1
|
Ghosh P, Davies LJ, Nitsche C. Engineered Nanobodies Bind Bismuth, Indium and Gallium for Applications in Theranostics. Angew Chem Int Ed Engl 2025; 64:e202419455. [PMID: 39481115 DOI: 10.1002/anie.202419455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/02/2024]
Abstract
Targeted theranostics heavily rely on metal isotopes conjugated to antibodies. Single-domain antibodies, known as nanobodies, are much smaller in size without compromising specificity and affinity. The conventional way of conjugating metals to nanobodies involves non-specific modification of amino acid residues with bifunctional chelating agents. We demonstrate that mutagenesis of a single residue in a nanobody creates a triple cysteine motif that selectively binds bismuth which is, for example, used in targeted alpha therapy. Two mutations create a quadruple cysteine mutant specific for gallium and indium used in positron emission tomography and single-photon emission computed tomography, respectively. Labelling is quantitative within a few minutes. The metal nanobodies maintain structural integrity and stability over weeks, resist competition from endogenous metal binders like glutathione, and retain functionality.
Collapse
Affiliation(s)
- Pritha Ghosh
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Lani J Davies
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Christoph Nitsche
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
2
|
Spiegelberg D, Hwang LA, Pua KH, Kumar SC, Koh XY, Koh XH, Selvaraju RK, Sabapathy K, Nestor M, Lane D. Targeting mutant p53: Evaluation of novel anti-p53 R175H monoclonal antibodies as diagnostic tools. Sci Rep 2025; 15:1000. [PMID: 39762369 PMCID: PMC11704002 DOI: 10.1038/s41598-024-83871-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
About 50% of all cancers carry a mutation in p53 that impairs its tumor suppressor function. The p53 missense mutation p53R175H (p53R172H in mice) is a hotspot mutation in various cancer types. Therefore, monoclonal antibodies selectively targeting clinically relevant mutations like p53R175H could prove immensely value. We aimed to evaluate the in vitro and in vivo binding properties of two novel anti-p53R175H monoclonal antibodies and to assess their performance as agents for molecular imaging. In vitro, 125I-4H5 and 125I-7B9 demonstrated long shelf life and antigen-specific binding. Our in vivo study design allowed head-to-head comparison of the antibodies in a double tumor model using repeated SPECT/CT imaging, followed by biodistribution and autoradiography. Both tracers performed similarly, with marginally faster blood clearance for 125I-7B9. Repeated molecular imaging demonstrated suitable imaging characteristics for both antibodies, with the best contrast images occurring at 48 h post-injection. Significantly higher uptake was detected in the mut-p53-expressing tumors, confirmed by ex vivo autoradiography. We conclude that molecular imaging with an anti-p53R175H tracer could be a promising approach for cancer diagnostics and could be further applied for patient stratification and treatment response monitoring of mutant p53-targeted therapeutics.
Collapse
Affiliation(s)
- Diana Spiegelberg
- Department of Immunology, Genetics, Pathology, Uppsala University, Uppsala, Sweden.
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Le-Ann Hwang
- Divisions of Cellular & Molecular Research, National Cancer Centre Singapore, Singapore, 168583, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Khian Hong Pua
- Institute of Molecular and Cellular Biology, ASTAR, Singapore, 138673, Singapore
| | - Sashwini Chandra Kumar
- Divisions of Cellular & Molecular Research, National Cancer Centre Singapore, Singapore, 168583, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Xin Yu Koh
- Institute of Molecular and Cellular Biology, ASTAR, Singapore, 138673, Singapore
| | - Xiao Hui Koh
- Institute of Molecular and Cellular Biology, ASTAR, Singapore, 138673, Singapore
| | - Ram Kumar Selvaraju
- Preclinical PET-MRI Platform, Part of Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Kanaga Sabapathy
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Marika Nestor
- Department of Immunology, Genetics, Pathology, Uppsala University, Uppsala, Sweden
| | - David Lane
- Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Movahed F, Navaei O, Taghlidi S, Nurzadeh M, Gharaati ME, Rabiei M. Radiolabeled HER2-targeted molecular probes in breast cancer imaging: current knowledge and future prospective. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03691-7. [PMID: 39751821 DOI: 10.1007/s00210-024-03691-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025]
Abstract
Breast cancer is the most frequent non-dermatologic malignancy in women. Breast cancer is characterized by the expression of the human epidermal growth factor receptor type 2 (HER2), and the presence or lack of estrogen receptor (ER) and progesterone receptor (PR) expression. HER2 overexpression is reported in about 20 to 25% of breast cancer patients, which is usually linked to cancer progression, metastases, and poor survival. Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) are the gold standards for determining HER2 status, even though IHC has largely focused on quantifying HER2+ status versus "other" HER2 status (including variants with low or no expression). Recent findings regarding the beneficial therapeutic effects of anti-HER2 monoclonal antibodies (mAb) in HER2low metastatic patients lead to changes in the classic definition of advanced breast cancer, and methods for precise assessment of HER2 status are being developed. As a result, various radiolabeled HER-targeted mAbs and antibody fragments have been designed to avoid repeated biopsies with potential bias due to tumor heterogeneity, including single-chain variable fragment (scFv), F(ab')2, affibody, and nanobody. These small targeting radiotracers displayed favorable biodistributions, clearance, and stability, allowing for higher image quality, shorter circulation half-life, and lower immunogenicity. This study aimed to comprehensively review the application of radiolabeled anti-HER2 antibody fragments in breast cancer in vivo imaging and provide a better understanding of targeted HER2 quantification.
