1
|
Vanermen M, Ligeour M, Oliveira MC, Gestin JF, Elvas F, Navarro L, Guérard F. Astatine-211 radiolabelling chemistry: from basics to advanced biological applications. EJNMMI Radiopharm Chem 2024; 9:69. [PMID: 39365487 PMCID: PMC11452365 DOI: 10.1186/s41181-024-00298-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND 211At-radiopharmaceuticals are currently the subject of growing studies for targeted alpha therapy of cancers, which leads to the widening of the scope of the targeting vectors, from small molecules to peptides and proteins. This has prompted, during the past decade, to a renewed interest in developing novel 211At-labelling approaches and novel prosthetic groups to address the diverse scenarios and to reach improved efficiency and robustness of procedures as well as an appropriate in vivo stability of the label. MAIN BODY Translated from the well-known (radio)iodine chemistry, the long preferred electrophilic astatodemetallation using trialkylaryltin precursors is now complemented by new approaches using electrophilic or nucleophilic At. Alternatives to the astatoaryl moiety have been proposed to improve labelling stability, and the range of prosthetic groups available to label proteins has expanded. CONCLUSION In this report, we cover the evolution of radiolabelling chemistry, from the initial strategies developed in the late 1970's to the most recent findings.
Collapse
Affiliation(s)
- Maarten Vanermen
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | - Mathilde Ligeour
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Maria-Cristina Oliveira
- Departamento de Engenharia e Ciências Nucleares and Centro de Ciências e Tecnologias Nucleares, IST, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
| | | | - Filipe Elvas
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | | | - François Guérard
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France.
| |
Collapse
|
2
|
Garaulet G, Báez BB, Medrano G, Rivas-Sánchez M, Sánchez-Alonso D, Martinez-Torrecuadrada JL, Mulero F. Radioimmunotheragnosis in Cancer Research. Cancers (Basel) 2024; 16:2896. [PMID: 39199666 PMCID: PMC11352548 DOI: 10.3390/cancers16162896] [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: 06/13/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
The combination of immunoPET-where an antibody (Ab) is labeled with an isotope for PET imaging-and radioimmunotherapy (RIT), using the same antibody with a therapeutic isotope, offers significant advantages in cancer management. ImmunoPET allows non-invasive imaging of antigen expression, which aids in patient selection for subsequent radioimmunotherapy. It also facilitates the assessment of tumor response to therapy, allowing for treatment adjustments if necessary. In addition, immunoPET provides critical pharmacokinetic data, including antibody biodistribution and clearance rates, which are essential for dosimetry calculations and treatment protocol optimization. There are still challenges to overcome. Identifying appropriate target antigens that are selectively expressed on cancer cells while minimally expressed on normal tissues remains a major hurdle to reduce off-target toxicity. In addition, it is critical to optimize the pharmacokinetics of radiolabeled antibodies to maximize tumor uptake and minimize normal tissue uptake, particularly in vital organs such as the liver and kidney. This approach offers the potential for targeted and personalized cancer therapy with reduced systemic toxicity by exploiting the specificity of monoclonal antibodies and the cytotoxic effects of radiation. However, further research is needed to address remaining challenges and to optimize these technologies for clinical use.
Collapse
Affiliation(s)
- Guillermo Garaulet
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Bárbara Beatriz Báez
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Guillermo Medrano
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - María Rivas-Sánchez
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | - David Sánchez-Alonso
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | | | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| |
Collapse
|
3
|
Zhao X, Jakobsson V, Tao Y, Zhao T, Wang J, Khong PL, Chen X, Zhang J. Targeted Radionuclide Therapy in Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39042829 DOI: 10.1021/acsami.4c07850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Despite the development of various novel therapies, glioblastoma (GBM) remains a devastating disease, with a median survival of less than 15 months. Recently, targeted radionuclide therapy has shown significant progress in treating solid tumors, with the approval of Lutathera for neuroendocrine tumors and Pluvicto for prostate cancer by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This achievement has shed light on the potential of targeted radionuclide therapy for other solid tumors, including GBM. This review presents the current status of targeted radionuclide therapy in GBM, highlighting the commonly used therapeutic radionuclides emitting alpha, beta particles, and Auger electrons that could induce potent molecular and cellular damage to treat GBM. We then explore a range of targeting vectors, including small molecules, peptides, and antibodies, which selectively target antigen-expressing tumor cells with minimal or no binding to healthy tissues. Considering that radiopharmaceuticals for GBM are often administered locoregionally to bypass the blood-brain barrier (BBB), we review prominent delivery methods such as convection-enhanced delivery, local implantation, and stereotactic injections. Finally, we address the challenges of this therapeutic approach for GBM and propose potential solutions.
