1
|
Li H, Lin WP, Zhang ZN, Sun ZJ. Tailoring biomaterials for monitoring and evoking tertiary lymphoid structures. Acta Biomater 2023; 172:1-15. [PMID: 37739247 DOI: 10.1016/j.actbio.2023.09.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/01/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
Despite the remarkable clinical success of immune checkpoint blockade (ICB) in the treatment of cancer, the response rate to ICB therapy remains suboptimal. Recent studies have strongly demonstrated that intratumoral tertiary lymphoid structures (TLSs) are associated with a good prognosis and a successful clinical response to immunotherapy. However, there is still a shortage of efficient and wieldy approaches to image and induce intratumoral TLSs in vivo. Biomaterials have made great strides in overcoming the deficiencies of conventional diagnosis and therapies for cancer, and antitumor therapy has also benefited from biomaterial-based drug delivery models. In this review, we summarize the reported methods for TLS imaging and induction based on biomaterials and provide potential strategies that can further enhance the effectiveness of imaging and stimulating intratumoral TLSs to predict and promote the response rates of ICB therapies for patients. STATEMENT OF SIGNIFICANCE: In this review, we focused on the promising of biomaterials for imaging and induction of TLSs. We reviewed the applications of biomaterials in molecular imaging and immunotherapy, identified the biomaterials that may be suitable for TLS imaging and induction, and provided outlooks for further research. Accurate imaging and effective induction of TLSs are of great significance for understanding the mechanism and clinical application. We highlighted the need for multidisciplinary coordination and cooperation in this field, and proposed the possible future direction of noninvasive imaging and artificial induction of TLSs based on biomaterials. We believe that it can facilitate collaboration and galvanize a broader effort.
Collapse
Affiliation(s)
- Hao Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, PR China; Department of Oral Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Wen-Ping Lin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, PR China
| | - Zhong-Ni Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, PR China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, PR China; Department of Oral Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China.
| |
Collapse
|
2
|
Ma X, Zhang MJ, Wang J, Zhang T, Xue P, Kang Y, Sun ZJ, Xu Z. Emerging Biomaterials Imaging Antitumor Immune Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2204034. [PMID: 35728795 DOI: 10.1002/adma.202204034] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/19/2022] [Indexed: 06/15/2023]
Abstract
Immunotherapy is one of the most promising clinical modalities for the treatment of malignant tumors and has shown excellent therapeutic outcomes in clinical settings. However, it continues to face several challenges, including long treatment cycles, high costs, immune-related adverse events, and low response rates. Thus, it is critical to predict the response rate to immunotherapy by using imaging technology in the preoperative and intraoperative. Here, the latest advances in nanosystem-based biomaterials used for predicting responses to immunotherapy via the imaging of immune cells and signaling molecules in the immune microenvironment are comprehensively summarized. Several imaging methods, such as fluorescence imaging, magnetic resonance imaging, positron emission tomography imaging, ultrasound imaging, and photoacoustic imaging, used in immune predictive imaging, are discussed to show the potential of nanosystems for distinguishing immunotherapy responders from nonresponders. Nanosystem-based biomaterials aided by various imaging technologies are expected to enable the effective prediction and diagnosis in cases of tumors, inflammation, and other public diseases.
Collapse
Affiliation(s)
- Xianbin Ma
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Meng-Jie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jingting Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Tian Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Peng Xue
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Yuejun Kang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| |
Collapse
|
3
|
Volpe A, Adusumilli PS, Schöder H, Ponomarev V. Imaging cellular immunotherapies and immune cell biomarkers: from preclinical studies to patients. J Immunother Cancer 2022; 10:jitc-2022-004902. [PMID: 36137649 PMCID: PMC9511655 DOI: 10.1136/jitc-2022-004902] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2022] [Indexed: 01/26/2023] Open
Abstract
Cellular immunotherapies have emerged as a successful therapeutic approach to fight a wide range of human diseases, including cancer. However, responses are limited to few patients and tumor types. An in-depth understanding of the complexity and dynamics of cellular immunotherapeutics, including what is behind their success and failure in a patient, the role of other immune cell types and molecular biomarkers in determining a response, is now paramount. As the cellular immunotherapy arsenal expands, whole-body non-invasive molecular imaging can shed a light on their in vivo fate and contribute to the reliable assessment of treatment outcome and prediction of therapeutic response. In this review, we outline the non-invasive strategies that can be tailored toward the molecular imaging of cellular immunotherapies and immune-related components, with a focus on those that have been extensively tested preclinically and are currently under clinical development or have already entered the clinical trial phase. We also provide a critical appraisal on the current role and consolidation of molecular imaging into clinical practice.
Collapse
Affiliation(s)
- Alessia Volpe
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA,Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York, USA,Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
4
|
The generation and application of antigen-specific T cell therapies for cancer and viral-associated disease. Mol Ther 2022; 30:2130-2152. [PMID: 35149193 PMCID: PMC9171249 DOI: 10.1016/j.ymthe.2022.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/27/2021] [Accepted: 02/03/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy with antigen-specific T cells is a promising, targeted therapeutic option for patients with cancer as well as for immunocompromised patients with virus infections. In this review, we characterize and compare current manufacturing protocols for the generation of T cells specific to viral and non-viral tumor-associated antigens. Specifically, we discuss: (1) the different methodologies to expand virus-specific T cell and non-viral tumor-associated antigen-specific T cell products, (2) an overview of the immunological principles involved when developing such manufacturing protocols, and (3) proposed standardized methodologies for the generation of polyclonal, polyfunctional antigen-specific T cells irrespective of donor source. Ex vivo expanded cells have been safely administered to treat numerous patients with virus-associated malignancies, hematologic malignancies, and solid tumors. Hence, we have performed a comprehensive review of the clinical trial results evaluating the safety, feasibility, and efficacy of these products in the clinic. In summary, this review seeks to provide new insights regarding antigen-specific T cell technology to benefit a rapidly expanding T cell therapy field.
Collapse
|
5
|
Sato N, Choyke PL. Whole-Body Imaging to Assess Cell-Based Immunotherapy: Preclinical Studies with an Update on Clinical Translation. Mol Imaging Biol 2022; 24:235-248. [PMID: 34816284 PMCID: PMC8983636 DOI: 10.1007/s11307-021-01669-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/28/2022]
Abstract
In the past decades, immunotherapies against cancers made impressive progress. Immunotherapy includes a broad range of interventions that can be separated into two major groups: cell-based immunotherapies, such as adoptive T cell therapies and stem cell therapies, and immunomodulatory molecular therapies such as checkpoint inhibitors and cytokine therapies. Genetic engineering techniques that transduce T cells with a cancer-antigen-specific T cell receptor or chimeric antigen receptor have expanded to other cell types, and further modulation of the cells to enhance cancer targeting properties has been explored. Because cell-based immunotherapies rely on cells migrating to target organs or tissues, there is a growing interest in imaging technologies that non-invasively monitor transferred cells in vivo. Here, we review whole-body imaging methods to assess cell-based immunotherapy using a variety of examples. Following a review of preclinically used cell tracking technologies, we consider the status of their clinical translation.
Collapse
Affiliation(s)
- Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 10/Rm. B3B406, 10 Center Dr, Bethesda, MD, 20892, USA.
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 10/Rm. B3B69F, 10 Center Dr, Bethesda, MD, 20892, USA
| |
Collapse
|
6
|
Li X, Yang X, Li Z, Zheng X, Peng YJ, Lin W, Zhou L, Cao D, Situ M, Tu Q, Huang H, Fan W, Feng G, Zhang X. Development of a Radiotracer for PET Imaging of the SNAP Tag. ACS OMEGA 2022; 7:7550-7555. [PMID: 35284707 PMCID: PMC8908366 DOI: 10.1021/acsomega.1c05856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/01/2022] [Indexed: 06/14/2023]
Abstract
Cell therapies have progressed to cures for hematopoietic disorders, neurodegenerative diseases, and cancer. However, only some patients can benefit from cell therapies even with prior screening. Due to the limited clinical methods to monitor the in vivo therapeutic functions of these transferred cells over time, the uncertain prognosis is hard to attenuate. Positron emission tomography (PET) cell tracking can provide comprehensive dynamic and spatial information on the proliferation status and whole-body distribution of the therapeutic cell. In this work, we designed and synthesized the first SNAP-tagged PET radiotracer. SNAP tag is an O 6-alkylguanine-DNA alkyltransferase that can form an irreversible bond with 18F-BG-surface for in vivo cell tracking based on a reporter gene system. 18F-BG-surface was obtained by the F-Al radiolabeling method in 32 ± 7% radiochemical yield and showed a high in vitro stability in mouse serum. SNAP-tagged cells could be selectively targeted by 18F-BG-surface both in vitro (4.81 ± 0.08%AD/106 cell vs 2.26 ± 0.10%AD/106 cell) and in vivo (1.90 ± 0.05 vs 0.55 ± 0.02% ID/g, p < 0.01).