Collapse
Affiliation(s)
- Fatemeh Movahed
- Department of Gynecology and Obstetrics, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ouldouz Navaei
- Department of Biotechnology, Università Milano-Bicocca Milano, Milan, Italy
| | - Shiva Taghlidi
- Medicine and Surgery, Università Degli Studi Di Milano-Bicocca, Milan, Italy
| | - Maryam Nurzadeh
- Department of Fetomaternal, Faculty of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Eslami Gharaati
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Rabiei
- Obstetrics and Gynecology Department, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Dev ID, Puranik AD, Singh B, Prasad V. Current and Future Perspectives of PDL1 PET and SPECT Imaging. Semin Nucl Med 2024; 54:966-975. [PMID: 39510854 DOI: 10.1053/j.semnuclmed.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 11/15/2024]
Abstract
Programmed Death 1 (PD1) and Programmed Death Ligand (PDL1) play a crucial role in tumor microenvironment by helping cancer cells evade innate immunity. Numerous inhibitor anticancer drugs targeting this interplay have been used in clinical practice and many more are in preclinical stage. These drugs have shown promising results in achieving good response and long-term clinical benefit, is routinely performed to identify patients who may benefit. However, there are major challenges associated with these immunohistochemistry tests which have opened the space for noninvasive imaging modalities using PD1 and PDL1 inhibitors labeled with either PET or SPECT radionuclides. These radiopharmaceuticals, although primarily developed for the field of immunotherapy, have great potential in expanding and optimizing the combination of radiopharmaceutical therapies with PD1-PDL1 targeting anticancer drugs. This review elaborates currently available PET and SPECT radiopharmaceuticals targeting PD1-PDL1 axis. It also explores the potential future role of newer targets which are being developed and tested in various preclinical studies.
Collapse
Affiliation(s)
- Indraja D Dev
- Assistant Professor, Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Center, Homi Bhabha National Institute, Mumbai, India
| | - Ameya D Puranik
- Professor, Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, India
| | - Baljinder Singh
- Professor, Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Vikas Prasad
- Director of Clinical Theranostics, Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington, WA.
| |
Collapse
|
5
|
Riessland M, Ximerakis M, Jarjour AA, Zhang B, Orr ME. Therapeutic targeting of senescent cells in the CNS. Nat Rev Drug Discov 2024; 23:817-837. [PMID: 39349637 DOI: 10.1038/s41573-024-01033-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 11/01/2024]
Abstract
Senescent cells accumulate throughout the body with advanced age, diseases and chronic conditions. They negatively impact health and function of multiple systems, including the central nervous system (CNS). Therapies that target senescent cells, broadly referred to as senotherapeutics, recently emerged as potentially important treatment strategies for the CNS. Promising therapeutic approaches involve clearing senescent cells by disarming their pro-survival pathways with 'senolytics'; or dampening their toxic senescence-associated secretory phenotype (SASP) using 'senomorphics'. Following the pioneering discovery of first-generation senolytics dasatinib and quercetin, dozens of additional therapies have been identified, and several promising targets are under investigation. Although potentially transformative, senotherapies are still in early stages and require thorough testing to ensure reliable target engagement, specificity, safety and efficacy. The limited brain penetrance and potential toxic side effects of CNS-acting senotherapeutics pose challenges for drug development and translation to the clinic. This Review assesses the potential impact of senotherapeutics for neurological conditions by summarizing preclinical evidence, innovative methods for target and biomarker identification, academic and industry drug development pipelines and progress in clinical trials.