Collapse
Affiliation(s)
- Xiaobin Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yucen Tao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jingyan Wang
- Xiamen University, School of Public Health, Xiang'an South Road, Xiamen 361102, China
| | - Pek-Lan Khong
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Departments of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
4
|
Hurley K, Cao M, Huang H, Wang Y. Targeted Alpha Therapy (TAT) with Single-Domain Antibodies (Nanobodies). Cancers (Basel) 2023; 15:3493. [PMID: 37444603 DOI: 10.3390/cancers15133493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
The persistent threat of cancer necessitates the development of improved and more efficient therapeutic strategies that limit damage to healthy tissues. Targeted alpha therapy (TαT), a novel form of radioimmuno-therapy (RIT), utilizes a targeting vehicle, commonly antibodies, to deliver high-energy, but short-range, alpha-emitting particles specifically to cancer cells, thereby reducing toxicity to surrounding normal tissues. Although full-length antibodies are often employed as targeting vehicles for TαT, their high molecular weight and the presence of an Fc-region lead to a long blood half-life, increased bone marrow toxicity, and accumulation in other tissues such as the kidney, liver, and spleen. The discovery of single-domain antibodies (sdAbs), or nanobodies, naturally occurring in camelids and sharks, has introduced a novel antigen-specific vehicle for molecular imaging and TαT. Given that nanobodies are the smallest naturally occurring antigen-binding fragments, they exhibit shorter relative blood half-lives, enhanced tumor uptake, and equivalent or superior binding affinity and specificity. Nanobody technology could provide a viable solution for the off-target toxicity observed with full-length antibody-based TαT. Notably, the pharmacokinetic properties of nanobodies align better with the decay characteristics of many short-lived α-emitting radionuclides. This review aims to encapsulate recent advancements in the use of nanobodies as a vehicle for TαT.
Collapse
Affiliation(s)
- Kate Hurley
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Meiyun Cao
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Haiming Huang
- Research Center, Forlong Biotechnology Inc., Suzhou 215004, China
| | - Yi Wang
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
5
|
Buck AK, Serfling SE, Kraus S, Samnick S, Dreher N, Higuchi T, Rasche L, Einsele H, Werner RA. Theranostics in Hematooncology. J Nucl Med 2023:jnumed.122.265199. [PMID: 37290799 DOI: 10.2967/jnumed.122.265199] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
In the early 2000s, major clinical trials provided evidence of a favorable outcome from antibody-mediated radioimmunotherapy for hematologic neoplasms, which then led to Food and Drug Administration approval. For instance, the theranostic armamentarium for the referring hematooncologist now includes 90Y-ibritumomab tiuxetan for refractory low-grade follicular lymphoma or transformed B-cell non-Hodgkin lymphoma, as well as 131I-tositumomab for rituximab-refractory follicular lymphoma. Moreover, the first interim results of the SIERRA phase III trial reported beneficial effects from the use of 131I-anti-CD45 antibodies (Iomab-B) in refractory or relapsed acute myeloid leukemia. During the last decade, the concept of theranostics in hematooncology has been further expanded by C-X-C motif chemokine receptor 4-directed molecular imaging. Beyond improved detection rates of putative sites of disease, C-X-C motif chemokine receptor 4-directed PET/CT also selects candidates for radioligand therapy using β-emitting radioisotopes targeting the identical chemokine receptor on the lymphoma cell surface. Such image-piloted therapeutic strategies provided robust antilymphoma efficacy, along with desired eradication of the bone marrow niche, such as in patients with T- or B-cell lymphoma. As an integral part of the treatment plan, such radioligand therapy-mediated myeloablation also allows one to line up patients for stem cell transplantation, which leads to successful engraftment during the further treatment course. In this continuing education article, we provide an overview of the current advent of theranostics in hematooncology and highlight emerging clinical applications.
