clinicians put the role of molecular imaging into perspective, the appropriate role and emerging opportunities for molecular imaging and theragnostics in thyroid cancer are discussed in our present review.
Collapse
Affiliation(s)
- Luca Giovanella
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Desiree’ Deandreis
- Division of Nuclear Medicine, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10126 Turin, Italy;
| | - Alexis Vrachimis
- Department of Nuclear Medicine, German Oncology Center, University Hospital of the European University, Limassol 4108, Cyprus;
| | - Alfredo Campenni
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98121 Messina, Italy;
| | - Petra Petranovic Ovcaricek
- Department of Oncology and Nuclear Medicine, University Hospital Center Sestre Milosrdnice, 10000 Zagreb, Croatia;
| |
Collapse
|
13
|
Naik M, Al-Nahhas A, Khan SR. Treatment of Neuroendocrine Neoplasms with Radiolabeled Peptides-Where Are We Now. Cancers (Basel) 2022; 14:761. [PMID: 35159027 PMCID: PMC8833798 DOI: 10.3390/cancers14030761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has been one of the most successful and exciting examples of theranostics in nuclear medicine in recent decades and is now firmly embedded in many treatment algorithms for unresectable or metastatic neuroendocrine neoplasms (NENs) worldwide. It is widely considered to be an effective treatment for well- or moderately differentiated neoplasms, which express high levels of somatostatin receptors that can be selectively targeted. This review article outlines the scientific basis of PRRT in treatment of NENs and describes its discovery dating back to the early 1990s. Early treatments utilizing Indium-111, a γ-emitter, showed promise in reduction in tumor size and improvement in biochemistry, but were also met with high radiation doses and myelotoxic and nephrotoxic effects. Subsequently, stable conjugation of DOTA-peptides with β-emitting radionuclides, such as Yttrium-90 and Lutetium-177, served as a breakthrough for PRRT and studies highlighted their potential in eliciting progression-free survival and quality of life benefits. This article will also elaborate on the key trials which paved the way for its approval and will discuss therapeutic considerations, such as patient selection and administration technique, to optimize its use.
Collapse
Affiliation(s)
- Mitesh Naik
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| | | | - Sairah R. Khan
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| |
Collapse
|
14
|
Gubbi S, Koch CA, Klubo-Gwiezdzinska J. Peptide Receptor Radionuclide Therapy in Thyroid Cancer. Front Endocrinol (Lausanne) 2022; 13:896287. [PMID: 35712243 PMCID: PMC9197113 DOI: 10.3389/fendo.2022.896287] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/19/2022] [Indexed: 01/03/2023] Open
Abstract
The treatment options that are currently available for management of metastatic, progressive radioactive iodine (RAI)-refractory differentiated thyroid cancers (DTCs), and medullary thyroid cancers (MTCs) are limited. While there are several systemic targeted therapies, such as tyrosine kinase inhibitors, that are being evaluated and implemented in the treatment of these cancers, such therapies are associated with serious, sometimes life-threatening, adverse events. Peptide receptor radionuclide therapy (PRRT) has the potential to be an effective and safe modality for treating patients with somatostatin receptor (SSTR)+ RAI-refractory DTCs and MTCs. MTCs and certain sub-types of RAI-refractory DTCs, such as Hürthle cell cancers which are less responsive to conventional modalities of treatment, have demonstrated a favorable response to treatment with PRRT. While the current literature offers hope for utilization of PRRT in thyroid cancer, several areas of this field remain to be investigated further, especially head-to-head comparisons with other systemic targeted therapies. In this review, we provide a comprehensive outlook on the current translational and clinical data on the use of various PRRTs, including diagnostic utility of somatostatin analogs, theranostic properties of PRRT, and the potential areas for future research.