Collapse
Affiliation(s)
- Markus Riessland
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| | | | | | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miranda E Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
- Salisbury VA Medical Center, Salisbury, NC, USA.
| |
Collapse
|
6
|
Northington KR, Calderon J, Bates EA. Netrin-1 stimulated axon growth requires the polyglutamylase TTLL1. Front Neurosci 2024; 18:1436312. [PMID: 39469034 PMCID: PMC11514365 DOI: 10.3389/fnins.2024.1436312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction In the developing brain, neurons extend an axonal process through a complex and changing environment to form synaptic connections with the correct targets in response to extracellular cues. Microtubule and actin filaments provide mechanical support and drive axon growth in the correct direction. The axonal cytoskeleton responds to extracellular guidance cues. Netrin-1 is a multifunctional guidance cue that can induce alternate responses based on the bound receptor. The mechanism by which actin responds to Netrin-1 is well described. However, how Netrin-1 influences the microtubule cytoskeleton is less understood. Appropriate microtubule function is required for axon pathfinding, as mutations in tubulin phenocopy axon crossing defects of Netrin-1 and DCC mutants. Microtubule stabilization is required for attractive guidance cue response. The C-terminal tails of microtubules can be post-translationally modified. Post-translational modifications (PTMs) help control the microtubule cytoskeleton. Methods We measured polyglutamylation in cultured primary mouse cortical neurons before and after Netrin-1 stimulation. We used immunohistochemistry to measure how Netrin-1 stimulation alters microtubule-associated protein localization. Next, we manipulated TTLL1 to determine if Netrin-1-induced axon growth and MAP localization depend on polyglutamylation levels. Results In this study, we investigated if Netrin-1 signaling alters microtubule PTMs in the axon. We found that microtubule polyglutamylation increases after Netrin-1 stimulation. This change in polyglutamylation is necessary for Netrin-1-induced axonal growth rate increases. We next determined that MAP1B and DCX localization changes in response to Netrin-1. These proteins can both stabilize the microtubule cytoskeleton and may be responsible for Netrin-1-induced growth response in neurons. The changes in DCX and MAP1B depend on TTLL1, a protein responsible for microtubule polyglutamylation.
Collapse
Affiliation(s)
| | | | - Emily A. Bates
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
7
|
Neilio JM, Ginat DT. Emerging Head and Neck Tumor Targeting Contrast Agents for the Purpose of CT, MRI, and Multimodal Diagnostic Imaging: A Molecular Review. Diagnostics (Basel) 2024; 14:1666. [PMID: 39125542 PMCID: PMC11311342 DOI: 10.3390/diagnostics14151666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The diagnosis and treatment of head and neck tumors present significant challenges due to their infiltrative nature and diagnostic hindrances such as the blood-brain barrier. The intricate anatomy of the head and neck region also complicates the clear identification of tumor boundaries and assessment of tumor characteristics. AIM This review aims to explore the efficacy of molecular imaging techniques that employ targeted contrast agents in head and neck cancer imaging. Head and neck cancer imaging benefits significantly from the combined advantages of CT and MRI. CT excels in providing swift, high-contrast images, enabling the accurate localization of tumors, while MRI offers superior soft tissue resolution, contributing to the detailed evaluation of tumor morphology in this region of the body. Many of these novel contrast agents have integration of dual-modal, triple-modal, or even dual-tissue targeting imaging, which have expanded the horizons of molecular imaging. Emerging contrast agents for the purpose of MRI and CT also include the widely used standards in imaging such as gadolinium and iodine-based agents, respectively, but with peptide, polypeptide, or polymeric functionalizations. Relevance for patients. For patients, the development and use of these targeted contrast agents have potentially significant implications. They benefit from the enhanced accuracy of tumor detection and characterization, which are critical for effective treatment planning. Additionally, these agents offer improved imaging contrast with the added benefit of reduced toxicity and bioaccumulation. The summarization of preclinical nanoparticle research in this review serves as a valuable resource for scientists and students working towards advancing tumor diagnosis and treatment with targeted contrast agents.