Collapse
Affiliation(s)
- Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany;
| | | | - Sabrina Kraus
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany; and
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Niklas Dreher
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany; and
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany; and
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
6
|
Gao Y, Liu Y, Wang Y, Zhang Q, Wu D, Ye X, Wu J, Xu W, Zhou J, Yang Y, Cen H, Zhang F, Xiang Y, Tang X, Ding K, Lin J, Ma L, Wang S, Yu H, Zhao Y, Song B, Lv F, Huang H. Bendamustine in the treatment of patients with indolent non-Hodgkin lymphoma refractory or relapse to rituximab treatment: An open-label, single-agent, multicenter study in China. Cancer 2023; 129:551-559. [PMID: 36541221 DOI: 10.1002/cncr.34544] [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: 06/08/2022] [Revised: 07/18/2022] [Accepted: 08/03/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND The optimal treatment strategy for refractory or relapse (R/R) indolent non-Hodgkin lymphoma (iNHL) has not been fully identified. This study aims to investigate the efficacy and tolerance of bendamustine hydrochloride developed in native Chinese corporation in the treatment of patients with R/R iNHL. METHODS A total of 101 patients from 19 centers were enrolled in this study from July 2016 to February 2019. Bendamustine hydrochloride (120 mg/m2 ) was given on days 1 and 2 of each 21-day treatment cycle for six planned cycles or up to eight cycles if tolerated. Parameters of efficacy and safety were analyzed. RESULTS The median age of the patients was 53.44 (range, 24.4-74.6) years old. A total of 56 (55.44%) patients completed at least six treatment cycles, and the relative dose intensity was 93.78%. The overall response rate was 72.28%, and the median duration of response was 15.84 months (95% confidence interval [CI], 13.77-27.48 months). Median progression-free survival was 16.52 months (95% CI, 14.72-23.41 months), and the median overall survival was not reached. Grade 3 or 4 hematologic toxicities included neutropenia (77.22%), thrombocytopenia (29.70%), and anemia (15.84%). The most frequent nonhematologic adverse events (any grade) included nausea, vomiting, fatigue, fever, decreased appetite, and weight loss. Seven patients died during the trial, and four cases may be related to the investigational drug. CONCLUSIONS This study reveals that bendamustine hydrochloride is a feasible treatment option for the indolent B-cell non-Hodgkin lymphoma patient who has not remitted or relapsed after treatment with rituximab. All adverse events were predictable and manageable.
Collapse
Affiliation(s)
- Yan Gao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yizhen Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yafei Wang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Depei Wu
- Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xu Ye
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianqiu Wu
- Department of Medical Oncology, Jiangsu Institute of Cancer Research, Jiangsu Red Cross Cancer Center, Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Xu
- Hematology Department, Jiangsu Province Hospital, Nanjing, China
| | - Jianfeng Zhou
- Hematology Department, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Yu Yang
- Department of Lymphoma and Head and Neck Cancer, The Affiliated Tumor Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Hong Cen
- Department of Hematology, Lymphoma and Pediatric Oncology, Guangxi Medical University Affiliated Tumor Hospital and Oncology Medical College, Nanning, China
| | - Feng Zhang
- Department of Hematology, Anhui Oncology Hospital, Bengbu Medical College, Bengbu, China
| | - Ying Xiang
- Department of Hematology and Oncology, Chongqing Cancer Hospital, Chongqing, China
| | - Xiaoqiong Tang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kaiyang Ding
- Department of Hematology, Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui Province, China
| | - JinYing Lin
- Department of Hematology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lei Ma
- Guangzhou Medical University Affiliated Tumor Hospital and Oncology Medical College, Nanning, China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou, Guangdong, China
| | - Hao Yu
- Department of Biostatistics, Nanjing Medical University, Nanjing, China
| | - Yang Zhao
- Nanjing Baosida Pharmaceutical Technology Co., Ltd, Nanjing, China
| | - Bin Song
- West China Hospital of Sichuan University, Sichuan, China
| | - Fangfang Lv
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huiqiang Huang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
7
|
Bodei L, Herrmann K, Schöder H, Scott AM, Lewis JS. Radiotheranostics in oncology: current challenges and emerging opportunities. Nat Rev Clin Oncol 2022; 19:534-550. [PMID: 35725926 PMCID: PMC10585450 DOI: 10.1038/s41571-022-00652-y] [Citation(s) in RCA: 129] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2022] [Indexed: 12/20/2022]
Abstract
Structural imaging remains an essential component of diagnosis, staging and response assessment in patients with cancer; however, as clinicians increasingly seek to noninvasively investigate tumour phenotypes and evaluate functional and molecular responses to therapy, theranostics - the combination of diagnostic imaging with targeted therapy - is becoming more widely implemented. The field of radiotheranostics, which is the focus of this Review, combines molecular imaging (primarily PET and SPECT) with targeted radionuclide therapy, which involves the use of small molecules, peptides and/or antibodies as carriers for therapeutic radionuclides, typically those emitting α-, β- or auger-radiation. The exponential, global expansion of radiotheranostics in oncology stems from its potential to target and eliminate tumour cells with minimal adverse effects, owing to a mechanism of action that differs distinctly from that of most other systemic therapies. Currently, an enormous opportunity exists to expand the number of patients who can benefit from this technology, to address the urgent needs of many thousands of patients across the world. In this Review, we describe the clinical experience with established radiotheranostics as well as novel areas of research and various barriers to progress.
