1
|
Yang X, Liu X, Liu Y, Wang XF, Chen Z, Wang X. Optimizing iodine capture performance by metal-organic framework containing with bipyridine units. Front Chem Sci Eng 2023; 17:395-403. [DOI: doi.org/10.1007/s11705-022-2218-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/01/2022] [Indexed: 06/25/2023]
|
2
|
Yang X, Liu X, Liu Y, Wang XF, Chen Z, Wang X. Optimizing iodine capture performance by metal-organic framework containing with bipyridine units. Front Chem Sci Eng 2022. [DOI: 10.1007/s11705-022-2218-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
3
|
Klein I, Mandel S, Burman K, Garber J, Haugen B. Effects of Radioiodine on the Nasolacrimal System. Endocr Pract 2022; 28:1272-1273. [PMID: 36174915 DOI: 10.1016/j.eprac.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Irwin Klein
- Department of Medicine, NYU School of Medicine, Melville, New York.
| | | | | | | | | |
Collapse
|
4
|
Qiu X, Wang P, Sa R, Cheng L, Jin Y, Song H, Chen L. Diagnosis and Treatment of Acute Pleural Effusion following Radioiodine Remnant Ablation Post Lobectomy for Thyroid Cancer. Diagnostics (Basel) 2022; 12:diagnostics12122982. [PMID: 36552989 PMCID: PMC9777309 DOI: 10.3390/diagnostics12122982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
Radioiodine remnant ablation (RRA) was previously demonstrated to be a safe and effective alternative to completion thyroidectomy for patients with differentiated thyroid cancer (DTC). However, its side effects have not been fully investigated, particularly in patients with lobectomy. We reported a young euthyroidal female who underwent RRA post lobectomy and lymph node dissection for papillary thyroid cancer, whose post-ablation 131I-whole-body scan accidentally showed diffuse radioiodine distribution on chest-mimicking pulmonary metastases. Immediately-added single-photon emission computed tomography/computed tomography (SPECT/CT), nevertheless, revealed a 131I-accumulating swollen left thyroid lobe and emerging pleural effusion, which relieved after short-term treatment with prednisone. In summary, acute pleural effusion ascribed to RRA-induced thoracic duct compression was reported for the first time. 131I-lobectomy-induced pleural effusion could be precisely diagnosed by SPECT/CT and efficiently manipulated via treating radiation thyroiditis with the short-term administration of corticosteroid.
Collapse
Affiliation(s)
- Xian Qiu
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Pengwen Wang
- Department of Thyroid Surgery, Panshi Hospital, 1 Kangfu Road, Panshi 132300, China
| | - Ri Sa
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Department of Nuclear Medicine, The First Hospital of Jilin University, 71 Xinmin St., Changchun 130021, China
| | - Lin Cheng
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Yuchen Jin
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Hongjun Song
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Libo Chen
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Correspondence: ; Tel.: +86-21-24058871; Fax: +86-21-64941720
| |
Collapse
|
5
|
Sellmyer MA, Lee IK, Mankoff DA. Building the Bridge: Molecular Imaging Biomarkers for 21 st Century Cancer Therapies. J Nucl Med 2021; 62:jnumed.121.262484. [PMID: 34446450 PMCID: PMC8612205 DOI: 10.2967/jnumed.121.262484] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 01/17/2023] Open
Abstract
Precision medicine, where the molecular underpinnings of the disease are assessed for tailored therapies, has greatly impacted cancer care. In parallel, a new pillar of therapeutics has emerged with profound success, including immunotherapies such as checkpoint inhibitors and cell-based therapies. Nonetheless, it remains essential to develop paradigms to predict and monitor for therapeutic response. Molecular imaging has the potential to add substantially to all phases of cancer patient care: predicative, companion diagnostics can illuminate therapeutic target density within a tumor, and pharmacodynamic imaging biomarkers can complement traditional modalities to judge a favorable treatment response. This "Focus on Molecular Imaging" article discusses the current role of molecular imaging in oncology and highlights an additional step in clinical paradigm termed a "therapeutic biomarker," which serves to assess whether next generation drugs reach their target to elicit a favorable clinical response.