Collapse
Affiliation(s)
- Xinling Li
- Department
of Nuclear Medicine, Sun Yat-sen University State Key Laboratory of
Oncology in South China; Collaborative Innovation Center for Cancer
Medicine, Sun Yat-sen University Cancer
Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Xiaochun Yang
- Department
of Nuclear Medicine, Sun Yat-sen University State Key Laboratory of
Oncology in South China; Collaborative Innovation Center for Cancer
Medicine, Sun Yat-sen University Cancer
Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Zhijian Li
- Department
of Nuclear Medicine, Sun Yat-sen University State Key Laboratory of
Oncology in South China; Collaborative Innovation Center for Cancer
Medicine, Sun Yat-sen University Cancer
Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Xiaobin Zheng
- Department
of Nuclear Medicine, Sun Yat-sen University State Key Laboratory of
Oncology in South China; Collaborative Innovation Center for Cancer
Medicine, Sun Yat-sen University Cancer
Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Yong-jian Peng
- State
Key Laboratory of Oncology in South China; Collaborative Innovation
Center for Cancer Medicine, Sun Yat-sen
University Cancer Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Wenjie Lin
- State
Key Laboratory of Oncology in South China; Collaborative Innovation
Center for Cancer Medicine, Sun Yat-sen
University Cancer Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Ling Zhou
- Sun
Yat-sen University State Key Laboratory of Oncology in South China;
Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Dehai Cao
- State
Key Laboratory of Oncology in South China; Collaborative Innovation
Center for Cancer Medicine, Sun Yat-sen
University Cancer Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Minyi Situ
- State
Key Laboratory of Oncology in South China; Collaborative Innovation
Center for Cancer Medicine, Sun Yat-sen
University Cancer Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Qingqiang Tu
- Laboratory
Animal Center, Sun Yat-sen University Zhongshan
School of Medicine, 74 Zhongshan 2th Road, Guangzhou, Guangdong Province 510085, China
| | - Huiqiang Huang
- State
Key Laboratory of Oncology in South China; Collaborative Innovation
Center for Cancer Medicine, Sun Yat-sen
University Cancer Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Wei Fan
- Department
of Nuclear Medicine, Sun Yat-sen University State Key Laboratory of
Oncology in South China; Collaborative Innovation Center for Cancer
Medicine, Sun Yat-sen University Cancer
Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Guokai Feng
- State
Key Laboratory of Oncology in South China; Collaborative Innovation
Center for Cancer Medicine, Sun Yat-sen
University Cancer Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| | - Xiaofei Zhang
- Department
of Nuclear Medicine, Sun Yat-sen University State Key Laboratory of
Oncology in South China; Collaborative Innovation Center for Cancer
Medicine, Sun Yat-sen University Cancer
Center, 651 Dongfengdong Road, Guangzhou, Guangdong Province 510060, China
| |
Collapse
|
7
|
Shalaby N, Dubois VP, Ronald J. Molecular imaging of cellular immunotherapies in experimental and therapeutic settings. Cancer Immunol Immunother 2021; 71:1281-1294. [PMID: 34657195 PMCID: PMC9122865 DOI: 10.1007/s00262-021-03073-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 09/28/2021] [Indexed: 11/27/2022]
Abstract
Cell-based cancer immunotherapies are becoming a routine part of the armamentarium against cancer. While remarkable successes have been seen, including durable remissions, not all patients will benefit from these therapies and many can suffer from life-threatening side effects. These differences in efficacy and safety across patients and across tumor types (e.g., blood vs. solid), are thought to be due to differences in how well the immune cells traffic to their target tissue (e.g., tumor, lymph nodes, etc.) whilst avoiding non-target tissues. Across patient variability can also stem from whether the cells interact with (i.e., communicate with) their intended target cells (e.g., cancer cells), as well as if they proliferate and survive long enough to yield potent and long-lasting therapeutic effects. However, many cell-based therapies are monitored by relatively simple blood tests that lack any spatial information and do not reflect how many immune cells have ended up at particular tissues. The ex vivo labeling and imaging of infused therapeutic immune cells can provide a more precise and dynamic understanding of whole-body immune cell biodistribution, expansion, viability, and activation status in individual patients. In recent years numerous cellular imaging technologies have been developed that may provide this much-needed information on immune cell fate. For this review, we summarize various ex vivo labeling and imaging approaches that allow for tracking of cellular immunotherapies for cancer. Our focus is on clinical imaging modalities and summarize the progression from experimental to therapeutic settings. The imaging information provided by these technologies can potentially be used for many purposes including improved real-time understanding of therapeutic efficacy and potential side effects in individual patients after cell infusion; the ability to more readily compare new therapeutic cell designs to current designs for various parameters such as improved trafficking to target tissues and avoidance of non-target tissues; and the long-term ability to identify patient populations that are likely to be positive responders and at low-risk of side effects.
Collapse
Affiliation(s)
- Nourhan Shalaby
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada.,Robarts Research Institute, London, Ontario, Canada
| | - Veronica Phyllis Dubois
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada.,Robarts Research Institute, London, Ontario, Canada
| | - John Ronald
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada. .,Robarts Research Institute, London, Ontario, Canada. .,Lawson Health Research Institute, London, Ontario, Canada.
| |
Collapse
|
8
|
Kiraga Ł, Kucharzewska P, Paisey S, Cheda Ł, Domańska A, Rogulski Z, Rygiel TP, Boffi A, Król M. Nuclear imaging for immune cell tracking in vivo – Comparison of various cell labeling methods and their application. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
9
|
Imaging CAR T-cell kinetics in solid tumors: Translational implications. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:355-367. [PMID: 34553024 PMCID: PMC8426175 DOI: 10.1016/j.omto.2021.06.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 06/08/2021] [Indexed: 01/22/2023]
Abstract
Success in solid tumor chimeric antigen receptor (CAR) T-cell therapy requires overcoming several barriers, including lung sequestration, inefficient accumulation within the tumor, and target-antigen heterogeneity. Understanding CAR T-cell kinetics can assist in the interpretation of therapy response and limitations and thereby facilitate developing successful strategies to treat solid tumors. As T-cell therapy response varies across metastatic sites, the assessment of CAR T-cell kinetics by peripheral blood analysis or a single-site tumor biopsy is inadequate for interpretation of therapy response. The use of tumor imaging alone has also proven to be insufficient to interpret response to therapy. To address these limitations, we conducted dual tumor and T-cell imaging by use of a bioluminescent reporter and positron emission tomography in clinically relevant mouse models of pleural mesothelioma and non-small cell lung cancer. We observed that the mode of delivery of T cells (systemic versus regional), T-cell activation status (presence or absence of antigen-expressing tumor), and tumor-antigen expression heterogeneity influence T-cell kinetics. The observations from our study underscore the need to identify and develop a T-cell reporter—in addition to standard parameters of tumor imaging and antitumor efficacy—that can be used for repeat imaging without compromising the efficacy of CAR T cells in vivo.
Collapse
|
10
|
Shao F, Long Y, Ji H, Jiang D, Lei P, Lan X. Radionuclide-based molecular imaging allows CAR-T cellular visualization and therapeutic monitoring. Am J Cancer Res 2021; 11:6800-6817. [PMID: 34093854 PMCID: PMC8171102 DOI: 10.7150/thno.56989] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy is a new and effective form of adoptive cell therapy that is rapidly entering the mainstream for the treatment of CD19-positive hematological cancers because of its impressive effect and durable responses. Huge challenges remain in achieving similar success in patients with solid tumors. The current methods of monitoring CAR-T, including morphological imaging (CT and MRI), blood tests, and biopsy, have limitations to assess whether CAR-T cells are homing to tumor sites and infiltrating into tumor bed, or to assess the survival, proliferation, and persistence of CAR-T cells in solid tumors associated with an immunosuppressive microenvironment. Radionuclide-based molecular imaging affords improved CAR-T cellular visualization and therapeutic monitoring through either a direct cellular radiolabeling approach or a reporter gene imaging strategy, and endogenous cell imaging is beneficial to reflect functional information and immune status of T cells. Focusing on the dynamic monitoring and precise assessment of CAR-T therapy, this review summarizes the current applications of radionuclide-based noninvasive imaging in CAR-T cells visualization and monitoring and presents current challenges and strategic choices.
Collapse
|
11
|
Volpe A, Pillarsetty NVK, Lewis JS, Ponomarev V. Applications of nuclear-based imaging in gene and cell therapy: probe considerations. MOLECULAR THERAPY-ONCOLYTICS 2021; 20:447-458. [PMID: 33718593 PMCID: PMC7907215 DOI: 10.1016/j.omto.2021.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/26/2021] [Indexed: 01/11/2023]
Abstract
Several types of gene- and cell-based therapeutics are now emerging in the cancer immunotherapy, transplantation, and regenerative medicine landscapes. Radionuclear-based imaging can be used as a molecular imaging tool for repetitive and non-invasive visualization as well as in vivo monitoring of therapy success. In this review, we discuss the principles of nuclear-based imaging and provide a comprehensive overview of its application in gene and cell therapy. This review aims to inform investigators in the biomedical field as well as clinicians on the state of the art of nuclear imaging, from probe design to available radiopharmaceuticals and advances of direct (probe-based) and indirect (transgene-based) strategies in both preclinical and clinical settings. Notably, as the nuclear-based imaging toolbox is continuously expanding, it will be increasingly incorporated into the clinical setting where the distribution, targeting, and persistence of a new generation of therapeutics can be imaged and ultimately guide therapeutic decisions.
Collapse
Affiliation(s)
- Alessia Volpe
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Naga Vara Kishore Pillarsetty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
12
|
Wu AM. Protein Engineering for Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00045-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
13
|
Abousaway O, Rakhshandehroo T, Van den Abbeele AD, Kircher MF, Rashidian M. Noninvasive Imaging of Cancer Immunotherapy. Nanotheranostics 2021; 5:90-112. [PMID: 33391977 PMCID: PMC7738948 DOI: 10.7150/ntno.50860] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy has revolutionized the treatment of several malignancies. Notwithstanding the encouraging results, many patients do not respond to treatments. Evaluation of the efficacy of treatments is challenging and robust methods to predict the response to treatment are not yet available. The outcome of immunotherapy results from changes that treatment evokes in the tumor immune landscape. Therefore, a better understanding of the dynamics of immune cells that infiltrate into the tumor microenvironment may fundamentally help in addressing this challenge and provide tools to assess or even predict the response. Noninvasive imaging approaches, such as PET and SPECT that provide whole-body images are currently seen as the most promising tools that can shed light on the events happening in tumors in response to treatment. Such tools can provide critical information that can be used to make informed clinical decisions. Here, we review recent developments in the field of noninvasive cancer imaging with a focus on immunotherapeutics and nuclear imaging technologies and will discuss how the field can move forward to address the challenges that remain unresolved.
Collapse
Affiliation(s)
- Omar Abousaway
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
| | - Taha Rakhshandehroo
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
| | - Annick D. Van den Abbeele
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02215, USA
| | - Moritz F. Kircher
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02215, USA
| | - Mohammad Rashidian
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
| |
Collapse
|
14
|
O'Reilly RJ, Prockop S, Hasan A, Doubrovina E. Therapeutic advantages provided by banked virus-specific T-cells of defined HLA-restriction. Bone Marrow Transplant 2020; 54:759-764. [PMID: 31431697 DOI: 10.1038/s41409-019-0614-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We have developed banks of EBV and CMV-specific T-cell lines generated from healthy seropositive third party donors and characterized them as to their HLA type, virus specificity, lack of alloreactivity, and HLA restriction. We here summarize results of studies employing these immediately accessible, broadly-applicable third party virus-specific T-cells for adoptive therapy of EBV lymphomas and CMV infections in allo-HCT recipients. We describe the characteristics contributing to their safety. We also discuss several distinctive advantages of banked third party virus-specific T-cells selected on the basis of their HLA restriction, particularly in the treatment of Rituximab-non-responsive EBV+ lymphomas and drug refractory CMV infections complicating HLA non-identical transplants.
Collapse
Affiliation(s)
- Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Susan Prockop
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aisha Hasan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ekaterina Doubrovina
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
15
|
Serganova I, Blasberg RG. Molecular Imaging with Reporter Genes: Has Its Promise Been Delivered? J Nucl Med 2020; 60:1665-1681. [PMID: 31792128 DOI: 10.2967/jnumed.118.220004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022] Open
Abstract
The first reporter systems were developed in the early 1980s and were based on measuring the activity of an enzyme-as a surrogate measure of promoter-driven transcriptional activity-which is now known as a reporter gene system. The initial objective and application of reporter techniques was to analyze the activity of a specific promoter (namely, the expression of a gene that is under the regulation of the specific promoter that is linked to the reporter gene). This system allows visualization of specific promoter activity with great sensitivity. In general, there are 2 classes of reporter systems: constitutively expressed (always-on) reporter constructs used for cell tracking, and inducible reporter systems sensitive to endogenous signaling molecules and transcription factors that characterize specific tissues, tumors, or signaling pathways.This review traces the development of different reporter systems, using fluorescent and bioluminescent proteins as well as radionuclide-based reporter systems. The development and application of radionuclide-based reporter systems is the focus of this review. The question at the end of the review is whether the "promise" of reporter gene imaging has been realized. What is required for moving forward with radionuclide-based reporter systems, and what is required for successful translation to clinical applications?