Collapse
Affiliation(s)
- Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA
| | - Ken Herrmann
- German Cancer Consortium, University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Pharmacology, Weill Cornell Medical School, New York, NY, 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: 16] [Impact Index Per Article: 4.0] [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
|
Radioimmunotherapy in Oncology. CURRENT RADIOLOGY REPORTS 2017. [DOI: 10.1007/s40134-017-0258-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
10
|
Yeong CH, Cheng MH, Ng KH. Therapeutic radionuclides in nuclear medicine: current and future prospects. J Zhejiang Univ Sci B 2015; 15:845-63. [PMID: 25294374 DOI: 10.1631/jzus.b1400131] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The potential use of radionuclides in therapy has been recognized for many decades. A number of radionuclides, such as iodine-131 ((131)I), phosphorous-32 ((32)P), strontium-90 ((90)Sr), and yttrium-90 ((90)Y), have been used successfully for the treatment of many benign and malignant disorders. Recently, the rapid growth of this branch of nuclear medicine has been stimulated by the introduction of a number of new radionuclides and radiopharmaceuticals for the treatment of metastatic bone pain and neuroendocrine and other malignant or non-malignant tumours. Today, the field of radionuclide therapy is enjoying an exciting phase and is poised for greater growth and development in the coming years. For example, in Asia, the high prevalence of thyroid and liver diseases has prompted many novel developments and clinical trials using targeted radionuclide therapy. This paper reviews the characteristics and clinical applications of the commonly available therapeutic radionuclides, as well as the problems and issues involved in translating novel radionuclides into clinical therapies.
Collapse
Affiliation(s)
- Chai-Hong Yeong
- Department of Biomedical Imaging & University of Malaya Research Imaging Centre, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; Department of Nuclear Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | | | | |
Collapse
|
11
|
Development of a dry distillation technology for the production of 131I using medium flux reactor for radiopharmaceutical applications. J Radioanal Nucl Chem 2014. [DOI: 10.1007/s10967-014-3423-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
12
|
Novy Z, Laznickova A, Mandikova J, Barta P, Laznicek M, Trejtnar F. The effect of chelator type onin vitroreceptor binding and stability in177Lu-labeled cetuximab and panitumumab. J Labelled Comp Radiopharm 2014; 57:448-52. [DOI: 10.1002/jlcr.3204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 02/01/2014] [Accepted: 04/11/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Zbynek Novy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove; Charles University in Prague; Czech Republic
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry; Palacky University Olomouc; Czech Republic
| | - Alice Laznickova
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy in Hradec Kralove; Charles University in Prague; Czech Republic
| | - Jana Mandikova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove; Charles University in Prague; Czech Republic
| | - Pavel Barta
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove; Charles University in Prague; Czech Republic
| | - Milan Laznicek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove; Charles University in Prague; Czech Republic
| | - Frantisek Trejtnar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove; Charles University in Prague; Czech Republic
| |
Collapse
|
13
|
Sharkey RM, Burton J, Goldenberg DM. Radioimmunotherapy of non-Hodgkin’s lymphoma: a critical appraisal. Expert Rev Clin Immunol 2014; 1:47-62. [DOI: 10.1586/1744666x.1.1.47] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
14
|
Pharmacokinetics, pharmacodynamics and physiologically-based pharmacokinetic modelling of monoclonal antibodies. Clin Pharmacokinet 2013; 52:83-124. [PMID: 23299465 DOI: 10.1007/s40262-012-0027-4] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Development of monoclonal antibodies (mAbs) and their functional derivatives represents a growing segment of the development pipeline in the pharmaceutical industry. More than 25 mAbs and derivatives have been approved for a variety of therapeutic applications. In addition, around 500 mAbs and derivatives are currently in different stages of development. mAbs are considered to be large molecule therapeutics (in general, they are 2-3 orders of magnitude larger than small chemical molecule therapeutics), but they are not just big chemicals. These compounds demonstrate much more complex pharmacokinetic and pharmacodynamic behaviour than small molecules. Because of their large size and relatively poor membrane permeability and instability in the conditions of the gastrointestinal tract, parenteral administration is the