Collapse
Affiliation(s)
- Jonathan M. Neilio
- Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA;
| | - Daniel T. Ginat
- Department of Radiology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
8
|
Badier L, Quelven I. Zirconium 89 and Copper 64 for ImmunoPET: From Antibody Bioconjugation and Radiolabeling to Molecular Imaging. Pharmaceutics 2024; 16:882. [PMID: 39065579 PMCID: PMC11279968 DOI: 10.3390/pharmaceutics16070882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Immunotherapy has transformed cancer treatment. Nevertheless, given the heterogeneity of clinical efficacy, the multiplicity of treatment options available and the possibility of serious adverse effects, selecting the most effective treatment has become the greatest challenge. Molecular imaging offers an attractive way for this purpose. ImmunoPET provides specific imaging with positron emission tomography (PET) using monoclonal antibodies (mAb) or its fragments as vector. By combining the high targeting specificity of mAb and the sensitivity of PET technique, immunoPET could noninvasively and dynamically reveal tumor antigens expression and provide theranostic tools of several types of malignancies. Because of their slow kinetics, mAbs require radioelements defined by a consistent half-life. Zirconium 89 (89Zr) and Copper 64 (64Cu) are radiometals with half-lives suitable for mAb labeling. Radiolabeling with a radiometal requires the prior use of a bifunctional chelate agent (BFCA) to functionalize mAb for radiometal chelation, in a second step. There are a number of BFCA available and much research is focused on antibody functionalization techniques or on developing the optimum chelating agent depending the selected radiometal. In this manuscript, we present a critical account of radiochemical techniques with radionuclides 89Zr and 64Cu and their applications in preclinical and clinical immuno-PET imaging.
Collapse
Affiliation(s)
| | - Isabelle Quelven
- Toulouse NeuroImaging Center (ToNIC), INSERM/UPS UMR 1214, University Hospital of Toulouse-Purpan, CEDEX 3, 31024 Toulouse, France;
| |
Collapse
|
9
|
Tripathy RK, Pande AH. Molecular and functional insight into anti-EGFR nanobody: Theranostic implications for malignancies. Life Sci 2024; 345:122593. [PMID: 38554946 DOI: 10.1016/j.lfs.2024.122593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/27/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Targeted therapy and imaging are the most popular techniques for the intervention and diagnosis of cancer. A potential therapeutic target for the treatment of cancer is the epidermal growth factor receptor (EGFR), primarily for glioblastoma, lung, and breast cancer. Over-production of ligand, transcriptional up-regulation due to autocrine/paracrine signalling, or point mutations at the genomic locus may contribute to the malfunction of EGFR in malignancies. This exploit makes use of EGFR, an established biomarker for cancer diagnostics and treatment. Despite considerable development in the last several decades in making EGFR inhibitors, they are still not free from limitations like toxicity and a short serum half-life. Nanobodies and antibodies share similar binding properties, but nanobodies have the additional advantage that they can bind to antigenic epitopes deep inside the target that conventional antibodies are unable to access. For targeted therapy, anti-EGFR nanobodies can be conjugated to various molecules such as drugs, peptides, toxins and photosensitizers. These nanobodies can be designed as novel immunoconjugates using the universal modular antibody-based platform technology (UniCAR). Furthermore, Anti-EGFR nanobodies can be expressed in neural stem cells and visualised by effective fluorescent and radioisotope labelling.
Collapse
Affiliation(s)
- Rajan K Tripathy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, (Mohali) 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, (Mohali) 160062, Punjab, India.
| |
Collapse
|
10
|
Suzuki H, Kannaka K, Uehara T. Approaches to Reducing Normal Tissue Radiation from Radiolabeled Antibodies. Pharmaceuticals (Basel) 2024; 17:508. [PMID: 38675468 PMCID: PMC11053530 DOI: 10.3390/ph17040508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Radiolabeled antibodies are powerful tools for both imaging and therapy in the field of nuclear medicine. Radiolabeling methods that do not release radionuclides from parent antibodies are essential for radiolabeling antibodies, and practical radiolabeling protocols that provide high in vivo stability have been established for many radionuclides, with a few exceptions. However, several limitations remain, including undesirable side effects on the biodistribution profiles of antibodies. This review summarizes the numerous efforts made to tackle this problem and the recent advances, mainly in preclinical studies. These include pretargeting approaches, engineered antibody fragments and constructs, the secondary injection of clearing agents, and the insertion of metabolizable linkages. Finally, we discuss the potential of these approaches and their prospects for further clinical application.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan; (K.K.); (T.U.)