Collapse
Affiliation(s)
- Sriram Gubbi
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Christian A. Koch
- Department of Medicine, Fox Chase Cancer Center, Philadelphia, PA, United States
- Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Joanna Klubo-Gwiezdzinska
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Joanna Klubo-Gwiezdzinska,
| |
Collapse
|
15
|
Puerto M, Borson-Chazot F, Tabarin A. Updates on therapy for medullary thyroid cancer in 2021. ANNALES D'ENDOCRINOLOGIE 2021; 83:114-118. [PMID: 34921811 DOI: 10.1016/j.ando.2021.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 12/07/2021] [Indexed: 11/26/2022]
Abstract
Medullary thyroid cancer (MTC) is a rare form of thyroid cancer, frequently linked to a germline or somatic mutation in the RET proto-oncogene. MTC has a good prognosis at the localized stage but prognosis is worse in case of metastases, although there is considerable heterogeneity in progression even in advanced stages. Classical chemotherapy shows little efficacy in this type of cancer. Over the last decade, new effective anti-cancer therapies, in particular multi-targeted tyrosine kinase inhibitors and selective anti-RET tyrosine kinase inhibitors, have changed the management of patients with advanced MTC. The aim of this review is to report the results of studies of these new treatments, and to update the state of knowledge from ongoing studies of treatments such as vectorized internal radiotherapy. In chronic forms, which are incurable but with slow progression, the development of new lines of treatment that can reduce the phenomena of acquired resistance is a major issue.
Collapse
Affiliation(s)
- Marie Puerto
- Hôpital Haut-Lévêque - CHU de Bordeaux, Endocrinology Department, 33600 Pessac, France
| | | | - Antoine Tabarin
- Hôpital Haut-Lévêque - CHU de Bordeaux, Endocrinology Department, 33600 Pessac, France
| |
Collapse
|
16
|
Vogel WV, van der Marck SC, Versleijen MWJ. Challenges and future options for the production of lutetium-177. Eur J Nucl Med Mol Imaging 2021; 48:2329-2335. [PMID: 33974091 PMCID: PMC8241800 DOI: 10.1007/s00259-021-05392-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- W V Vogel
- Department of Nuclear Medicine, The Netherlands Cancer Institute-Antoni van Leeuwenhoek (NKI-AVL), Plesmanlaan 121, 1066, CX, Amsterdam, the Netherlands.
| | | | - M W J Versleijen
- Department of Nuclear Medicine, The Netherlands Cancer Institute-Antoni van Leeuwenhoek (NKI-AVL), Plesmanlaan 121, 1066, CX, Amsterdam, the Netherlands
| |
Collapse
|
17
|
Multiple endocrine neoplasia type 2: A reveiw. Semin Cancer Biol 2021; 79:163-179. [PMID: 33812987 DOI: 10.1016/j.semcancer.2021.03.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 03/13/2021] [Accepted: 03/27/2021] [Indexed: 12/16/2022]
Abstract
Multiple endocrine neoplasias are rare hereditary syndromes some of them with malignant potential. Multiple endocrine neoplasia type 2 (MEN 2) is an autosomal dominant hereditary cancer syndrome due to germline variants in the REarranged during Transfection (RET) proto-oncogene. There are two distinct clinical entities: MEN 2A and MEN 2B. MEN 2A is associated with medullary thyroid carcinoma (MTC), phaeochromocytoma, primary hyperparathyroidism, cutaneous lichen amyloidosis and Hirschprung's disease and MEN 2B with MTC, phaeochromocytoma, ganglioneuromatosis of the aerodigestive tract, musculoskeletal and ophthalmologic abnormalities. Germline RET variants causing MEN 2 result in gain-of-function; since the discovery of the genetic variants a thorough search for genotype-phenotype associations began in order to understand the high variability both between families and within family members. These studies have successfully led to improved risk classification of prognosis in relation to the genotype, thus improving the management of the patients by thorough genetic counseling. The present review summarizes the recent developments in the knowledge of these hereditary syndromes as well as the impact on clinical management, including genetic counseling, of both individual patients and families. It furthermore points to future directions of research for better clarification of timing of treatments of the various manifestations of the syndromes in order to improve survival and morbidity in these patients.