Collapse
Affiliation(s)
- Inna Serganova
- Department of Neurology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronald G Blasberg
- Department of Neurology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York .,Department of Radiology, Memorial Hospital, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York; and.,Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
16
|
McCarthy CE, White JM, Viola NT, Gibson HM. In vivo Imaging Technologies to Monitor the Immune System. Front Immunol 2020; 11:1067. [PMID: 32582173 PMCID: PMC7280489 DOI: 10.3389/fimmu.2020.01067] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
The past two decades have brought impressive advancements in immune modulation, particularly with the advent of both cancer immunotherapy and biologic therapeutics for inflammatory conditions. However, the dynamic nature of the immune response often complicates the assessment of therapeutic outcomes. Innovative imaging technologies are designed to bridge this gap and allow non-invasive visualization of immune cell presence and/or function in real time. A variety of anatomical and molecular imaging modalities have been applied for this purpose, with each option providing specific advantages and drawbacks. Anatomical methods including magnetic resonance imaging (MRI), computed tomography (CT), and ultrasound provide sharp tissue resolution, which can be further enhanced with contrast agents, including super paramagnetic ions (for MRI) or nanobubbles (for ultrasound). Conjugation of the contrast material to an antibody allows for specific targeting of a cell population or protein of interest. Protein platforms including antibodies, cytokines, and receptor ligands are also popular choices as molecular imaging agents for positron emission tomography (PET), single-photon emission computerized tomography (SPECT), scintigraphy, and optical imaging. These tracers are tagged with either a radioisotope or fluorescent molecule for detection of the target. During the design process for immune-monitoring imaging tracers, it is important to consider any potential downstream physiologic impact. Antibodies may deplete the target cell population, trigger or inhibit receptor signaling, or neutralize the normal function(s) of soluble proteins. Alternatively, the use of cytokines or other ligands as tracers may stimulate their respective signaling pathways, even in low concentrations. As in vivo immune imaging is still in its infancy, this review aims to describe the modalities and immunologic targets that have thus far been explored, with the goal of promoting and guiding the future development and application of novel imaging technologies.
Collapse
Affiliation(s)
- Claire E McCarthy
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Jordan M White
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Nerissa T Viola
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Heather M Gibson
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| |
Collapse
|
17
|
Pancreatic Cancer UK Grand Challenge: Developments and challenges for effective CAR T cell therapy for pancreatic ductal adenocarcinoma. Pancreatology 2020; 20:394-408. [PMID: 32173257 DOI: 10.1016/j.pan.2020.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
Death from pancreatic ductal adenocarcinoma (PDAC) is rising across the world and PDAC is predicted to be the second most common cause of cancer death in the USA by 2030. Development of effective biotherapies for PDAC are hampered by late presentation, a low number of differentially expressed molecular targets and a tumor-promoting microenvironment that forms both a physical, collagen-rich barrier and is also immunosuppressive. In 2017 Pancreatic Cancer UK awarded its first Grand Challenge Programme award to tackle this problem. The team plan to combine the use of novel CAR T cells with strategies to overcome the barriers presented by the tumor microenvironment. In advance of publication of those data this review seeks to highlight the key problems in effective CAR T cell therapy of PDAC and to describe pre-clinical and clinical progress in CAR T bio-therapeutics.
Collapse
|
18
|
Martinez O, Sosabowski J, Maher J, Papa S. New Developments in Imaging Cell-Based Therapy. J Nucl Med 2019; 60:730-735. [PMID: 30979822 PMCID: PMC6581223 DOI: 10.2967/jnumed.118.213348] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/19/2019] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy is now established as a central therapeutic pillar in hematologic oncology. Cell-based therapies, with or without genetic modification ex vivo, have reached the clinic as the standard of care in limited indications and remain the subject of intense preclinical and translational development. Expanding on this, related therapeutic approaches are in development for solid-tumor and nonmalignant indications, broadening the scope of this technology. It has long been recognized that in vivo tracking of infused cellular therapies would provide unique opportunities to optimize their efficacy and aid in the assessment and management of toxicity. Recently, we have witnessed the introduction of novel tracers for passive labeling of cell products and advances in the introduction and use of reporter genes to enable longitudinal imaging. This review highlights the key developments over the last 5 y.
Collapse
Affiliation(s)
- Olivier Martinez
- ImmunoEngineering Group, School of Cancer and Pharmaceutical Sciences, King's Health Partners Integrated Cancer Centre, Guy's Hospital, King's College London, London, United Kingdom
| | - Jane Sosabowski
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - John Maher
- CAR Mechanics Group, School of Cancer and Pharmaceutical Sciences, King's Health Partners Integrated Cancer Centre, Guy's Hospital, King's College London, London, United Kingdom
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Department of Immunology, Eastbourne Hospital, Eastbourne, United Kingdom; and
| | - Sophie Papa
- ImmunoEngineering Group, School of Cancer and Pharmaceutical Sciences, King's Health Partners Integrated Cancer Centre, Guy's Hospital, King's College London, London, United Kingdom
- Department of Medical Oncology, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
19
|
Che W, Zhang L, Li Y, Zhu D, Xie Z, Li G, Zhang P, Su Z, Dou C, Tang BZ. Ultrafast and Noninvasive Long-Term Bioimaging with Highly Stable Red Aggregation-Induced Emission Nanoparticles. Anal Chem 2019; 91:3467-3474. [PMID: 30693764 DOI: 10.1021/acs.analchem.8b05024] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Strongly red luminescent and water-soluble probes are very important for studying biological events and processes. Fluorescent nanoparticles (NPs) built from the aggregation-induced emission luminogen (AIEgen) and amphipathic polymeric matrixes have been considered as promising candidates for bioimaging. However, AIE NPs with long-wavelength absorption suitable for in vivo application are still scarce. In this work, three AIE-active red-emissive BODIPY derivatives with long-wavelength absorption were rationally designed and synthesized. Then three NPs based on these AIEgens exhibit bright red photoluminescence with high fluorescence quantum yield in aqueous media. These NPs uniformly dispersed in water and showed excellent stability and good biocompatibility. They can be readily internalized by HeLa cells, and the staining process is performed by simply shaking the culture with cells for just a few seconds at room temperature, which indicates an ultrafast and easy-to-operate staining protocol. More importantly, long-term tracing in living cells and mouse over 15 days is successfully achieved. The strong fluorescence signals, ultrafast staining procedure, and long-term tracing abilities indicate that these AIE NPs hold great potential for monitoring biological processes.
Collapse
Affiliation(s)
- Weilong Che
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry , Northeast Normal University , 5268 Renmin Street , Changchun , Jilin Province 130024 , P. R. China
| | - Liping Zhang
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry , Northeast Normal University , 5268 Renmin Street , Changchun , Jilin Province 130024 , P. R. China
| | - Yuanyuan Li
- State Key Laboratory of Polymer Physics and Chemistry , Changchun Institute of Applied Chemistry Chinese Academy of Sciences , Changchun , 130022 , P. R. China
| | - Dongxia Zhu
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry , Northeast Normal University , 5268 Renmin Street , Changchun , Jilin Province 130024 , P. R. China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry , Changchun Institute of Applied Chemistry Chinese Academy of Sciences , Changchun , 130022 , P. R. China
| | - Guangfu Li
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry , Northeast Normal University , 5268 Renmin Street , Changchun , Jilin Province 130024 , P. R. China
| | - Pengfei Zhang
- Department of Chemistry Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction , The Hong Kong University of Science and Technology , Clear Water Bay, Kowloon , Hong Kong , China
| | - Zhongmin Su
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry , Northeast Normal University , 5268 Renmin Street , Changchun , Jilin Province 130024 , P. R. China
- School of Chemistry and Environmental Engineering , Changchun University of Science and Technology , Changchun , 130022 , P. R. China
| | - Chuandong Dou
- State Key Laboratory of Polymer Physics and Chemistry , Changchun Institute of Applied Chemistry Chinese Academy of Sciences , Changchun , 130022 , P. R. China
| | - Ben Zhong Tang
- Department of Chemistry Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction , The Hong Kong University of Science and Technology , Clear Water Bay, Kowloon , Hong Kong , China
| |
Collapse
|
20
|
Krzastek SC, Goliadze E, Zhou S, Petrossian A, Youniss F, Sundaresan G, Wang L, Zweit J, Guruli G. Dendritic cell trafficking in tumor-bearing mice. Cancer Immunol Immunother 2018; 67:1939-1947. [PMID: 29943070 PMCID: PMC11028156 DOI: 10.1007/s00262-018-2187-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 06/13/2018] [Indexed: 12/16/2022]
Abstract
Prostate cancer is one of the leading causes of cancer deaths, with no curative treatments once it spreads. Alternative therapies, including immunotherapy, have shown limited efficacy. Dendritic cells (DC) have been widely used in the treatment of various malignancies. DC capture antigens and move to the lymphoid organs where they prime naive T cells. Interaction between DC and T cells are most active in lymph nodes and suppression of DC trafficking to lymph nodes impairs the immune response. In this work, we aimed to study trafficking of DC in vivo via various routes of delivery, to optimize the effectiveness of DC-based therapy. A DC labeling system was developed using 1,1'-dioctadecyltetramethyl indotricarbocyanine Iodine for in vivo fluorescent imaging. DC harvested from C57B/6 mice were matured, labeled, and injected intravenously, subcutaneously, or intratumorally, with or without antigen loading with whole tumor lysate, into C57B/6 mice inoculated with RM-1 murine prostate tumor cells. Signal intensity was measured in vivo and ex vivo. Signal intensity at the tumor site increased over time, suggesting trafficking of DC to the tumor with all modes of injection. Subcutaneous injection showed preferential trafficking to lymph nodes and tumor. Intravenous injection showed trafficking to lungs, intestines, and spleen. Subcutaneous injection of DC pulsed with whole tumor lysate resulted in the highest increase in signal intensity at the tumor site and lymph nodes, suggesting subcutaneous injection of primed DC leads to highest preferential trafficking of DC to the immunocompetent organs.