most usual route of administration. The rate and extent of mAb distribution is very slow and depends on extravasation in tissue, distribution within the particular tissue, and degradation. Elimination primarily happens via catabolism to peptides and amino acids. Although not definitive, work has been published to define the human tissues mainly involved in the elimination of mAbs, and it seems that many cells throughout the body are involved. mAbs can be targeted against many soluble or membrane-bound targets, thus these compounds may act by a variety of mechanisms to achieve their pharmacological effect. mAbs targeting soluble antigen generally exhibit linear elimination, whereas those targeting membrane-bound antigen often exhibit non-linear elimination, mainly due to target-mediated drug disposition (TMDD). The high-affinity interaction of mAbs and their derivatives with the pharmacological target can often result in non-linear pharmacokinetics. Because of species differences (particularly due to differences in target affinity and abundance) in the pharmacokinetics and pharmacodynamics of mAbs, pharmacokinetic/pharmacodynamic modelling of mAbs has been used routinely to expedite the development of mAbs and their derivatives and has been utilized to help in the selection of appropriate dose regimens. Although modelling approaches have helped to explain variability in both pharmacokinetic and pharmacodynamic properties of these drugs, there is a clear need for more complex models to improve understanding of pharmacokinetic processes and pharmacodynamic interactions of mAbs with the immune system. There are different approaches applied to physiologically based pharmacokinetic (PBPK) modelling of mAbs and important differences between the models developed. Some key additional features that need to be accounted for in PBPK models of mAbs are neonatal Fc receptor (FcRn; an important salvage mechanism for antibodies) binding, TMDD and lymph flow. Several models have been described incorporating some or all of these features and the use of PBPK models are expected to expand over the next few years.
Collapse
|
15
|
Litvak-Greenfeld D, Benhar I. Risks and untoward toxicities of antibody-based immunoconjugates. Adv Drug Deliv Rev 2012; 64:1782-99. [PMID: 22659123 DOI: 10.1016/j.addr.2012.05.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/18/2012] [Accepted: 05/24/2012] [Indexed: 01/08/2023]
Abstract
Antibody-based immunoconjugates are specifically targeted monoclonal antibodies that deliver a cytotoxic payload to their target. The cytotoxic agents can be highly potent drugs, radionuclides or toxins. Such molecules, referred to as antibody-drug conjugates, radioimmunoconjugates and immunotoxins, respectively, represent a promising approach for enhancing the efficacy of unconjugated (naked) antibodies for improved therapeutic results. Though tremendous progress has been achieved over the last few decades, the safety of these molecules still remains a matter of concern and a careful design is required for achieving a relatively safe toxicity profile along with therapeutic effectiveness. This review focuses on the toxicities arising from the use of these potent agents.
Collapse
|
16
|
Abstract
BACKGROUND Radioimmunotherapy (RIT) is a safe and effective therapeutic option for patients with indolent B-cell non-Hodgkin lymphomas (NHL), in both up-front and relapsed/refractory settings. Two approved agents (90Y-ibritumomab tiuxetan and 131I-tositumomab) are available in the United States. Both target CD20 with similar clinical outcomes but with unique clinical considerations and radiation precautions due to the use of varying radioisotopes. METHODS This paper reviews the available evidence for these approved RIT agents and examines the recently published and ongoing clinical trials of potential novel indications for aggressive B-cell NHL. RESULTS A pretreatment biodistribution evaluation required before administering the 90Y-ibritumomab tiuxetan therapeutic dose has been removed, which once limited its usage. The potential clinical applications of RIT include relapsed/refractory indolent B-cell NHL, diffuse large B-cell lymphoma, indolent lymphoma in the front-line setting, and mantle cell lymphoma. Multiple novel RIT agents are in preclinical and clinical development, and the addition of radiosensitizers or external-beam radiotherapy may act in synergy with RIT for both indolent and aggressive lymphomas. The risk of treatment-related myelodysplastic syndrome does not appear to be higher in patients treated with RIT over those receiving chemotherapy alone. CONCLUSIONS RIT is a safe, effective, and significantly underutilized therapy for patients with B-cell NHL, and many studies have demonstrated the efficacy of 90Y-ibritumomab tiuxetan and 131I-tositumomab for relapsed/refractory indolent B-cell lymphomas. Continued research to establish its efficacy for other lymphoma subtypes is warranted.