Collapse
|
18
|
Panzuto F. Clinical Challenges in the Management of Neuroendocrine Tumors. J Clin Med 2021; 10:jcm10020257. [PMID: 33445572 PMCID: PMC7826968 DOI: 10.3390/jcm10020257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Francesco Panzuto
- Digestive Diseases Unit, Sant'Andrea University Hospital, 00189 Rome, Italy
| |
Collapse
|
19
|
Bartz-Kurycki MA, Oluwo OE, Morris-Wiseman LF. Medullary thyroid carcinoma: recent advances in identification, treatment, and prognosis. Ther Adv Endocrinol Metab 2021; 12:20420188211049611. [PMID: 34659736 PMCID: PMC8511962 DOI: 10.1177/20420188211049611] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 09/12/2021] [Indexed: 12/13/2022] Open
Abstract
Medullary thyroid carcinoma (MTC) is a neuroendocrine tumor that represents <5% of all thyroid malignancies and is generally more aggressive than differentiated thyroid cancer. The aim of this study is to provide an update, through review of clinical studies of patients with MTC published between January 1, 2016, and June 1, 2021, on recent advances in the diagnosis and treatment of MTC. This review focuses on updates in biochemical testing, imaging, hereditary disease, surgical management, adjuvant therapies, and prognosis. Recent advances reviewed herein have sought to diagnose MTC at earlier stages of disease, predict when patients with a hereditary syndrome may develop MTC, use functional imaging to assess for distant metastases, perform optimal initial surgery with appropriate lymphadenectomy, employ targeted systemic therapies for patients with progressive metastatic disease, and better predict patient-specific outcomes.
Collapse
Affiliation(s)
| | - Omowunmi E. Oluwo
- Department of Surgery, University of Arizona
Tucson College of Medicine, Tucson, AZ, USA
| | | |
Collapse
|
20
|
Di Molfetta S, Dotto A, Fanciulli G, Florio T, Feola T, Colao A, Faggiano A. Immune Checkpoint Inhibitors: New Weapons Against Medullary Thyroid Cancer? Front Endocrinol (Lausanne) 2021; 12:667784. [PMID: 33935977 PMCID: PMC8081349 DOI: 10.3389/fendo.2021.667784] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/24/2021] [Indexed: 12/31/2022] Open
Abstract
Medullary thyroid carcinoma is a rare neuroendocrine neoplasm that originates from thyroid C cells. Surgery, with complete resection of the tumor, is the only curative approach. However, in most cases, the tumor recurs at locoregional or metastatic level. In this setting, the management remains challenging. In recent years, the immune checkpoint inhibitors have provided promise for changing the cancer treatment paradigm through the application of new approaches that enhance the body's natural antitumor defenses. The aim of this review is to summarize and discuss available data on efficacy and safety of the Food and Drug Administration-approved immune checkpoint inhibitors in patients with medullary thyroid carcinoma. After an extensive search, we found 7 useful data sources (one single-case report, one short article with very preliminary data, five ongoing registered clinical trials). Despite the lack of published evidence regarding the use of immune check point inhibitors, it must be considered that all the ongoing registered clinical trials saw first light in the last three years, thus indicating a growing interest of researchers in this field. Results coming from these trials, and hopefully, in the next future, from additional trials, will help to clarify whether this class of drugs may represent a new weapon in favor of patients with medullary thyroid carcinoma.
Collapse
Affiliation(s)
- Sergio Di Molfetta
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
- *Correspondence: Sergio Di Molfetta,
| | - Andrea Dotto
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Giuseppe Fanciulli
- Neuroendocrine Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari—Endocrine Unit, Azienda Ospedaliera Universitaria Sassari, Sassari, Italy
| | - Tullio Florio
- Department of Internal Medicine, University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Tiziana Feola
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, Naples, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
21
|
Okafor C, Hogan J, Raygada M, Thomas BJ, Akshintala S, Glod JW, Del Rivero J. Update on Targeted Therapy in Medullary Thyroid Cancer. Front Endocrinol (Lausanne) 2021; 12:708949. [PMID: 34489865 PMCID: PMC8416904 DOI: 10.3389/fendo.2021.708949] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/30/2021] [Indexed: 12/20/2022] Open
Abstract
Medullary thyroid carcinoma (MTC) is a rare neuroendocrine tumor that accounts for 2-4% of all thyroid cancers. All inherited MTC and approximately 50% of sporadic cases are driven by mutations in the REarranged during Transfection (RET) proto-oncogene. The recent expansion of the armamentarium of RET-targeting tyrosine kinase inhibitors (TKIs) has provided effective options for systemic therapy for patients with metastatic and progressive disease. However, patients that develop resistant disease as well as those with other molecular drivers such as RAS have limited options. An improved understanding of mechanisms of resistance to TKIs as well as identification of novel therapeutic targets is needed to improve outcomes for patients with MTC.
Collapse
Affiliation(s)
- Christian Okafor
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Julie Hogan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Margarita Raygada
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Barbara J. Thomas
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Srivandana Akshintala
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - John W. Glod
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Jaydira Del Rivero,
| |
Collapse
|