Collapse
Affiliation(s)
- Sarah C Krzastek
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Ekaterine Goliadze
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Shaoqing Zhou
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Albert Petrossian
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Fatma Youniss
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Gobalakrishnan Sundaresan
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Li Wang
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Jamal Zweit
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Georgi Guruli
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
21
|
Krebs S, Ahad A, Carter LM, Eyquem J, Brand C, Bell M, Ponomarev V, Reiner T, Meares CF, Gottschalk S, Sadelain M, Larson SM, Weber WA. Antibody with Infinite Affinity for In Vivo Tracking of Genetically Engineered Lymphocytes. J Nucl Med 2018; 59:1894-1900. [PMID: 29903928 DOI: 10.2967/jnumed.118.208041] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/07/2018] [Indexed: 12/12/2022] Open
Abstract
There remains an urgent need for the noninvasive tracking of transfused chimeric antigen receptor (CAR) T cells to determine their biodistribution, viability, expansion, and antitumor functionality. DOTA antibody reporter 1 (DAbR1) comprises a single-chain fragment of the antilanthanoid-DOTA antibody 2D12.5/G54C fused to the human CD4-transmembrane domain and binds irreversibly to lanthanoid (S)-2-(4-acrylamidobenzyl)-DOTA (AABD). The aim of this study was to investigate whether DAbR1 can be expressed on lymphocytes and used as a reporter gene as well as a suicide gene for therapy of immune-related adverse effects. Methods: DAbR1 was subcloned together with green fluorescent protein into an SFG-retroviral vector and used to transduce CD3/CD28-activated primary human T cells and second-generation 1928z (CAR) T cells. Cell surface expression of DAbR1 was confirmed by cell uptake studies with radiolabeled AABD. In addition, the feasibility of imaging of DAbR1-positive T cells in vivo after intravenous injection of 86Y/177Lu-AABD was studied and radiation doses determined. Results: A panel of DAbR1-expressing T cells and CAR T cells exhibited greater than 8-fold increased uptake of 86Y-AABD in vitro when compared with nontransduced cells. Imaging studies showed 86Y-AABD was retained by DAbR1-positive T cells while it continuously cleared from normal tissues, allowing for in vivo tracking of intravenously administered CAR T cells. Normal-organ dose estimates were favorable for repeated PET/CT studies. Selective T cell ablation in vivo with 177Lu-AABD seems feasible for clustered T-cell populations. Conclusion: We have demonstrated for the first time that T cells can be modified with DAbR1, enabling their in vivo tracking via PET and SPECT. The favorable biodistribution and high image contrast observed warrant further studies of this new reporter gene.
Collapse
Affiliation(s)
- Simone Krebs
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Afruja Ahad
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lukas M Carter
- Radiochemistry and Molecular Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin Eyquem
- Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, New York, New York
| | - Christian Brand
- Radiochemistry and Molecular Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Meghan Bell
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Ponomarev
- Radiochemistry and Molecular Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas Reiner
- Radiochemistry and Molecular Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Claude F Meares
- Chemistry Department, University of California, Davis, California
| | - Stephen Gottschalk
- Department of Bone Marrow Transplant and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee; and
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, New York, New York
| | - Steven M Larson
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wolfgang A Weber
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
22
|
Emami-Shahri N, Foster J, Kashani R, Gazinska P, Cook C, Sosabowski J, Maher J, Papa S. Clinically compliant spatial and temporal imaging of chimeric antigen receptor T-cells. Nat Commun 2018. [PMID: 29540684 PMCID: PMC5852048 DOI: 10.1038/s41467-018-03524-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The unprecedented efficacy of chimeric antigen receptor (CAR) T-cell immunotherapy of CD19+ B-cell malignancy has established a new therapeutic pillar of hematology–oncology. Nonetheless, formidable challenges remain for the attainment of comparable success in patients with solid tumors. To accelerate progress and rapidly characterize emerging toxicities, systems that permit the repeated and non-invasive assessment of CAR T-cell bio-distribution would be invaluable. An ideal solution would entail the use of a non-immunogenic reporter that mediates specific uptake of an inexpensive, non-toxic and clinically established imaging tracer by CAR T cells. Here we show the utility of the human sodium iodide symporter (hNIS) for the temporal and spatial monitoring of CAR T-cell behavior in a cancer-bearing host. This system provides a clinically compliant toolkit for high-resolution serial imaging of CAR T cells in vivo, addressing a fundamental unmet need for future clinical development in the field. Adoptive transfer of chimeric antigen receptor (CAR) T cells has shown promising anticancer results in clinical trials. Here the authors use the human sodium iodide symporter (hNIS) as a reporter gene to image human CAR T cells in cancer-bearing mice using broadly available tracers and imaging platforms.
Collapse
Affiliation(s)
- Nia Emami-Shahri
- ImmunoEngineering Group, King's College London, Division of Cancer Studies, 3rd Floor Bermondsey Wing, King's Health Partners Integrated Cancer Centre, Great Maze Pond, Guy's Hospital, London, SE1 9RT, UK
| | - Julie Foster
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Roxana Kashani
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Patrycja Gazinska
- Breast Cancer Now, Division of Cancer Studies, Guy's Cancer Centre, Great Maze Pond, London, SE1 9RT, UK
| | - Celia Cook
- ImmunoEngineering Group, King's College London, Division of Cancer Studies, 3rd Floor Bermondsey Wing, King's Health Partners Integrated Cancer Centre, Great Maze Pond, Guy's Hospital, London, SE1 9RT, UK
| | - Jane Sosabowski
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - John Maher
- CAR Mechanics Group, King's College London, Division of Cancer Studies, 3rd Floor Bermondsey Wing, King's Health Partners Integrated Cancer Centre, Great Maze Pond, Guy's Hospital, London, SE1 9RT, UK.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, SE5 9RS, UK.,Department of Immunology, Eastbourne Hospital, King's Drive, Eastbourne, BN21 2UD, UK
| | - Sophie Papa
- ImmunoEngineering Group, King's College London, Division of Cancer Studies, 3rd Floor Bermondsey Wing, King's Health Partners Integrated Cancer Centre, Great Maze Pond, Guy's Hospital, London, SE1 9RT, UK. .,Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, SE1 9RT, UK.
| |
Collapse
|
23
|
Abstract
Immunotherapies include various approaches, ranging from stimulating effector mechanisms to counteracting inhibitory and suppressive mechanisms, and creating a forum for discussing the most effective means of advancing these therapies through imaging is the focus of the newly formed Imaging in Cellular and Immune Therapies (ICIT) interest group within the World Molecular Imaging Society. Efforts are being made in the identification and validation of predictive biomarkers for a number of immunotherapies. Without predictive biomarkers, a considerable number of patients may receive treatments that have no chance of offering a benefit. This will reflect poorly on the field of immunotherapy and will yield false hopes in patients while at the same time contributing to significant cost to the healthcare system. This review summarizes the main strategies in cancer immune and cell-based therapies and discusses recent advances in imaging strategies aimed to improve cancer immunotherapy outcomes.
Collapse
Affiliation(s)
- Vladimir Ponomarev
- Department of Radiology, Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Ave Z-2063, Box 501, New York, NY, 10065, USA.
| |
Collapse
|
24
|
Jurgielewicz P, Harmsen S, Wei E, Bachmann MH, Ting R, Aras O. New imaging probes to track cell fate: reporter genes in stem cell research. Cell Mol Life Sci 2017; 74:4455-4469. [PMID: 28674728 DOI: 10.1007/s00018-017-2584-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 06/06/2017] [Accepted: 06/26/2017] [Indexed: 01/09/2023]
Abstract
Cell fate is a concept used to describe the differentiation and development of a cell in its organismal context over time. It is important in the field of regenerative medicine, where stem cell therapy holds much promise but is limited by our ability to assess its efficacy, which is mainly due to the inability to monitor what happens to the cells upon engraftment to the damaged tissue. Currently, several imaging modalities can be used to track cells in the clinical setting; however, they do not satisfy many of the criteria necessary to accurately assess several aspects of cell fate. In recent years, reporter genes have become a popular option for tracking transplanted cells, via various imaging modalities in small mammalian animal models. This review article examines the reporter gene strategies used in imaging modalities such as MRI, SPECT/PET, Optoacoustic and Bioluminescence Imaging. Strengths and limitations of the use of reporter genes in each modality are discussed.
Collapse
Affiliation(s)
- Piotr Jurgielewicz
- Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Stefan Harmsen
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | | | | | - Richard Ting
- Department of Radiology, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, Suite 1511, New York, NY, 10065, USA.
| |
Collapse
|
25
|
Yusufi N, Mall S, Bianchi HDO, Steiger K, Reder S, Klar R, Audehm S, Mustafa M, Nekolla S, Peschel C, Schwaiger M, Krackhardt AM, D`Alessandria C. In-depth Characterization of a TCR-specific Tracer for Sensitive Detection of Tumor-directed Transgenic T Cells by Immuno-PET. Am J Cancer Res 2017; 7:2402-2416. [PMID: 28744323 PMCID: PMC5525745 DOI: 10.7150/thno.17994] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/22/2017] [Indexed: 12/19/2022] Open
Abstract
A number of different technologies have been developed to monitor in vivo the distribution of gene-modified T cells used in immunotherapy. Nevertheless, in-depth characterization of novel approaches with respect to sensitivity and clinical applicability are so far missing. We have previously described a novel method to track engineered human T cells in tumors using 89Zr-Df-aTCRmu-F(ab')2 targeting the murinized part of the TCR beta domain (TCRmu) of a transgenic TCR. Here, we performed an in-depth in vitro characterization of the tracer in terms of antigen affinity, immunoreactivity, influence on T-cell functionality and stability in vitro and in vivo. Of particular interest, we have developed diverse experimental settings to quantify TCR-transgenic T cells in vivo. Local application of 89Zr-Df-aTCRmu-F(ab')2-labeled T cells in a spot-assay revealed signal detection down to approximately 1.8x104 cells. In a more clinically relevant model, NSG mice were intravenously injected with different numbers of transgenic T cells, followed by injection of the 89Zr-Df-aTCRmu-F(ab')2 tracer, PET/CT imaging and subsequent ex vivo T-cell quantification in the tumor. Using this setting, we defined a comparable detection limit of 1.0x104 T cells. PET signals correlated well to total numbers of transgenic T cells detected ex vivo independently of the engraftment rates observed in different individual experiments. Thus, these findings confirm the high sensitivity of our novel PET/CT T-cell tracking method and provide critical information about the quantity of transgenic T cells in the tumor environment suggesting our technology being highly suitable for further clinical translation.
Collapse
|
26
|
Abstract
Chimaeric antigen receptor (CAR) therapy is entering the mainstream for the treatment of CD19(+)cancers. As is does we learn more about resistance to therapy and the role, risks and management of toxicity. In solid tumour CAR therapy research the route to the clinic is less smooth with a wealth of challenges facing translating this, potentially hugely valuable, therapeutic option for patients. As we strive to understand our successes, and navigate the challenges, having a clear understanding of how adoptively transferred CAR-T-cells behavein vivoand in human trials is invaluable. Harnessing reporter gene imaging to enable detection and tracking of small numbers of CAR-T-cells after adoptive transfer is one way by which we can accomplish this. The compatibility of certain reporter gene systems with tracers available routinely in the clinic makes this approach highly useful for future appraisal of CAR-T-cell success in humans.