Collapse
Affiliation(s)
- Mark A. Sellmyer
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Iris K. Lee
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A. Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Wu Y, Fang Q, Jin Q, Du W, Zhang X, Chen D. Parathyroid Hormone Level Changes Following Radioiodine Therapy for Thyroid Cancer: A Prospective Observational Study. Endocr Pract 2021; 27:342-347. [PMID: 33715818 DOI: 10.1016/j.eprac.2020.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/19/2020] [Accepted: 09/28/2020] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Our objective was to analyze the effect of radioiodine (RAI) therapy on parathyroid hormone (PTH) secretion. METHODS A total of 137 patients were included and divided into 2 groups based on pretherapy PTH levels. The residual thyroid tissue volume was classified into 4 grades (0-3), and a value of 0 indicated that there was no apparent residual tissue. We analyzed the PTH level changes among different time points in each group and the factors that could predict the PTH level changes. RESULTS In 113 patients with normal parathyroid gland function, the PTH level at baseline, 1 day, 7 days, 1 month, 3 months, and 6 months after RAI therapy did not show any significant difference; in 24 patients with decreased parathyroid gland function, the level of PTH immediately decreased after the implementation of RAI therapy but gradually returned to a pre-RAI therapy level within 6 months. On the seventh day after therapy, the mean value of PTH in patients with a residual thyroid tissue volume of extent of 0/1 was 8.0 ± 2.3 pg/mL, which was significantly higher than that in patients with a residual thyroid tissue volume of extent of 2/3 (P = .011). Similar phenomena were observed 1 month, 3 months, and 6 months after therapy. CONCLUSION RAI therapy had a significant transient adverse effect on parathyroid gland function in patients with decreased PTH secretion pretherapy, and the extent was associated with the amount of residual thyroid tissue.
Collapse
Affiliation(s)
- Yao Wu
- Department of Head Neck and Thyroid, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Qigen Fang
- Department of Head Neck and Thyroid, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.
| | - Qiufeng Jin
- Department of Head Neck and Thyroid, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Wei Du
- Department of Head Neck and Thyroid, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xu Zhang
- Department of Head Neck and Thyroid, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Defeng Chen
- Department of Head Neck and Thyroid, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
7
|
Kunos CA, Mankoff DA, Schultz MK, Graves SA, Pryma DA. Radiopharmaceutical Chemistry and Drug Development-What's Changed? Semin Radiat Oncol 2021; 31:3-11. [PMID: 33246634 DOI: 10.1016/j.semradonc.2020.07.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Radiation oncologists and nuclear medicine physicians have seen a resurgence in the clinical use of radiopharmaceuticals for the curative or palliative treatment of cancer. To enable the discovery and the development of new targeted radiopharmaceutical treatments, the United States National Cancer Institute has adapted its clinical trial enterprise to accommodate the requirements of a development program with investigational agents that have a radioactive isotope as part of the studied drug product. One change in perspective has been the consideration of investigational radiopharmaceuticals as drugs, with maximum tolerable doses determined by normal organ toxicity frequency like in drug clinical trials. Other changes include new clinical trial enterprise elements for biospecimen handling, adverse event reporting, regulatory conduct, writing services, drug master files, and reporting of patient outcomes. Arising from this enterprise, the study and clinical use of alpha-particle and beta-particle emitters have emerged as an important approach to cancer treatment. Resources allocated to this enterprise have brought forward biomarkers of molecular pathophysiology now used to select treatment or to evaluate clinical performance of radiopharmaceuticals. The clinical use of diagnostic and therapeutic radionuclide pairs is anticipated to accelerate radiopharmaceutical clinical development.