| | | | | |
Collapse
|
11
|
Yao R, Zhu M, Guo Z, Shen J. Refining nanoprobes for monitoring of inflammatory bowel disease. Acta Biomater 2024; 177:37-49. [PMID: 38364928 DOI: 10.1016/j.actbio.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/11/2024] [Accepted: 02/09/2024] [Indexed: 02/18/2024]
Abstract
Inflammatory bowel disease (IBD) is a gastrointestinal immune disease that requires clear diagnosis, timely treatment, and lifelong monitoring. The diagnosis and monitoring methods of IBD mainly include endoscopy, imaging examination, and laboratory examination, which are constantly developed to achieve early definite diagnosis and accurate monitoring. In recent years, with the development of nanotechnology, the diagnosis and monitoring methods of IBD have been remarkably enriched. Nanomaterials, characterized by their minuscule dimensions that can be tailored, along with their distinctive optical, magnetic, and biodistribution properties, have emerged as valuable contrast agents for imaging and targeted agents for endoscopy. Through both active and passive targeting mechanisms, nanoparticles accumulate at the site of inflammation, thereby enhancing IBD detection. This review comprehensively outlines the existing IBD detection techniques, expounds upon the utilization of nanoparticles in IBD detection and diagnosis, and offers insights into the future potential of in vitro diagnostics. STATEMENT OF SIGNIFICANCE: Due to their small size and unique physical and chemical properties, nanomaterials are widely used in the biological and medical fields. In the area of oncology and inflammatory disease, an increasing number of nanomaterials are being developed for diagnostics and drug delivery. Here, we focus on inflammatory bowel disease, an autoimmune inflammatory disease that requires early diagnosis and lifelong monitoring. Nanomaterials can be used as contrast agents to visualize areas of inflammation by actively or passively targeting them through the intestinal mucosal epithelium where gaps exist due to inflammation stimulation. In this article, we summarize the utilization of nanoparticles in inflammatory bowel disease detection and diagnosis, and offers insights into the future potential of in vitro diagnostics.
Collapse
Affiliation(s)
- Ruchen Yao
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China; NHC Key Laboratory of Digestive Diseases, China
| | - Mingming Zhu
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China; NHC Key Laboratory of Digestive Diseases, China
| | - Zhiqian Guo
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Jun Shen
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China; NHC Key Laboratory of Digestive Diseases, China.
| |
Collapse
|
12
|
Zhang T, Lei H, Chen X, Dou Z, Yu B, Su W, Wang W, Jin X, Katsube T, Wang B, Zhang H, Li Q, Di C. Carrier systems of radiopharmaceuticals and the application in cancer therapy. Cell Death Discov 2024; 10:16. [PMID: 38195680 PMCID: PMC10776600 DOI: 10.1038/s41420-023-01778-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 01/11/2024] Open
Abstract
Radiopharmaceuticals play a vital role in cancer therapy. The carrier of radiopharmaceuticals can precisely locate and guide radionuclides to the target, where radionuclides kill surrounding tumor cells. Effective application of radiopharmaceuticals depends on the selection of an appropriate carrier. Herein, different types of carriers of radiopharmaceuticals and the characteristics are briefly described. Subsequently, we review radiolabeled monoclonal antibodies (mAbs) and their derivatives, and novel strategies of radiolabeled mAbs and their derivatives in the treatment of lymphoma and colorectal cancer. Furthermore, this review outlines radiolabeled peptides, and novel strategies of radiolabeled peptides in the treatment of neuroendocrine neoplasms, prostate cancer, and gliomas. The emphasis is given to heterodimers, bicyclic peptides, and peptide-modified nanoparticles. Last, the latest developments and applications of radiolabeled nucleic acids and small molecules in cancer therapy are discussed. Thus, this review will contribute to a better understanding of the carrier of radiopharmaceuticals and the application in cancer therapy.