Collapse
|
27
|
Vedvyas Y, Shevlin E, Zaman M, Min IM, Amor-Coarasa A, Park S, Park S, Kwon KW, Smith T, Luo Y, Kim D, Kim Y, Law B, Ting R, Babich J, Jin MM. Longitudinal PET imaging demonstrates biphasic CAR T cell responses in survivors. JCI Insight 2016; 1:e90064. [PMID: 27882353 DOI: 10.1172/jci.insight.90064] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Clinical monitoring of adoptive T cell transfer (ACT) utilizes serial blood analyses to discern T cell activity. While useful, these data are 1-dimensional and lack spatiotemporal information related to treatment efficacy or toxicity. We utilized a human genetic reporter, somatostatin receptor 2 (SSTR2), and PET, to quantitatively and longitudinally visualize whole-body T cell distribution and antitumor dynamics using a clinically approved radiotracer. Initial evaluations determined that SSTR2-expressing T cells were detectable at low densities with high sensitivity and specificity. SSTR2-based PET was applied to ACT of chimeric antigen receptor (CAR) T cells targeting intercellular adhesion molecule-1, which is overexpressed in anaplastic thyroid tumors. Timely CAR T cell infusions resulted in survival of tumor-bearing mice, while later infusions led to uniform death. Real-time PET imaging revealed biphasic T cell expansion and contraction at tumor sites among survivors, with peak tumor burden preceding peak T cell burden by several days. In contrast, nonsurvivors displayed unrelenting increases in tumor and T cell burden, indicating that tumor growth was outpacing T cell killing. Thus, longitudinal PET imaging of SSTR2-positive ACT dynamics enables prognostic, spatiotemporal monitoring with unprecedented clarity and detail to facilitate comprehensive therapy evaluation with potential for clinical translation.
Collapse
Affiliation(s)
- Yogindra Vedvyas
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA.,Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Enda Shevlin
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Marjan Zaman
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Irene M Min
- Department of Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Alejandro Amor-Coarasa
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Spencer Park
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA.,Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Susan Park
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Keon-Woo Kwon
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Turner Smith
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Yonghua Luo
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Dohyun Kim
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Young Kim
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA.,Department of Pathology, Chonnam National University Medical School, Gwangju, South Korea
| | - Benedict Law
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Richard Ting
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - John Babich
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Moonsoo M Jin
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York, USA.,Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA.,Department of Surgery, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
28
|
Li A, Wu Y, Linnoila J, Pulli B, Wang C, Zeller M, Ali M, Lewandrowski GK, Li J, Tricot B, Keliher E, Wojtkiewicz GR, Fulci G, Feng X, Tannous BA, Yao Z, Chen JW. Surface biotinylation of cytotoxic T lymphocytes for in vivo tracking of tumor immunotherapy in murine models. Cancer Immunol Immunother 2016; 65:1545-1554. [PMID: 27722909 DOI: 10.1007/s00262-016-1911-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 09/29/2016] [Indexed: 10/20/2022]
Abstract
Currently, there is no stable and flexible method to label and track cytotoxic T lymphocytes (CTLs) in vivo in CTL immunotherapy. We aimed to evaluate whether the sulfo-hydroxysuccinimide (NHS)-biotin-streptavidin (SA) platform could chemically modify the cell surface of CTLs for in vivo tracking. CD8+ T lymphocytes were labeled with sulfo-NHS-biotin under different conditions and then incubated with SA-Alexa647. Labeling efficiency was proportional to sulfo-NHS-biotin concentration. CD8+ T lymphocytes could be labeled with higher efficiency with sulfo-NHS-biotin in DPBS than in RPMI (P < 0.05). Incubation temperature was not a key factor. CTLs maintained sufficient labeling for at least 72 h (P < 0.05), without altering cell viability. After co-culturing labeled CTLs with mouse glioma stem cells (GSCs) engineered to present biotin on their surface, targeting CTLs could specifically target biotin-presenting GSCs and inhibited cell proliferation (P < 0.01) and tumor spheres formation. In a biotin-presenting GSC brain tumor model, targeting CTLs could be detected in biotin-presenting gliomas in mouse brains but not in the non-tumor-bearing contralateral hemispheres (P < 0.05). In vivo fluorescent molecular tomography imaging in a subcutaneous U87 mouse model confirmed that targeting CTLs homed in on the biotin-presenting U87 tumors but not the control U87 tumors. PET imaging with 89Zr-deferoxamine-biotin and SA showed a rapid clearance of the PET signal over 24 h in the control tumor, while only minimally decreased in the targeted tumor. Thus, sulfo-NHS-biotin-SA labeling is an efficient method to noninvasively track the migration of adoptive transferred CTLs and does not alter CTL viability or interfere with CTL-mediated cytotoxic activity.
Collapse
Affiliation(s)
- Anning Li
- Department of Radiology, Huashan Hospital, Fudan University, 12 Urumchi Road, Shanghai, 200040, China.,Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA.,Department of Radiology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, China
| | - Yue Wu
- Department of Radiology, Huashan Hospital, Fudan University, 12 Urumchi Road, Shanghai, 200040, China.,Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Jenny Linnoila
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Benjamin Pulli
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA.,Department of Radiology, Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Cuihua Wang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Matthias Zeller
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Muhammad Ali
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Grant K Lewandrowski
- Molecular Neurogenetics Unit, Neuroscience Center, 149 13th St., Charlestown, MA, 02129, USA
| | - Jinghui Li
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Benoit Tricot
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Edmund Keliher
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Gregory R Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Giulia Fulci
- Brain Tumor Research Center, Simches Research Building, Neurosurgery Service, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Xiaoyuan Feng
- Department of Radiology, Huashan Hospital, Fudan University, 12 Urumchi Road, Shanghai, 200040, China
| | - Bakhos A Tannous
- Molecular Neurogenetics Unit, Neuroscience Center, 149 13th St., Charlestown, MA, 02129, USA
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, 12 Urumchi Road, Shanghai, 200040, China.
| | - John W Chen
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA. .,Department of Radiology, Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, MA, 02114, USA.
| |
Collapse
|
29
|
Mall S, Yusufi N, Wagner R, Klar R, Bianchi H, Steiger K, Straub M, Audehm S, Laitinen I, Aichler M, Peschel C, Ziegler S, Mustafa M, Schwaiger M, D'Alessandria C, Krackhardt AM. Immuno-PET Imaging of Engineered Human T Cells in Tumors. Cancer Res 2016; 76:4113-23. [PMID: 27354381 DOI: 10.1158/0008-5472.can-15-2784] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 03/13/2016] [Indexed: 11/16/2022]
Abstract
Sensitive in vivo imaging technologies applicable to the clinical setting are still lacking for adoptive T-cell-based immunotherapies, an important gap to fill if mechanisms of tumor rejection or escape are to be understood. Here, we propose a highly sensitive imaging technology to track human TCR-transgenic T cells in vivo by directly targeting the murinized constant TCR beta domain (TCRmu) with a zirconium-89 ((89)Zr)-labeled anti-TCRmu-F(ab')2 fragment. Binding of the labeled or unlabeled F(ab')2 fragment did not impair functionality of transgenic T cells in vitro and in vivo Using a murine xenograft model of human myeloid sarcoma, we monitored by Immuno-PET imaging human central memory T cells (TCM), which were transgenic for a myeloid peroxidase (MPO)-specific TCR. Diverse T-cell distribution patterns were detected by PET/CT imaging, depending on the tumor size and rejection phase. Results were confirmed by IHC and semiquantitative evaluation of T-cell infiltration within the tumor corresponding to the PET/CT images. Overall, these findings offer a preclinical proof of concept for an imaging approach that is readily tractable for clinical translation. Cancer Res; 76(14); 4113-23. ©2016 AACR.
Collapse
Affiliation(s)
- Sabine Mall
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Nahid Yusufi
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Munich, Germany
| | - Ricarda Wagner
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Richard Klar
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Henrique Bianchi
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Katja Steiger
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Melanie Straub
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Stefan Audehm
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Iina Laitinen
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Munich, Germany
| | - Michaela Aichler
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Munich, Germany
| | - Christian Peschel
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany. German Cancer Consortium (DKTK), Munich, Germany
| | - Sibylle Ziegler
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Munich, Germany
| | - Mona Mustafa
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Munich, Germany
| | - Markus Schwaiger
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Munich, Germany. German Cancer Consortium (DKTK), Munich, Germany
| | - Calogero D'Alessandria
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Munich, Germany
| | - Angela M Krackhardt
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany. German Cancer Consortium (DKTK), Munich, Germany. Clinical Cooperation Group Antigen Specific T-Cell Therapy, Helmholtz Zentrum München, Munich, Germany.
| |
Collapse
|
30
|
Abstract
Positron emission tomography (PET) is a powerful noninvasive imaging technique able to measure distinct biological processes in vivo by administration of a radiolabeled probe. Whole-body measurements track the probe accumulation providing a means to measure biological changes such as metabolism, cell location, or tumor burden. PET can also be applied to both preclinical and clinical studies providing three-dimensional information. For immunotherapies (in particular understanding T cell responses), PET can be utilized for spatial and longitudinal tracking of T lymphocytes. Although PET has been utilized clinically for over 30 years, the recent development of additional PET radiotracers have dramatically expanded the use of PET to detect endogenous or adoptively transferred T cells in vivo. Novel probes have identified changes in T cell quantity, location, and function. This has enabled investigators to track T cells outside of the circulation and in hematopoietic organs such as spleen, lymph nodes, and bone marrow, or within tumors. In this review, we cover advances in PET detection of the antitumor T cell response and areas of focus for future studies.
Collapse
|
31
|
Juergens RA, Zukotynski KA, Singnurkar A, Snider DP, Valliant JF, Gulenchyn KY. Imaging Biomarkers in Immunotherapy. BIOMARKERS IN CANCER 2016; 8:1-13. [PMID: 26949344 PMCID: PMC4768940 DOI: 10.4137/bic.s31805] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/20/2015] [Accepted: 12/22/2015] [Indexed: 12/23/2022]
Abstract
Immune-based therapies have been in use for decades but recent work with immune checkpoint inhibitors has now changed the landscape of cancer treatment as a whole. While these advances are encouraging, clinicians still do not have a consistent biomarker they can rely on that can accurately select patients or monitor response. Molecular imaging technology provides a noninvasive mechanism to evaluate tumors and may be an ideal candidate for these purposes. This review provides an overview of the mechanism of action of varied immunotherapies and the current strategies for monitoring patients with imaging. We then describe some of the key researches in the preclinical and clinical literature on the current uses of molecular imaging of the immune system and cancer.