Collapse
Affiliation(s)
- Charles A Kunos
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA.
| | - David A Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Daniel A Pryma
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Asadian S, Mirzaei H, Kalantari BA, Davarpanah MR, Mohamadi M, Shpichka A, Nasehi L, Es HA, Timashev P, Najimi M, Gheibi N, Hassan M, Vosough M. β-radiating radionuclides in cancer treatment, novel insight into promising approach. Pharmacol Res 2020; 160:105070. [PMID: 32659429 DOI: 10.1016/j.phrs.2020.105070] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022]
Abstract
Targeted radionuclide therapy, known as molecular radiotherapy is a novel therapeutic module in cancer medicine. β-radiating radionuclides have definite impact on target cells via interference in cell cycle and particular signalings that can lead to tumor regression with minimal off-target effects on the surrounding tissues. Radionuclides play a remarkable role not only in apoptosis induction and cell cycle arrest, but also in the amelioration of other characteristics of cancer cells. Recently, application of novel β-radiating radionuclides in cancer therapy has been emerged as a promising therapeutic modality. Several investigations are ongoing to understand the underlying molecular mechanisms of β-radiating elements in cancer medicine. Based on the radiation dose, exposure time and type of the β-radiating element, different results could be achieved in cancer cells. It has been shown that β-radiating radioisotopes block cancer cell proliferation by inducing apoptosis and cell cycle arrest. However, physical characteristics of the β-radiating element (half-life, tissue penetration range, and maximum energy) and treatment protocol determine whether tumor cells undergo cell cycle arrest, apoptosis or both and to which extent. In this review, we highlighted novel therapeutic effects of β-radiating radionuclides on cancer cells, particularly apoptosis induction and cell cycle arrest.
Collapse
Affiliation(s)
- Samieh Asadian
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | | | | | - Morteza Mohamadi
- Department of Physical Chemistry, Faculty of Science, University of Tehran, Tehran, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Leila Nasehi
- Department of Medical Laboratory Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia; Department of Polymers and Composites, NN Semenov Institute of Chemical Physics, Moscow, Russia.
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research, Université Catholique de Louvain, B-1200 Brussels, Belgium
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
9
|
Kevadiya BD, Ottemann BM, Thomas MB, Mukadam I, Nigam S, McMillan J, Gorantla S, Bronich TK, Edagwa B, Gendelman HE. Neurotheranostics as personalized medicines. Adv Drug Deliv Rev 2019; 148:252-289. [PMID: 30421721 PMCID: PMC6486471 DOI: 10.1016/j.addr.2018.10.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022]
Abstract
The discipline of neurotheranostics was forged to improve diagnostic and therapeutic clinical outcomes for neurological disorders. Research was facilitated, in largest measure, by the creation of pharmacologically effective multimodal pharmaceutical formulations. Deployment of neurotheranostic agents could revolutionize staging and improve nervous system disease therapeutic outcomes. However, obstacles in formulation design, drug loading and payload delivery still remain. These will certainly be aided by multidisciplinary basic research and clinical teams with pharmacology, nanotechnology, neuroscience and pharmaceutic expertise. When successful the end results will provide "optimal" therapeutic delivery platforms. The current report reviews an extensive body of knowledge of the natural history, epidemiology, pathogenesis and therapeutics of neurologic disease with an eye on how, when and under what circumstances neurotheranostics will soon be used as personalized medicines for a broad range of neurodegenerative, neuroinflammatory and neuroinfectious diseases.
Collapse
Affiliation(s)
- Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brendan M Ottemann
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Midhun Ben Thomas
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saumya Nigam
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - JoEllyn McMillan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
10
|
Hosono M. Perspectives for Concepts of Individualized Radionuclide Therapy, Molecular Radiotherapy, and Theranostic Approaches. Nucl Med Mol Imaging 2019; 53:167-171. [PMID: 31231436 PMCID: PMC6554368 DOI: 10.1007/s13139-019-00586-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 11/29/2022] Open
Abstract
Radionuclide therapy (RNT) stands on the delivery of radiation to tumors or non-tumor target organs using radiopharmaceuticals that are designed to have specific affinity to targets. RNT is recently called molecular radiotherapy (MRT) by some advocators in order to emphasize its characteristics as radiotherapy and the relevance of dosimetry-guided optimization of treatment. Moreover, RNT requires relevant radiation protection standards because it employs unsealed radionuclides and gives therapeutic radiation doses in humans. On the basis of these radiation protection standards, the development and use of radiopharmaceuticals for combined application through diagnostics and therapeutics lead to theranostic approaches that will enhance the efficacy and safety of treatment by implementing dosimetry-based individualization.