Collapse
Affiliation(s)
- Michael Tomblyn
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
17
|
Cang S, Mukhi N, Wang K, Liu D. Novel CD20 monoclonal antibodies for lymphoma therapy. J Hematol Oncol 2012; 5:64. [PMID: 23057966 PMCID: PMC3479003 DOI: 10.1186/1756-8722-5-64] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 10/09/2012] [Indexed: 01/14/2023] Open
Abstract
Rituximab (RTX), a monoclonal antibody (mAb) against CD20, has been widely used for lymphoma therapy. RTX in combination with cyclophosphamide /doxorubicin /vincristine /prednisone (R-CHOP) remains the standard frontline regimen for diffuse large B-cell lymphoma. However, suboptimal response and /or resistance to rituximab have remained a challenge in the therapy of B-cell non-Hodgkin's lymphoma (NHL). Novel agents are under active clinical trials. This review will summarize the latest development in new mAbs against CD20, which include second-generation mAbs, ofatumumab, veltuzumab (IMMU-106), ocrelizumab (PRO70769), and third-generation mAbs, AME-133v (ocaratuzumab), PRO131921 and GA101 (obinutumumab).
Collapse
Affiliation(s)
- Shundong Cang
- Department of Oncology, People’s Hospital, Henan Province, China
| | - Nikhil Mukhi
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY, 10595, USA
| | - Kemeng Wang
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY, 10595, USA
| | - Delong Liu
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY, 10595, USA
| |
Collapse
|
18
|
Zhao J, Xu Z, Liu D, Lu Q. Rituximab and new regimens for indolent lymphoma: a brief update from 2012 ASCO Annual Meeting. Cancer Cell Int 2012; 12:38. [PMID: 22913602 PMCID: PMC3480919 DOI: 10.1186/1475-2867-12-38] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 08/17/2012] [Indexed: 11/10/2022] Open
Abstract
Indolent lymphoma (IL), the second most common lymphoma, remains incurable with chemotherapy alone. While R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone) remains the standard frontline regimen for diffuse Large B -cell lymphoma, the optimal chemotherapy regimen for frontline therapy of advanced IL remains uncertain. FCR (fludarabine, cyclophosphamide, rituximab) has been shown to be better than fludarabine alone and fludarabine plus cyclophosphamide for IL. In FOLL05 trial, R-CHOP was compared with R-CVP (cyclophosphamide, vincristine, prednisone) and R-FM (fludarabine, mitoxantrone). The study showed that R-CHOP appears to have the best risk-benefit ratio for IL. The StiL NHL1 trial showed that BR (bendamustine, rituximab) has longer progression free survival and is better tolerated than R-CHOP. Long-term complications with secondary malignancies between the two regimens appear to be comparable. In this review, new combination regimens reported at 2012 ASCO annual meeting were evaluated for frontline and salvage therapy of indolent lymphoma.
Collapse
Affiliation(s)
- Jiangning Zhao
- Department of Hematology, Zhongshan Hospital of Xiamen University, Xiamen 361004, China
| | - Zhenshu Xu
- Institute of Hematology, Fujian Union Hospital, Fuzhou, China
| | - Delong Liu
- Division of Hematology and Oncology, New York Medical College and Westchester Medical Center, Valhalla, NY 10595, USA
| | - Quanyi Lu
- Department of Hematology, Zhongshan Hospital of Xiamen University, Xiamen 361004, China
| |
Collapse
|
19
|
Ahmed S, Winter JN, Gordon LI, Evens AM. Radioimmunotherapy for the treatment of non-Hodgkin lymphoma: current status and future applications. Leuk Lymphoma 2010; 51:1163-77. [PMID: 20470217 DOI: 10.3109/10428191003793366] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Radioimmunotherapy (RIT) has proved to be a safe and effective treatment for patients with relapsed or refractory indolent non-Hodgkin lymphoma (NHL) including rituximab-refractory follicular lymphoma. Further, FDA approval was recently granted for use in newly diagnosed follicular lymphoma as consolidative therapy immediately following induction chemotherapy. We detail herein the scope of clinical studies performed in relapsed/refractory and newly diagnosed indolent lymphoma and summarize the associated safety data. In addition, we discuss new applications of RIT that have been investigated in a variety of clinical scenarios (e.g. single-agent and sequential therapy in aggressive NHLs and as a component of stem cell transplant conditioning). The wide array of RIT-based studies have yielded encouraging data, although randomized controlled trials will be needed to prove superiority over conventional therapy. Novel therapeutic RIT-based strategies that continue to be explored include radiation-enhancing agents combined with RIT, pre-targeting, RIT fractionation, as well as the integration of new humanized antibodies. The field of RIT continues to evolve scientifically and grow clinically. A reappraisal of prior data and examination of recently published and ongoing studies will be important in recognizing the potential benefit of RIT in the treatment of NHL.