Collapse
Affiliation(s)
| | - Katherine A Zukotynski
- Department of Radiology, McMaster University, Hamilton, ON, Canada.; Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amit Singnurkar
- Department of Radiology, McMaster University, Hamilton, ON, Canada.; Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Denis P Snider
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - John F Valliant
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Karen Y Gulenchyn
- Department of Radiology, McMaster University, Hamilton, ON, Canada.; Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
32
|
Vaidyanathan G, McDougald D, Koumarianou E, Choi J, Hens M, Zalutsky MR. Synthesis and evaluation of 4-[18F]fluoropropoxy-3-iodobenzylguanidine ([18F]FPOIBG): A novel 18F-labeled analogue of MIBG. Nucl Med Biol 2015; 42:673-84. [PMID: 25956997 PMCID: PMC4481138 DOI: 10.1016/j.nucmedbio.2015.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Radioiodinated meta-iodobenzylguanidine (MIBG), a norepinephrine transporter (NET) substrate, has been extensively used as an imaging agent to study the pathophysiology of the heart and for the diagnosis and treatment of neuroendocrine tumors. The goal of this study was to develop an (18)F-labeled analogue of MIBG that like MIBG itself could be synthesized in a single radiochemical step. Towards this end, we designed 4-fluoropropoxy-3-iodobenzylguanidine (FPOIBG). METHODS Standards of FPOIBG and 4-fluoropropoxy-3-bromobenzylguanidine (FPOBBG) as well as their tosylate precursors for labeling with (18)F, and a tin precursor for the preparation of radioiodinated FPOIBG were synthesized. Radiolabeled derivatives were synthesized by nucleophilic substitution and electrophilic iododestannylation from the corresponding precursors. Labeled compounds were evaluated for NET transporter recognition in in vitro assays using three NET-expressing cell lines and in biodistribution experiments in normal mice, with all studies performed in a paired-label format. Competitive inhibition of [(125)I]MIBG uptake by unlabeled benzylguanidine compounds was performed in UVW-NAT cell line to determine IC50 values. RESULTS [(18)F]FPOIBG was synthesized from the corresponding tosylate precursor in 5.2 ± 0.5% (n = 6) overall radiochemical yields starting with aqueous fluoride in about 105 min. In a paired-label in vitro assay, the uptake of [(18)F]FPOIBG at 2h was 10.2 ± 1.5%, 39.6 ± 13.4%, and 13.3 ± 2.5%, in NET-expressing SK-N-SH, UVW-NAT, and SK-N-BE(2c) cells, respectively, while these values for [(125)I]MIBG were 57.3 ± 8.1%, 82.7 ± 8.9%, and 66.3 ± 3.6%. The specificity of uptake of both tracers was demonstrated by blocking with desipramine. The (125)I-labeled congener of FPOIBG gave similar results. On the other hand, [(18)F]FPOBBG, a compound recently reported in the literature, demonstrated much higher uptake, albeit less than that of co-incubated [(125)I]MIBG. IC50 values for FPOIBG were higher than those obtained for MIBG and FPOBBG. Unlike the case with [(18)F]FPOBBG, the heart uptake [(18)F]FPOIBG in normal mice was significantly lower than that of MIBG. CONCLUSION Although [(18)F]FPOIBG does not appear to warrant further consideration as an (18)F-labeled MIBG analogue, analogues wherein the iodine in it is replaced with a chlorine, fluorine or hydrogen might be worth pursuing. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE An (18)F-labeled analogue of the well-known radiopharmaceutical MIBG could have significant impact, potentially improving imaging of NET related disease in cardiology and in the imaging of neuroendocrine tumors. Although (18)F-labeled analogues of MIBG have been reported including LMI1195, we undertook this work hypothesizing that based on its greater structural similarity to MIBG, FPOIBG might be a better analogue than LMI1195.
Collapse
Affiliation(s)
- Ganesan Vaidyanathan
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710.
| | - Darryl McDougald
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Eftychia Koumarianou
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Jaeyeon Choi
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Marc Hens
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
33
|
Moroz MA, Zhang H, Lee J, Moroz E, Zurita J, Shenker L, Serganova I, Blasberg R, Ponomarev V. Comparative Analysis of T Cell Imaging with Human Nuclear Reporter Genes. J Nucl Med 2015; 56:1055-60. [PMID: 26025962 DOI: 10.2967/jnumed.115.159855] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 04/25/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Monitoring genetically altered T cells is an important component of adoptive T cell therapy in patients, and the ability to visualize their trafficking/targeting, proliferation/expansion, and retention/death using highly sensitive reporter systems that do not induce an immunologic response would provide useful information. Therefore, we focused on human reporter gene systems that have the potential for translation to clinical studies. The objective of the in vivo imaging studies was to determine the minimum number of T cells that could be visualized with the different nuclear reporter systems. We determined the imaging sensitivity (lower limit of T cell detection) of each reporter using appropriate radiolabeled probes for PET or SPECT imaging. METHODS Human T cells were transduced with retroviral vectors encoding for the human norepinephrine transporter (hNET), human sodium-iodide symporter (hNIS), a human deoxycytidine kinase double mutant (hdCKDM), and herpes simplex virus type 1 thymidine kinase (hsvTK) reporter genes. After viability and growth were assessed, 10(5) to 3 × 10(6) reporter T cells were injected subcutaneously on the shoulder area. The corresponding radiolabeled probe was injected intravenously 30 min later, followed by sequential PET or SPECT imaging. Radioactivity at the T cell injection sites and in the thigh (background) was measured. RESULTS The viability and growth of experimental cells were unaffected by transduction. The hNET/meta-(18)F-fluorobenzylguanidine ((18)F-MFBG) reporter system could detect less than 1 × 10(5) T cells because of its high uptake in the transduced T cells and low background activity. The hNIS/(124)I-iodide reporter system could detect approximately 1 × 10(6) T cells; (124)I-iodide uptake at the T cell injection site was time-dependent and associated with high background. The hdCKDM/2'-(18)F-fluoro-5-ethyl-1-β-d-arabinofuranosyluracil ((18)F-FEAU) and hsvTK/(18)F-FEAU reporter systems detected approximately 3 × 10(5) T cells, respectively. (18)F-FEAU was a more efficient probe (higher uptake, lower background) than (124)I-1-(2-deoxy-2-fluoro-1-d-arabinofuranosyl)-5-iodouracil for both hdCKDM and hsvTK. CONCLUSION A comparison of different reporter gene-reporter probe systems for imaging of T cell number was performed, and the hNET/(18)F-MFBG PET reporter system was found to be the most sensitive and capable of detecting approximately 35-40 × 10(3) T cells at the site of T cell injection in the animal model.
Collapse
Affiliation(s)
- Maxim A Moroz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hanwen Zhang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason Lee
- Crump Institute for Molecular Imaging, University of California, Los Angeles, California
| | - Ekaterina Moroz
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Juan Zurita
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Larissa Shenker
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Inna Serganova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Ronald Blasberg
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York; and Sloan Kettering Institute Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York Sloan Kettering Institute Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
34
|
Badar A, Kiru L, Kalber TL, Jathoul A, Straathof K, Årstad E, Lythgoe MF, Pule M. Fluorescence-guided development of a tricistronic vector encoding bimodal optical and nuclear genetic reporters for in vivo cellular imaging. EJNMMI Res 2015; 5:18. [PMID: 25853023 PMCID: PMC4385325 DOI: 10.1186/s13550-015-0097-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/10/2015] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In vivo imaging using genetic reporters is a central supporting tool in the development of cell and gene therapies affording us the ability to selectively track the therapeutic indefinitely. Previous studies have demonstrated the utility of the human norepinephrine transporter (hNET) as a positron emission tomography/single photon emission computed tomography (PET/SPECT) genetic reporter for in vivo cellular imaging. Here, our aim was to extend on this work and construct a tricistronic vector with dual optical (firefly luciferase) and nuclear (hNET) in vivo imaging and ex vivo histochemical capabilities. Guiding this development, we describe how a fluorescent substrate for hNET, 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP(+)), can be used to optimise vector design and serve as an in vitro functional screen. METHODS Vectors were designed to co-express a bright red-shifted firefly luciferase (FLuc), hNET and a small marker gene RQR8. Genes were co-expressed using 2A peptide linkage, and vectors were transduced into a T cell line, SupT1. Two vectors were constructed with different gene orientations; FLuc.2A.RQR8.2A.hNET and hNET.2A.FLuc.2A.RQR8. hNET function was assessed using ASP(+)-guided flow cytometry. In vivo cellular conspicuity was confirmed using sequential bioluminescence imaging (BLI) and SPECT imaging of transduced SupT1 cells injected into the flanks of mice. RESULTS SupT1/FLuc.2A.RQR8.2A.hNET cells resulted in >4-fold higher ASP(+) uptake compared to SupT1/hNET.2A.FLuc.2A.RQR8, suggesting that 2A orientation effected hNET function. SupT1/FLuc.2A.RQR8.2A.hNET cells were readily visualised with both BLI and SPECT, demonstrating high signal to noise at 24 h post (123)I-meta-iodobenzylguanidine (MIBG) administration. CONCLUSIONS In this study, a pre-clinical tricistronic vector with flow cytometry, BLI, SPECT and histochemical capabilities was constructed, which can be widely applied in cell tracking studies supporting the development of cell therapies. The study further demonstrates that hNET function in engineered cells can be assessed using ASP(+)-guided flow cytometry in place of costly radiosubstrate methodologies. This fluorogenic approach is unique to the hNET PET/SPECT reporter and may prove valuable when screening large numbers of cell lines or vector/mutant constructs.
Collapse
Affiliation(s)
- Adam Badar
- />Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Louise Kiru
- />Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
- />UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Tammy L Kalber
- />Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Amit Jathoul
- />UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Karin Straathof
- />UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Erik Årstad
- />Department of Chemistry and Institute of Nuclear Medicine, University College London, 235 Euston Road (T-5), London, NW1 2BU UK
| | - Mark F Lythgoe
- />Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Martin Pule
- />UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| |
Collapse
|
35
|
Streby KA, Shah N, Ranalli MA, Kunkler A, Cripe TP. Nothing but NET: a review of norepinephrine transporter expression and efficacy of 131I-mIBG therapy. Pediatr Blood Cancer 2015; 62:5-11. [PMID: 25175627 PMCID: PMC4237663 DOI: 10.1002/pbc.25200] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/07/2014] [Indexed: 12/21/2022]
Abstract
Neuroblastoma is unique amongst common pediatric cancers for its expression of the norepinephrine transporter (NET), enabling tumor-selective imaging and therapy with radioactive analogues of norepinephrine. The majority of neuroblastoma tumors are avid for (123)I-metaiodobenzaguanidine (mIBG) on imaging, yet the therapeutic response to (131) I-mIBG is only 30% in clinical trials, and off-target effects cause short- and long-term morbidity. We review the contemporary understanding of the tumor-selective uptake, retention, and efflux of meta-iodobenzylguanidine (mIBG) and strategies currently in development for improving its efficacy. Combination treatment strategies aimed at enhancing NET are likely necessary to reach the full potential of (131)I-mIBG therapy.