Collapse
Affiliation(s)
- Makoto Hosono
- Institute of Advanced Clinical Medicine and Department of Radiology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511 Japan
| |
Collapse
|
11
|
Affiliation(s)
- Makoto Hosono
- Department of Radiology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
12
|
Abstract
The fundamental foundation for precision medicine is accurate and specific targeting of cancer cells. Advances in the understanding of cancer biology, developments in diagnostic technologies, and expansion of therapeutic options have all contributed to the concept of personalized cancer care. Theranostics is the systematic integration of targeted diagnostics and therapeutics. The theranostic platform includes an imaging component that "sees" the lesions followed by administration of the companion therapy agent that "treats" the same lesions. This strategy leads to enhanced therapy efficacy, manageable adverse events, improved patient outcome, and lower overall costs. Radiotheranostics refers to the use of radionuclides for the paired imaging and therapy agents. Radioiodine is the classic radiotheranostic agent that has been used clinically in management of thyroid diseases for nearly 75 years. More recently there have been major exciting strides in radiotheranostics for neuroendocrine tumors and prostate cancer, among other conditions. Regulatory approval of a number of radiotheranostic pairs is anticipated in the near future. Continued support will be needed in research and development to keep pace with the current momentum in radiotheranostics innovations. Moreover, regulatory and reimbursement agencies need to streamline their requirements for seamless transfer of the radiotheranostic agents from the bench to the bedside. In this review, the concept, history, recent developments, current challenges, and outlook for radiotheranostics in the treatment of patients with cancer will be discussed. © RSNA, 2018.
Collapse
Affiliation(s)
- Hossein Jadvar
- From the Department of Radiology, Division of Nuclear Medicine, Keck School of Medicine, University of Southern California, 2250 Alcazar St, CSC/IGM 102, Los Angeles, CA 90033 (H.J.); Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Md (X.C.); Department of Radiology, University of Wisconsin-Madison, Madison, Wis (W.C.); and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (U.M.)
| | - Xiaoyuan Chen
- From the Department of Radiology, Division of Nuclear Medicine, Keck School of Medicine, University of Southern California, 2250 Alcazar St, CSC/IGM 102, Los Angeles, CA 90033 (H.J.); Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Md (X.C.); Department of Radiology, University of Wisconsin-Madison, Madison, Wis (W.C.); and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (U.M.)
| | - Weibo Cai
- From the Department of Radiology, Division of Nuclear Medicine, Keck School of Medicine, University of Southern California, 2250 Alcazar St, CSC/IGM 102, Los Angeles, CA 90033 (H.J.); Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Md (X.C.); Department of Radiology, University of Wisconsin-Madison, Madison, Wis (W.C.); and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (U.M.)
| | - Umar Mahmood
- From the Department of Radiology, Division of Nuclear Medicine, Keck School of Medicine, University of Southern California, 2250 Alcazar St, CSC/IGM 102, Los Angeles, CA 90033 (H.J.); Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Md (X.C.); Department of Radiology, University of Wisconsin-Madison, Madison, Wis (W.C.); and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (U.M.)
| |
Collapse
|
13
|
Chen F, Yin S, Zhu J, Jia L, Zhang H, Yang C, Liu C, Deng Z. Effects of nuclear factor‑κB on the uptake of 131iodine and apoptosis of thyroid carcinoma cells. Mol Med Rep 2018; 17:4959-4964. [PMID: 29393421 PMCID: PMC5865955 DOI: 10.3892/mmr.2018.8481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/04/2017] [Indexed: 01/21/2023] Open
Abstract
Thyroid carcinoma is primarily treated by surgery combined with radioactive 131iodine (131I) treatment; however, certain patients exhibit resistance to 131I treatment. Previous research indicated that nuclear factor-κB (NF-κB) was associated with resistance to 131I in cancer cells. The present study aimed to investigate the effects of NF-κB on 131I uptake and apoptosis in thyroid carcinoma cells. TPC-1 and BCPAP cell lines were employed as research models in the present study, and the expression of NF-κB was inhibited by RNA interference (RNAi). The ability of TPC-1 and BCPAP cells to uptake 131I was measured and the cell viability was detected by an MTT assay. Finally, the expression of the apoptosis-associated proteins X-linked inhibitor of apoptosis (XIAP), cellular inhibitor of apoptosis protein 1 (cIAP1) and caspase-3 in TCP-1 and BCPAP cells was determined by western blotting. Western blotting results demonstrated that the expression levels of NF-κB in TPC-1 and BCPAP cells were successfully downregulated by RNAi (P<0.05), while analysis of 131I uptake revealed no significant alterations in the 131I uptake ability of cells following RNAi (P>0.05). MTT experiments demonstrated that the inhibition of NF-κB expression in combination with radiation (131I treatment) led to a marked reduction in cell viability (P<0.05). Furthermore, western blot analysis revealed that the inhibition of NF-κB expression downregulated the expression levels of XIAP and cIAP1 (P<0.05), while the expression levels of caspase-3 were upregulated, indicating that the observed reduction in cell viability following NF-κB inhibition may be due to an increased level of apoptosis. Although NF-κB inhibition did not affect the 131I uptake of thyroid cancer cells, this inhibition may increase the apoptotic effects of radioactive 131I.