Collapse
Affiliation(s)
- Sairah Ahmed
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | |
Collapse
|
20
|
Schaefer-Cutillo J, Friedberg JW. Non-myeloablative Radioimmunotherapy for Non-Hodgkin’s Lymphoma. Semin Hematol 2008; 45:110-7. [DOI: 10.1053/j.seminhematol.2008.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
21
|
Torres-García E, Ferro-Flores G, Arteaga de Murphy C, Correa-González L, Pichardo-Romero PA. Biokinetics and Dosimetry of 188Re-anti-CD20 in Patients with Non-Hodgkin's Lymphoma: Preliminary Experience. Arch Med Res 2008; 39:100-9. [DOI: 10.1016/j.arcmed.2007.06.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Accepted: 06/18/2007] [Indexed: 11/25/2022]
|
22
|
Leonard JP, Goldenberg DM. Preclinical and clinical evaluation of epratuzumab (anti-CD22 IgG) in B-cell malignancies. Oncogene 2007; 26:3704-13. [PMID: 17530024 DOI: 10.1038/sj.onc.1210370] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The vast majority of non-Hodgkin's lymphomas are of B-cell phenotype. Development of unlabeled or radiolabeled therapeutic monoclonal antibodies against the cell surface antigen, CD20, has revolutionized the treatment of these malignancies. It is clear that antibodies targeting other B-cell-specific molecules, such as CD22, also offer potential therapeutic benefit. Epratuzumab is a humanized anti-CD22 monoclonal, which has undergone preclinical and phase I/II clinical evaluation in patients with indolent or aggressive lymphoma. Data suggest that this agent is well tolerated, and can induce tumor regressions. Trials are currently evaluating its safety and activity in combination with rituximab (chimeric anti-CD20) and standard chemotherapy are ongoing. Initial results suggest that these regimens have acceptable toxicity, and that epratuzumab warrants further evaluation as an adjunct to standard lymphoma treatment regimens.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Drug Screening Assays, Antitumor
- Humans
- Immunotherapy
- Leukemia, B-Cell/drug therapy
- Leukemia, B-Cell/immunology
- Leukemia, B-Cell/pathology
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Sialic Acid Binding Ig-like Lectin 2/immunology
Collapse
Affiliation(s)
- J P Leonard
- Department of Medicine, Center for Lymphoma and Myeloma, Weill Medical College of Cornell University and New York Presbyterian Hospital, New York, NY 10021, USA.
| | | |
Collapse
|
23
|
Tawara T, Hasegawa K, Sugiura Y, Tahara T, Ishida I, Kataoka S. Fully human antibody exhibits pan-human leukocyte antigen-DR recognition and high in vitro/vivo efficacy against human leukocyte antigen-DR-positive lymphomas. Cancer Sci 2007; 98:921-8. [PMID: 17428256 PMCID: PMC11160095 DOI: 10.1111/j.1349-7006.2007.00469.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
HD8, a fully human monoclonal antibody specific for human leukocyte antigen-DR (HLA-DR), was generated by using the transchromosome mouse that bears the human immunoglobulin genes. HD8 could bind to all 13 tested HLA-DR-positive cell lines and 35 B-cells from healthy donors. Epitope mapping revealed that while the antibody recognizes the most polymorphic region of the HLA-DRB chain, its critical epitope residues are conserved in the major alleles. Indeed, HD8 could recognize 99.2% of HLA-DRB alleles. Since its essential epitope residues are also largely conserved in HLA-DP and HLA-DQ, HD8 could recognize 100% and 66% of the HLA-DP and HLA-DQ alleles tested, respectively. HD8 exerted strong antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity in vitro, and significantly extended the life span of immunocompromised mice inoculated with non-Hodgkin lymphoma cell lines. The HD8 antibody may be highly useful in HLA-DR-targeted immunotherapy as it is likely to evoke similarly strong responses in individuals carrying different HLA-DR alleles.
Collapse
Affiliation(s)
- Tomonori Tawara
- Pharmaceutical Research Laboratories, Kirin Brewing Company, Ltd, Gunma, Japan
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
B cells play an important role in the pathogenesis of certain lymphomas and leukemias, as well as many autoimmune diseases. Antagonistic B-cell antibodies are thus gaining an increasing role in the management of these diseases. The first antibody target in this regard was CD20, with the development and introduction of rituximab in the management of B-cell malignancies, as well as rheumatoid arthritis. A second candidate target is CD22. The first antagonistic antibody to this B-cell marker, epratuzumab, appears to function, in contrast to CD20 antibodies, more by modulation of B cells rather than by their high depletion in circulation. Originally developed for the treatment of non-Hodgkin's lymphoma, epratuzumab has now been found to be effective, with a very good safety profile, in two prototype autoimmune diseases: systemic lupus erythematosus and primary Sjögren's syndrome. Recent studies have demonstrated the activity and safety of epratuzumab in non-Hodgkin's lymphoma patients who have relapsed or are refractive to conventional therapy, including rituximab, and has also shown good activity in follicular and diffuse large B-cell lymphoma in combination with rituximab. As such, this new investigative antibody may have a significant market potential owing to the multitude of diseases and patients who may benefit from a CD22, B-cell antibody immunotherapy that is complementary to the known effects and role of CD20 antibodies, but can usually be administered within 1 h and depletes approximately 50% of circulating B cells.