Collapse
Affiliation(s)
- Keri A Streby
- Division of Hematology/Oncology/Blood and Marrow Transplant, The Ohio State UniversityColumbus, Ohio
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State UniversityColumbus, Ohio
| | - Nilay Shah
- Division of Hematology/Oncology/Blood and Marrow Transplant, The Ohio State UniversityColumbus, Ohio
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State UniversityColumbus, Ohio
| | - Mark A Ranalli
- Division of Hematology/Oncology/Blood and Marrow Transplant, The Ohio State UniversityColumbus, Ohio
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State UniversityColumbus, Ohio
| | - Anne Kunkler
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State UniversityColumbus, Ohio
| | - Timothy P Cripe
- Division of Hematology/Oncology/Blood and Marrow Transplant, The Ohio State UniversityColumbus, Ohio
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State UniversityColumbus, Ohio
| |
Collapse
|
36
|
|
37
|
Bernsen MR, Vaissier PEB, Van Holen R, Booij J, Beekman FJ, de Jong M. The role of preclinical SPECT in oncological and neurological research in combination with either CT or MRI. Eur J Nucl Med Mol Imaging 2014; 41 Suppl 1:S36-49. [PMID: 24895751 PMCID: PMC4003405 DOI: 10.1007/s00259-013-2685-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 12/20/2013] [Indexed: 01/03/2023]
Abstract
Preclinical imaging with SPECT combined with CT or MRI is used more and more frequently and has proven to be very useful in translational research. In this article, an overview of current preclinical research applications and trends of SPECT combined with CT or MRI, mainly in tumour imaging and neuroscience imaging, is given and the advantages and disadvantages of the different approaches are described. Today SPECT and CT systems are often integrated into a single device (commonly called a SPECT/CT system), whereas at present combined SPECT and MRI is almost always carried out with separate systems and fiducial markers to combine the separately acquired images. While preclinical SPECT/CT is most widely applied in oncology research, SPECT combined with MRI (SPECT/MRI when integrated in one system) offers the potential for both neuroscience applications and oncological applications. Today CT and MRI are still mainly used to localize radiotracer binding and to improve SPECT quantification, although both CT and MRI have additional potential. Future technology developments may include fast sequential or simultaneous acquisition of (dynamic) multimodality data, spectroscopy, fMRI along with high-resolution anatomic MRI, advanced CT procedures, and combinations of more than two modalities such as combinations of SPECT, PET, MRI and CT all together. This will all strongly depend on new technologies. With further advances in biology and chemistry for imaging molecular targets and (patho)physiological processes in vivo, the introduction of new imaging procedures and promising new radiopharmaceuticals in clinical practice may be accelerated.
Collapse
Affiliation(s)
- Monique R. Bernsen
- Department of Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology, Erasmus MC, Rotterdam, The Netherlands
| | - Pieter E. B. Vaissier
- Section Radiation Detection and Medical Imaging, Delft University of Technology, Delft, The Netherlands
| | - Roel Van Holen
- ELIS Department, MEDISIP, Ghent University, iMinds, Ghent, Belgium
| | - Jan Booij
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Freek J. Beekman
- Section Radiation Detection and Medical Imaging, Delft University of Technology, Delft, The Netherlands
- MILabs B.V., Utrecht, The Netherlands
| | - Marion de Jong
- Department of Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
38
|
Matthews PM, Comley R. Advances in the molecular imaging of multiple sclerosis. Expert Rev Clin Immunol 2014; 5:765-77. [DOI: 10.1586/eci.09.66] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
39
|
Narunsky L, Oren R, Bochner F, Neeman M. Imaging aspects of the tumor stroma with therapeutic implications. Pharmacol Ther 2013; 141:192-208. [PMID: 24134903 DOI: 10.1016/j.pharmthera.2013.10.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 09/13/2013] [Indexed: 12/25/2022]
Abstract
Cancer cells rely on extensive support from the stroma in order to survive, proliferate and invade. The tumor stroma is thus an important potential target for anti-cancer therapy. Typical changes in the stroma include a shift from the quiescence promoting-antiangiogenic extracellular matrix to a provisional matrix that promotes invasion and angiogenesis. These changes in the extracellular matrix are induced by changes in the secretion of extracellular matrix proteins and glucose amino glycans, extravasation of plasma proteins from hyperpermeable vessels and release of matrix modifying enzymes resulting in cleavage and cross-linking of matrix macromolecules. These in turn alter the rigidity of the matrix and the exposure and release of cytokines. Changes in matrix rigidity and vessel permeability affect drug delivery and mediate resistance to cytotoxic therapy. These stroma changes are brought about not only by the cancer cells, but also through the action of many cell types that are recruited by tumors including immune cells, fibroblasts and endothelial cells. Within the tumor, these normal host cells are activated resulting in loss of inhibitory and induction of cancer promoting activities. Key to the development of stroma-targeted therapies, selective biomarkers were developed for specific imaging of key aspects of the tumor stroma.
Collapse
Affiliation(s)
- Lian Narunsky
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Roni Oren
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Filip Bochner
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Michal Neeman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
40
|
Leech JM, Sharif-Paghaleh E, Maher J, Livieratos L, Lechler RI, Mullen GE, Lombardi G, Smyth LA. Whole-body imaging of adoptively transferred T cells using magnetic resonance imaging, single photon emission computed tomography and positron emission tomography techniques, with a focus on regulatory T cells. Clin Exp Immunol 2013; 172:169-77. [PMID: 23574314 DOI: 10.1111/cei.12087] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2013] [Indexed: 01/03/2023] Open
Abstract
Cell-based therapies using natural or genetically modified regulatory T cells (T(regs)) have shown significant promise as immune-based therapies. One of the main difficulties facing the further advancement of these therapies is that the fate and localization of adoptively transferred T(regs) is largely unknown. The ability to dissect the migratory pathway of these cells in a non-invasive manner is of vital importance for the further development of in-vivo cell-based immunotherapies, as this technology allows the fate of the therapeutically administered cell to be imaged in real time. In this review we will provide an overview of the current clinical imaging techniques used to track T cells and T(regs) in vivo, including magnetic resonance imaging (MRI) and positron emission tomography (PET)/single photon emission computed tomography (SPECT). In addition, we will discuss how the finding of these studies can be used, in the context of transplantation, to define the most appropriate T(reg) subset required for cellular therapy.
Collapse
Affiliation(s)
- J M Leech
- Medical Research Council, Centre for Transplantation, King's College London, King's Health Partners, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Li K, Qin W, Ding D, Tomczak N, Geng J, Liu R, Liu J, Zhang X, Liu H, Liu B, Tang BZ. Photostable fluorescent organic dots with aggregation-induced emission (AIE dots) for noninvasive long-term cell tracing. Sci Rep 2013; 3:1150. [PMID: 23359649 PMCID: PMC3556677 DOI: 10.1038/srep01150] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 12/11/2012] [Indexed: 12/24/2022] Open
Abstract
Long-term noninvasive cell tracing by fluorescent probes is of great importance to life science and biomedical engineering. For example, understanding genesis, development, invasion and metastasis of cancerous cells and monitoring tissue regeneration after stem cell transplantation require continual tracing of the biological processes by cytocompatible fluorescent probes over a long period of time. In this work, we successfully developed organic far-red/near-infrared dots with aggregation-induced emission (AIE dots) and demonstrated their utilities as long-term cell trackers. The high emission efficiency, large absorptivity, excellent biocompatibility, and strong photobleaching resistance of the AIE dots functionalized by cell penetrating peptides derived from transactivator of transcription proteins ensured outstanding long-term noninvasive in vitro and in vivo cell tracing. The organic AIE dots outperform their counterparts of inorganic quantum dots, opening a new avenue in the development of fluorescent probes for following biological processes such as carcinogenesis.
Collapse
Affiliation(s)
- Kai Li
- Institute of Materials Research and Engineering, 3, Research Link, Singapore 117602
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117576
- These authors contributed equally to this work
| | - Wei Qin
- Department of Chemistry, Division of Biomedical Engineering, Institute for Advanced Study, State Key Laboratory of Molecular Neuroscience, and Institute of Molecular Functional Materials, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, China
- These authors contributed equally to this work
| | - Dan Ding
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117576
| | - Nikodem Tomczak
- Institute of Materials Research and Engineering, 3, Research Link, Singapore 117602
| | - Junlong Geng
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117576
| | - Rongrong Liu
- Institute of Materials Research and Engineering, 3, Research Link, Singapore 117602
| | - Jianzhao Liu
- Department of Chemistry, Division of Biomedical Engineering, Institute for Advanced Study, State Key Laboratory of Molecular Neuroscience, and Institute of Molecular Functional Materials, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xinhai Zhang
- Institute of Materials Research and Engineering, 3, Research Link, Singapore 117602
| | - Hongwei Liu
- Institute of Materials Research and Engineering, 3, Research Link, Singapore 117602
| | - Bin Liu
- Institute of Materials Research and Engineering, 3, Research Link, Singapore 117602
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117576
| | - Ben Zhong Tang
- Institute of Materials Research and Engineering, 3, Research Link, Singapore 117602
- Department of Chemistry, Division of Biomedical Engineering, Institute for Advanced Study, State Key Laboratory of Molecular Neuroscience, and Institute of Molecular Functional Materials, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, China
- SCUT-HKUST Joint Research Laboratory, Guangdong Innovative Research Team, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, China, 510640
| |
Collapse
|
42
|
Maher J. Immunotherapy of malignant disease using chimeric antigen receptor engrafted T cells. ISRN ONCOLOGY 2012; 2012:278093. [PMID: 23304553 PMCID: PMC3523553 DOI: 10.5402/2012/278093] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 11/14/2012] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor- (CAR-) based immunotherapy has been under development for almost 25 years, over which period it has progressed from a new but cumbersome technology to an emerging therapeutic modality for malignant disease. The approach involves the genetic engineering of fusion receptors (CARs) that couple the HLA-independent binding of cell surface target molecules to the delivery of a tailored activating signal to host immune cells. Engineered CARs are delivered most commonly to peripheral blood T cells using a range of vector systems, most commonly integrating viral vectors. Preclinical refinement of this approach has proceeded over several years to the point that clinical testing is now being undertaken at several centres, using increasingly sophisticated and therapeutically successful genetic payloads. This paper considers several aspects of the pre-clinical and clinical development of CAR-based immunotherapy and how this technology is acquiring an increasing niche in the treatment of both solid and haematological malignancies.
Collapse
Affiliation(s)
- John Maher
- CAR Mechanics Group, Department of Research Oncology, King's Health Partners Integrated Cancer Centre, King's College London, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK
- Department of Immunology, Barnet and Chase Farm Hospitals NHS Trust, Barnet, Hertfordshire EN5 3DJ, UK
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| |
Collapse
|
43
|
Evaluation of Norepinephrine Transporter Expression and Metaiodobenzylguanidine Avidity in Neuroblastoma: A Report from the Children's Oncology Group. INTERNATIONAL JOURNAL OF MOLECULAR IMAGING 2012; 2012:250834. [PMID: 23050139 PMCID: PMC3463166 DOI: 10.1155/2012/250834] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 08/23/2012] [Indexed: 11/18/2022]
Abstract
Purpose. (123)I-metaiodobenzylguanidine (MIBG) is used for the diagnostic evaluation of neuroblastoma. We evaluated the relationship between norepinephrine transporter (NET) expression and clinical MIBG uptake. Methods. Quantitative reverse transcription PCR (N = 82) and immunohistochemistry (IHC; N = 61) were performed for neuroblastoma NET mRNA and protein expression and correlated with MIBG avidity on diagnostic scans. The correlation of NET expression with clinical features was also performed. Results. Median NET mRNA expression level for the 19 MIBG avid patients was 12.9% (range 1.6-73.7%) versus 5.9% (range 0.6-110.0%) for the 8 nonavid patients (P = 0.31). Median percent NET protein expression was 50% (range 0-100%) in MIBG avid patients compared to 10% (range 0-80%) in nonavid patients (P = 0.027). MYCN amplified tumors had lower NET protein expression compared to nonamplified tumors (10% versus 50%; P = 0.0002). Conclusions. NET protein expression in neuroblastoma correlates with MIBG avidity. MYCN amplified tumors have lower NET protein expression.