Collapse
Affiliation(s)
- Fukun Chen
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Shuting Yin
- Third Ward of The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Jialun Zhu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Li Jia
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Huaping Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Chuanzhou Yang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Chao Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Zhiyong Deng
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
14
|
Kiyota N, Robinson B, Shah M, Hoff AO, Taylor MH, Li D, Dutcus CE, Lee EK, Kim SB, Tahara M. Defining Radioiodine-Refractory Differentiated Thyroid Cancer: Efficacy and Safety of Lenvatinib by Radioiodine-Refractory Criteria in the SELECT Trial. Thyroid 2017; 27:1135-1141. [PMID: 28665259 PMCID: PMC5646742 DOI: 10.1089/thy.2016.0549] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND While there is a clear consensus for defining radioiodine-refractory differentiated thyroid cancer (RR-DTC), it is unknown whether these criteria are equally valid for determining when radioiodine (RAI) therapy is no longer beneficial and systemic treatment should be considered. Lenvatinib, a multikinase inhibitor, significantly prolonged progression-free survival (PFS) compared to placebo in a Phase 3 trial in RR-DTC (SELECT; hazard ratio [HR]: 0.21 [99% confidence interval (CI) 0.14-0.31]; p < 0.001). This sub-analysis compared clinical outcomes of lenvatinib-treated patients in SELECT stratified by RR-DTC inclusion criteria. METHODS In SELECT, patients with measurable RR-DTC and radiologic evidence of disease progression ≤13 months prior to study entry were randomized 2:1 to lenvatinib (24 mg/day; 28-day cycle) or placebo. In this analysis, patients were stratified based on the following RR-DTC inclusion criteria: no RAI uptake, disease progression within 12 months of RAI therapy despite RAI avidity at the time of treatment, and extensive (>600 mCi) cumulative RAI exposure. All had disease progression as an inclusion criterion for SELECT. RESULTS Of 392 patients (261 lenvatinib; 131 placebo) enrolled, 275, 235, and 73 patients met the inclusion criteria for no RAI uptake, disease progression despite RAI avidity, and extensive RAI exposure, respectively. There was significant overlap between the patient groups, with 167 (42.6%) patients meeting more than one inclusion criterion. Lenvatinib improved median PFS compared to placebo in all groups ("no RAI uptake": lenvatinib not quantifiable [NQ; CI 14.8-NQ] vs. placebo, 3.7 months [CI 2.5-5.3]; "disease progression despite RAI avidity": lenvatinib 16.5 months [CI 12.8-NQ] vs. placebo, 3.7 months [CI 1.9-5.4]; "extensive RAI exposure": lenvatinib 18.7 months [CI 10.7-NQ] vs. placebo, 3.6 months [CI 1.9-5.5]). Objective response rates were 71.8%, 60.0%, and 56.0% for patients with no RAI uptake, disease progression despite RAI avidity, and extensive RAI exposure, respectively. Lenvatinib-related adverse events were similar across groups. CONCLUSIONS Comparable efficacy and safety profiles were observed in lenvatinib-treated patients regardless of RR-DTC criteria, possibly because of a large overlap among patients fulfilling each criterion. However, differing definitions for RR-DTC may be equally valid because both lenvatinib and placebo arms exhibited similar PFS outcomes across groups.