Collapse
Affiliation(s)
- David M Goldenberg
- Garden State Cancer Center, Center for Molecular Medicine and Immunology 520 Belleville Avenue, Belleville, New Jersey 07109, USA.
| |
Collapse
|
25
|
Ding H, Prodinger WM, Kopecek J. Identification of CD21-binding peptides with phage display and investigation of binding properties of HPMA copolymer-peptide conjugates. Bioconjug Chem 2006; 17:514-23. [PMID: 16536485 DOI: 10.1021/bc0503162] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cancer targeting with peptides has become promising with the emergence of combinatorial peptide techniques such as phage display. Using phage display under stringent screening conditions, we selected five distinct peptides that specifically recognized the CD21 receptor, a cell surface marker of malignant B cell lymphoma. Two highly hydrophobic sequences were excluded (RLAYWCFSGLFLLVC and PVAAVSFVPYLVKTY). The binding affinity toward CD21 of the other three selected peptides (RMWPSSTVNLSAGRR, PNLDFSPTCSFRFGC, and GRVPSMFGGHFFFSR) was analyzed with fluorescence quenching. Their dissociation constants were determined to be within the micromolar range. On the basis of the results of phage ELISA, competitive phage ELISA, and fluorescence quenching, the binding sites of the three selected peptides were found to reside within the first four short consensus repeats of CD21 (SCR1-4). The peptide RMWPSSTVNLSAGRR (P1) was bound to the N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer, a potential drug carrier for chemotherapeutic agents, and the surface binding properties of HPMA copolymer-P1 conjugates were investigated. Specific interactions were observed between HPMA copolymer-P1 conjugates and surface-bound receptor. Binding of HPMA copolymer-P1 conjugates was directly related to the amount of surface (MaxiSorp plate) bound receptor, and the binding of the conjugates could be inhibited by the application of a 3-4 orders-of-magnitude excess of free peptide over the peptide concentration in conjugates. The enhanced binding of polymer-bound peptide was ascribed to multivalent interactions between the HPMA copolymer-P1 conjugate and the surface-bound CD21 receptor.
Collapse
Affiliation(s)
- Hui Ding
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
| | | | | |
Collapse
|
26
|
Current concepts of 131I therapy in oncology: Indications, methods and follow up. ARCHIVE OF ONCOLOGY 2006. [DOI: 10.2298/aoo0602045m] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Radioiodine in various forms (as sodium iodide and as the iodinated compounds MIBG, LIPIODOL, et al.) has been used as a therapeutic agent in oncology. Differentiated thyroid carcinoma (DTC) has been successfully treated by 131I therapy. Neuroendocrine tumors can be treated by palliative therapy, including Meta 131Iodobenzylguanidine therapy (131I-MIBG). Diagnostic 131I or 123I-MIBG scintigraphy is usually performed to image neuroblastoma and malignant pheochromocytoma. Following the establishment of the diagnosis, 131I-MIBG may be applied as a therapeutic agent but with limited success. Hepatocellular carcinoma (HCC) is treated by surgery only in 10% of patients. In others, palliative therapy should be administered. Radionuclide therapy for this disease is a therapeutic option with a major advantage compared to systemic chemotherapy, estrogen and progesterone therapy, and immunotherapy. 131I-lipiodol can be used to treat HCC without side effects. Compared to untreated patients, those who received 131Ilipiodol, showed significantly better survival and a decreased recurrence rate. The modern aspect of the neoplasm treatment involves radioimmunotherapy with radioiodine and some other radionuclides. Monoclonal antibody therapy with radioiodine has been extensively succeeded in the therapy of B-cell non-Hodgkin?s lymphoma, prostate cancer. Radioimmunotherapy is also efficiently performed in some other malignancies such as: medullary thyroid carcinoma, breast cancer, colorectal cancer and malignant brain tumors. Radioimmunotherapy will play a key role in the treatment of malignant diseases, especially hematopoietic neoplasms during this millennium.
Collapse
|