Collapse
|
44
|
Abstract
During the past two decades, stem cells have created enthusiasm as a regenerative therapy for ischemic heart disease. Transplantation of bone marrow stem cells, skeletal myoblasts, and endothelial progenitor cells has shown to improve myocardial function after infarction. Recently, attention has focused on the potential use of embryonic stem cells and induced pluripotent stem cells because they possess the capacity to differentiate into various cell types, including cardiac and endothelial cells. Clinical trials have shown positive effects on the functional recovery of heart after myocardial infarction and have answered questions on timing, dosage, and cell delivery route of stem cells such as those derived from bone marrow. Despite the current advances in stem cell research, one main hurdle remains the lack of reliable information about the fate of cell engraftment, survival, and proliferation after transplantation. This review discusses the different cell types used in cardiac cell therapy as well as molecular imaging modalities relevant to survival issues.
Collapse
|
45
|
Yaghoubi SS, Campbell DO, Radu CG, Czernin J. Positron emission tomography reporter genes and reporter probes: gene and cell therapy applications. Am J Cancer Res 2012; 2:374-91. [PMID: 22509201 PMCID: PMC3326723 DOI: 10.7150/thno.3677] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 02/09/2012] [Indexed: 12/22/2022] Open
Abstract
Positron emission tomography (PET) imaging reporter genes (IRGs) and PET reporter probes (PRPs) are amongst the most valuable tools for gene and cell therapy. PET IRGs/PRPs can be used to non-invasively monitor all aspects of the kinetics of therapeutic transgenes and cells in all types of living mammals. This technology is generalizable and can allow long-term kinetics monitoring. In gene therapy, PET IRGs/PRPs can be used for whole-body imaging of therapeutic transgene expression, monitoring variations in the magnitude of transgene expression over time. In cell or cellular gene therapy, PET IRGs/PRPs can be used for whole-body monitoring of therapeutic cell locations, quantity at all locations, survival and proliferation over time and also possibly changes in characteristics or function over time. In this review, we have classified PET IRGs/PRPs into two groups based on the source from which they were derived: human or non-human. This classification addresses the important concern of potential immunogenicity in humans, which is important for expansion of PET IRG imaging in clinical trials. We have then discussed the application of this technology in gene/cell therapy and described its use in these fields, including a summary of using PET IRGs/PRPs in gene and cell therapy clinical trials. This review concludes with a discussion of the future direction of PET IRGs/PRPs and recommends cell and gene therapists collaborate with molecular imaging experts early in their investigations to choose a PET IRG/PRP system suitable for progression into clinical trials.
Collapse
|
46
|
Abstract
Cell-based therapies, such as adoptive immunotherapy and stem-cell therapy, have received considerable attention as novel therapeutics in oncological research and clinical practice. The development of effective therapeutic strategies using tumor-targeted cells requires the ability to determine in vivo the location, distribution, and long-term viability of the therapeutic cell populations as well as their biological fate with respect to cell activation and differentiation. In conjunction with various noninvasive imaging modalities, cell-labeling methods, such as exogenous labeling or transfection with a reporter gene, allow visualization of labeled cells in vivo in real time, as well as monitoring and quantifying cell accumulation and function. Such cell-tracking methods also have an important role in basic cancer research, where they serve to elucidate novel biological mechanisms. In this Review, we describe the basic principles of cell-tracking methods, explain various approaches to cell tracking, and highlight recent examples for the application of such methods in animals and humans.
Collapse
|
47
|
Abstract
Purpose The objective of this article is to develop internalizing positron emission tomography (PET) reporter genes for tracking genetically modified T cells in vivo. Procedures The transmembrane and cytoplasmic domains of the human transferrin receptor (TfR) and CD5 were each fused to the carcinoembryonic (CEA) minigene N-A3 and expressed in Jurkat T cells. Internalization was evaluated by confocal microscopy or by intracellular uptake of 125I-labeled anti-CEA scFv-Fc. Reporter gene-transfected Jurkat xenografts in mice were analyzed by immunohistochemistry (IHC) and imaged by PET using 124I- or 64Cu-scFv-Fc as tracers. Results Surface expression of TR(1–99)-NA3 was lower than that of NA3-CD5. Both reporter genes were internalized following binding of the anti-CEA antibody fragment. IHC of tumors showed strong staining of NA3-CD5, whereas TR(1–99)-NA3 stained weakly. Specific targeting of TR(1–99)-NA3 or NA3-CD5 was shown by PET in xenografted mice. Conclusions The in vivo imaging studies suggest a potential application of the internalizing form of CEA (N-A3) as a PET reporter gene.
Collapse
|
48
|
Cheng D, Wang Y, Liu X, Pretorius PH, Liang M, Rusckowski M, Hnatowich DJ. Comparison of 18F PET and 99mTc SPECT imaging in phantoms and in tumored mice. Bioconjug Chem 2011; 21:1565-70. [PMID: 20681508 DOI: 10.1021/bc1001467] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Our objective was to compare the performance of a micro-single photon emission computed tomography (micro-SPECT) with that of a micro-positron emission tomography (microPET) in a Her2+ tumored mice using an anti-Her2 nanoparticle radiolabeled with (99m)Tc and (18)F. Camera performance was first compared using phantoms; then a tumored mouse administered the (99m)Tc-nanoparticle was imaged on a Bioscan NanoSPECT/CT, while another tumored mouse received the identical nanoparticle, labeled now with (18)F, and was imaged on a Philips Mosaic HP PET camera. The nanoparticle was radiolabeled with (99m)Tc via MAG(3) chelation and with (18)F via SFB as an intermediate. Phantom imaging showed that the resolution of the SPECT camera was clearly superior, but even with 4 heads and multipinhole collimators, detection sensitivity was 15-fold lower. Radiolabeling of the nanoparticle by chelation with (99m)Tc was considerably easier and safer than manual covalent attachment of (18)F. Both cameras provided accurate quantitation of radioactivity over a broad range. In conclusion, when deciding between (99m)Tc vs (18)F, an advantage rests with the chelation of (99m)Tc over covalent attachment of (18)F, achieved manually or otherwise, but with these small animal cameras, this choice also results in trading lower sensitivity for higher resolution.
Collapse
Affiliation(s)
- Dengfeng Cheng
- Division of Nuclear Medicine, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Ottobrini L, Martelli C, Trabattoni DL, Clerici M, Lucignani G. In vivo imaging of immune cell trafficking in cancer. Eur J Nucl Med Mol Imaging 2010; 38:949-68. [PMID: 21170525 DOI: 10.1007/s00259-010-1687-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 11/15/2010] [Indexed: 12/14/2022]
Abstract
Tumour establishment, progression and regression can be studied in vivo using an array of imaging techniques ranging from MRI to nuclear-based and optical techniques that highlight the intrinsic behaviour of different cell populations in the physiological context. Clinical in vivo imaging techniques and preclinical specific approaches have been used to study, both at the macroscopic and microscopic level, tumour cells, their proliferation, metastasisation, death and interaction with the environment and with the immune system. Fluorescent, radioactive or paramagnetic markers were used in direct protocols to label the specific cell population and reporter genes were used for genetic, indirect labelling protocols to track the fate of a given cell subpopulation in vivo. Different protocols have been proposed to in vivo study the interaction between immune cells and tumours by different imaging techniques (intravital and whole-body imaging). In particular in this review we report several examples dealing with dendritic cells, T lymphocytes and macrophages specifically labelled for different imaging procedures both for the study of their physiological function and in the context of anti-neoplastic immunotherapies in the attempt to exploit imaging-derived information to improve and optimise anti-neoplastic immune-based treatments.
Collapse
Affiliation(s)
- Luisa Ottobrini
- Department of Biomedical Sciences and Technologies, University of Milan, Milan, Italy
| | | | | | | | | |
Collapse
|
50
|
Vilekar P, Awasthi V, Lagisetty P, King C, Shankar N, Awasthi S. In vivo trafficking and immunostimulatory potential of an intranasally-administered primary dendritic cell-based vaccine. BMC Immunol 2010; 11:60. [PMID: 21143974 PMCID: PMC3018378 DOI: 10.1186/1471-2172-11-60] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 12/10/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Coccidioidomycosis or Valley fever is caused by a highly virulent fungal pathogen: Coccidioides posadasii or immitis. Vaccine development against Coccidioides is of contemporary interest because a large number of relapses and clinical failures are reported with antifungal agents. An efficient Th1 response engenders protection. Thus, we have focused on developing a dendritic cell (DC)-based vaccine for coccidioidomycosis. In this study, we investigated the immunostimulatory characteristics of an intranasal primary DC-vaccine in BALB/c mouse strain that is most susceptible to coccidioidomycosis. The DCs were transfected nonvirally with Coccidioides-Ag2/PRA-cDNA. Expression of DC-markers, Ag2/PRA and cytokines were studied by flow cytometry, dot-immunoblotting and cytometric bead array methods, respectively. The T cell activation was studied by assessing the upregulation of activation markers in a DC-T cell co-culture assay. For trafficking, the DCs were co-transfected with a plasmid DNA encoding HSV1 thymidine kinase (TK) and administered intranasally into syngeneic mice. The trafficking and homing of TK-expressing DCs were monitored with positron emission tomography (PET) using 18F-FIAU probe. Based on the PET-probe accumulation in vaccinated mice, selected tissues were studied for antigen-specific response and T cell phenotypes using ELISPOT and flow cytometry, respectively. RESULTS We found that the primary DCs transfected with Coccidioides-Ag2/PRA-cDNA were of immature immunophenotype, expressed Ag2/PRA and activated naïve T cells. In PET images and subsequent biodistribution, intranasally-administered DCs were found to migrate in blood, lung and thymus; lymphocytes showed generation of T effector memory cell population (T(EM)) and IFN-γ release. CONCLUSIONS In conclusion, our results demonstrate that the intranasally-administered primary DC vaccine is capable of inducing Ag2/PRA-specific T cell response. Unique approaches utilized in our study represent an attractive and novel means of producing and evaluating an autologous DC-based vaccine.
Collapse
Affiliation(s)
- Prachi Vilekar
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, OK 73117, USA
| | | | | | | | | | | |
Collapse
|