Collapse
Affiliation(s)
- Naomi Kiyota
- Department of Medical Oncology and Hematology, Kobe University Hospital and Kobe University Hospital Cancer Center, Kobe, Japan
| | - Bruce Robinson
- Kolling Institute of Medical Research, University of Sydney, New South Wales, Australia
| | - Manisha Shah
- Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Ana O. Hoff
- Department of Endocrinology, Endocrine Oncology Unit, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Matthew H. Taylor
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Di Li
- Eisai, Inc., Woodcliff Lake, New Jersey
| | | | - Eun Kyung Lee
- Center for Thyroid Cancer, National Cancer Center, Goyang, South Korea
| | - Sung-Bae Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Makoto Tahara
- Division of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
15
|
Abstract
OBJECTIVE This article reviews recent developments in targeted radionuclide therapy (TRT) approaches directed to malignant liver lesions, bone metastases, neuroendocrine tumors, and castrate-resistant metastatic prostate cancer and discusses challenges and opportunities in this field. CONCLUSION TRT has been employed since the first radioiodine thyroid treatment almost 75 years ago. Progress in the understanding of the complex underlying biology of cancer and advances in radiochemistry science, multimodal imaging techniques including the concept of "see and treat" within the framework of theranostics, and universal traction with the notion of precision medicine have all contributed to a resurgence of TRT.
Collapse
|
16
|
Samadi R, Ghanbari M, Shafiei B, Gheibi S, Azizi F, Ghasemi A. High dose of radioactive iodine per se has no effect on glucose metabolism in thyroidectomized rats. Endocrine 2017; 56:399-407. [PMID: 28283939 DOI: 10.1007/s12020-017-1274-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/23/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE Thyroid concentrates radioactive iodine by sodium-iodide symporter; this is used for treating hyperthyroidism and thyroid cancer. Pancreas expresses NIS and radioactive iodine uptake may damage pancreatic beta-cells and predispose patients to type 2 diabetes. The aim of this study was to determine whether radioactive iodine is associated with glucose metabolism in thyroidectomized rats. METHODS Forty male Wistar rats were divided into four groups (n = 10/each); control, thyroidectomized, thyroidectomized-treated with 131-I (TX+I), and thyroidectomized-treated with 131-I and L-thyroxine (TX+I+T4). At the end of study, serum fasting glucose, insulin, thyroid-stimulating hormone, and free tetraiodothyronine were measured, intraperitoneal glucose tolerance test was performed, and homeostasis model assessment-insulin resistance was calculated. In in vitro experiments, glucose-stimulated insulin secretion from pancreatic islets and sodium-iodide symporter mRNA expression in thyroid and islets were determined. RESULTS Compared to control group, free tetraiodothyronine was lower by 41 and 77% and thyroid-stimulating hormone was higher by 36 and 126% in thyroidectomized and TX+I groups, respectively. Compared to controls, rats in TX+I group had glucose intolerance as assessed using the area under curve of intraperitoneal glucose tolerance test (12,376 ± 542 vs. 20,769 ± 1070, P < 0.001) and L-thyroxine replacement therapy restored the value (14,286 ± 328.24) to near normal. Fasting insulin and homeostasis model assessment-insulin resistance were comparable in all groups, however fasting glucose was higher in TX+I group. In in vitro experiments, glucose-stimulated insulin secretion from islets did not differ between groups. CONCLUSION Radioactive iodine therapy per se had no effect on glucose metabolism, just intensified thyroid hormone deficiency and the alterations on glucose metabolism in thyroidectomized rats. L-thyroxine therapy restored the glucose intolerance observed in radioactive iodine-treated thyroidectomized rats.
Collapse
Affiliation(s)
- Roghaieh Samadi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Ghanbari
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Shafiei
- Department of Nuclear Medicine, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sevda Gheibi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Abstract
The goal of individualized and targeted treatment and precision medicine requires the assessment of potential therapeutic targets to direct treatment selection. The biomarkers used to direct precision medicine, often termed companion diagnostics, for highly targeted drugs have thus far been almost entirely based on in vitro assay of biopsy material. Molecular imaging companion diagnostics offer a number of features complementary to those from in vitro assay, including the ability to measure the heterogeneity of each patient's cancer across the entire disease burden and to measure early changes in response to treatment. We discuss the use of molecular imaging methods as companion diagnostics for cancer therapy with the goal of predicting response to targeted therapy and measuring early (pharmacodynamic) response as an indication of whether the treatment has "hit" the target. We also discuss considerations for probe development for molecular imaging companion diagnostics, including both small-molecule probes and larger molecules such as labeled antibodies and related constructs. We then describe two examples where both predictive and pharmacodynamic molecular imaging markers have been tested in humans: endocrine therapy for breast cancer and human epidermal growth factor receptor type 2-targeted therapy. The review closes with a summary of the items needed to move molecular imaging companion diagnostics from early studies into multicenter trials and into the clinic.
Collapse
Affiliation(s)
- David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
| | - Christine E Edmonds
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel A Pryma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
18
|
Prpic M, Kruljac I, Kust D, Kirigin LS, Jukic T, Dabelic N, Bolanca A, Kusic Z. Re-ablation I-131 activity does not predict treatment success in low- and intermediate-risk patients with differentiated thyroid carcinoma. Endocrine 2016; 52:602-8. [PMID: 26732041 DOI: 10.1007/s12020-015-0846-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/22/2015] [Indexed: 12/14/2022]
Abstract
The aim of this study was to evaluate the efficacy of different radioactive iodine (I-131) activities used for re-ablation, to compare various combinations of treatment activities, and to identify predictors of re-ablation failure in low- and intermediate-risk differentiated thyroid carcinoma (DTC) patients. The study included 128 consecutive low- and intermediate-risk patients with DTC with ablation failure after total thyroidectomy. Patient characteristics, T status, tumor size, lymph node involvement, postoperative remnant size on whole-body scintigraphy, serum thyroglobulin (Tg), thyroid-stimulating hormone (TSH), anti-Tg antibody (TgAb), and Tg/TSH ratio were analyzed as potential predictors of the re-ablation success. Re-ablation was successful in 113 out of 128 patients (88.3 %). Mean first I-131 activity was 2868 ± 914 MBq (77.5 ± 24.7 mCi) and mean second I-131 activity 3004 ± 699 MBq (81.2 ± 18.9 mCi). There was no association between the first, second, and cumulative activity with re-ablation treatment outcome. Treatment failure was associated with higher Tg levels prior to re-ablation (Tg2) (OR 1.16, 95 % CI 1.05-1.29, P = 0.003) and N1a status (OR 3.89, 95 % CI 1.13-13.41, P = 0.032). After excluding patients with positive-to-negative TgAb conversion, Tg2 level of 3.7 ng/mL predicted treatment failure with a sensitivity of 75.0 %, specificity of 80.5 %, and a negative predictive value of 97.1 %. Patients with positive-to-negative TgAb conversion had higher failure rates (OR 2.96, 95 % CI 0.94-9.29). Re-ablation success was high in all subgroups of patients and I-131 activity did not influence treatment outcome. Tg may serve as a good predictor of re-ablation failure. Patients with positive-to-negative TgAb conversion represent a specific group, in whom Tg level should not be used as a predictive marker of treatment outcome.
Collapse
Affiliation(s)
- Marin Prpic
- Department of Oncology and Nuclear Medicine, University Hospital Center "Sestre milosrdnice", Vinogradska cesta 29, 10000, Zagreb, Croatia.
| | - Ivan Kruljac
- Division of Endocrinology, Diabetes and Metabolic Diseases "Mladen Sekso", Department of Internal medicine, University Hospital Center "Sestre Milosrdnice", Vinogradska cesta 29, 10000, Zagreb, Croatia
| | - Davor Kust
- Department of Oncology and Nuclear Medicine, University Hospital Center "Sestre milosrdnice", Vinogradska cesta 29, 10000, Zagreb, Croatia
| | - Lora S Kirigin
- Division of Endocrinology, Diabetes and Metabolic Diseases "Mladen Sekso", Department of Internal medicine, University Hospital Center "Sestre Milosrdnice", Vinogradska cesta 29, 10000, Zagreb, Croatia
| | - Tomislav Jukic
- Department of Oncology and Nuclear Medicine, University Hospital Center "Sestre milosrdnice", Vinogradska cesta 29, 10000, Zagreb, Croatia
| | - Nina Dabelic
- Department of Oncology and Nuclear Medicine, University Hospital Center "Sestre milosrdnice", Vinogradska cesta 29, 10000, Zagreb, Croatia
| | - Ante Bolanca
- Department of Oncology and Nuclear Medicine, University Hospital Center "Sestre milosrdnice", Vinogradska cesta 29, 10000, Zagreb, Croatia
| | - Zvonko Kusic
- Department of Oncology and Nuclear Medicine, University Hospital Center "Sestre milosrdnice", Vinogradska cesta 29, 10000, Zagreb, Croatia
| |
Collapse
|
19
|
McFarland DC, Dang RP, Miles BA, Misiukiewicz K. Four Difficult Thyroid Cancer Cases: Incorporating Medical Therapies. Semin Oncol 2015; 42:e83-98. [PMID: 26615140 DOI: 10.1053/j.seminoncol.2015.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Daniel C McFarland
- Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Rajan P Dang
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Brett A Miles
- Department of Otolaryngology Head and Neck Surgery, Division of Head and Neck Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Krzysztof Misiukiewicz
- Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
20
|
Eckelman WC, Mankoff DA. Choosing a single target as a biomarker or therapeutic using radioactive probes. Nucl Med Biol 2015; 42:421-425. [PMID: 25684651 DOI: 10.1016/j.nucmedbio.2015.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 01/12/2015] [Indexed: 11/15/2022]
Affiliation(s)
| | - David A Mankoff
- Division of Nuclear Medicine, University of Pennsylvania, 116 Donner Building, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104-4283
| |
Collapse
|
21
|
Timmermand OV, Tran TA, Strand SE, Axelsson J. Intratherapeutic biokinetic measurements, dosimetry parameter estimates, and monitoring of treatment efficacy using cerenkov luminescence imaging in preclinical radionuclide therapy. J Nucl Med 2015; 56:444-9. [PMID: 25655628 DOI: 10.2967/jnumed.114.148544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED In recent years, there has been increasing interest in noninvasive Cerenkov luminescence imaging (CLI) of in vivo radionuclide distribution in small animals, a method proven to be a high-throughput modality for confirmation of tracer uptake. 11B6 is an IgG1 monoclonal antibody that is specific to free human kallikrein-related peptidase 2, an antigen abundant in malignant prostatic tissue. Free human kallikrein-related peptidase 2 was targeted in prostate cancer xenografts using (177)Lu-labeled 11B6 in either murine or humanized forms for radionuclide therapy. In this setting, CLI was investigated as a tool for providing parameters of dosimetric importance during radionuclide therapy. First, longitudinal imaging of biokinetics using CLI and SPECT was compared. Second, the CLI signal was correlated to quantitative ex vivo tumor activity measurements. Finally, CLI was used to monitor the radionuclide treatment, and the integrated CLI radiance was found to correlate well with subject-specific tumor volume reduction. METHODS 11B6 was radiolabeled with (177)Lu through the CHX-A″-DTPA chelator. In vivo CLI and SPECT imaging of (177)Lu-DTPA-11B6 uptake was performed on NMRI and BALB/c nude mice with subcutaneous LNCaP xenografts up to 14 d after injection. Tumor size was measured to assess response to radionuclide therapy. RESULTS CLI correlated well with SPECT imaging and could be applied up to 14 d after injection of 20 MBq with the specific tracer used. Through integration of the CLI radiance as a function of time, a dose metric for the tumors could be formed that correlated exponentially with tumor volume reduction. CONCLUSION CLI provided valuable intratherapeutic biokinetic measurements for treatment monitoring and could be used as a tool for subject-specific absorbed dose estimation.
Collapse
Affiliation(s)
| | - Thuy A Tran
- Lund University Bioimaging Center, Lund University, Lund, Sweden; and
| | - Sven-Erik Strand
- Department of Medical Radiation Physics, Lund University, Lund, Sweden Lund University Bioimaging Center, Lund University, Lund, Sweden; and
| | - Johan Axelsson
- Division of Atomic Physics, Department of Physics, Lund University, Lund, Sweden
| |
Collapse
|