Collapse
Affiliation(s)
- Taotao Zhang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Huiwen Lei
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Xiaohua Chen
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Zhihui Dou
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Boyi Yu
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Wei Su
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Wei Wang
- College of Life Science, Northwest Normal University, Lanzhou, 730000, China
| | - Xiaodong Jin
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Takanori Katsube
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Hong Zhang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| | - Qiang Li
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| | - Cuixia Di
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| |
Collapse
|
13
|
Kim S, Bae Y, Park SH, Chen N, Eom S, Kang S, Park J. Compact and modular bioprobe: Integrating SpyCatcher/SpyTag recombinant proteins with zwitterionic polymer-coated quantum dots. J Colloid Interface Sci 2023; 652:184-194. [PMID: 37595436 DOI: 10.1016/j.jcis.2023.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/05/2023] [Accepted: 08/04/2023] [Indexed: 08/20/2023]
Abstract
The development of quantum dot (QD)-based modular bioprobe that has a compact size and enable a facile conjugation of various biofunctional groups is in high demand. To address this, we surface engineered QDs with zwitterion polymer ligands to have an inherent compact size and derivatized them sequentially with the recombinant proteins SpyCatcher/SpyTag (SC/ST) to use their protein ligation system. SC/ST spontaneously form one complex through the isopeptide bond between them. SC-conjugated QDs (QD-SC) were used as base building blocks. Then, ST-biomolecules were added for modular biofunctionalization. We synthesized compact sized (∼15 nm) QD-SC-ST-affibody (antibody-mimicking small protein for tumor detection) conjugates, which showed successful cell-receptor targeting. The target cell-receptor could be easily tuned by changing the type of ST-affibody. We also demonstrated that anti-human-chorionic-gonadotropin mouse IgG1 antibodies can be labeled on the QD surface by mixing QD-SC with the ST-MG1Nb (mouse-IgG1-specific protein). The immunoassay performance of the antibody-labeled QDs was evaluated using a pregnancy test kit, displaying equivalent detection sensitivity to a commercially available kit. This study proposed an innovative strategy for QD biofunctionalization in a modular manner, which can be expanded to a diverse range of colloidal particle derivatization.
Collapse
Affiliation(s)
- Sunghwan Kim
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Yoonji Bae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Sung Han Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Ning Chen
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Soomin Eom
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| | - Jongnam Park
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| |
Collapse
|
14
|
Abenavoli EM, Linguanti F, Calabretta R, Delgado Bolton RC, Berti V, Lopci E. Clinical Application of ImmunoPET Targeting Checkpoint Inhibitors. Cancers (Basel) 2023; 15:5675. [PMID: 38067379 PMCID: PMC10705400 DOI: 10.3390/cancers15235675] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 10/16/2024] Open
Abstract
In the last decade, monoclonal antibodies (mAbs) targeting CTLA-4, PD-1, or PD-L1 have been developed and immune checkpoint inhibitors (ICIs) have become the main approach in cancer immunotherapy. However, not all patients benefit from ICI therapy and some are at risk of developing treatment-induced side-effects. These aspects, in parallel with the imaging challenges related to response assessments during immunotherapy, have driven scientific research to the discovery of new predictive biomarkers to individualize patients who could benefit from ICIs. In this context, molecular imaging using PET (positron emission tomography), which allows for whole-body tumor visualization, may be a promising non-invasive method for the determination of patients' sensitivity to antibody drugs. Several PET tracers, diverse from 2-[18F]FDG (or 2-Deoxy-2-[18F]fluoroglucose), have been developed to image immune checkpoints (ICs) or key elements of the immune system, although most of them are still in preclinical phases. Herein, we present the current state of the ImmunoPET-targeting of IC proteins with mAbs and antibody fragments, with a main focus on the latest developments in clinical molecular imaging studies of solid tumors. Moreover, given the relevance of the immune system and of tumor-infiltrating lymphocytes in particular in the prediction of the benefit of ICIs, we dedicate a portion of this review to ImmunoPET-targeting T cells.
Collapse
Affiliation(s)
| | - Flavia Linguanti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy (V.B.)
- Nuclear Medicine Department, Ospedale San Donato, 52100 Arezzo, Italy
| | - Raffaella Calabretta
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Roberto C. Delgado Bolton
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
- Servicio Cántabro de Salud, 39011 Santander, Spain
| | - Valentina Berti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy (V.B.)
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS—Humanitas Research Hospital, 20089 Rozzano, Italy
| |
Collapse
|
15
|
Ducharme M, Hall L, Eckenroad W, Cingoranelli SJ, Houson HA, Jaskowski L, Hunter C, Larimer BM, Lapi SE. Evaluation of [ 89Zr]Zr-DFO-2Rs15d Nanobody for Imaging of HER2-Positive Breast Cancer. Mol Pharm 2023; 20:4629-4639. [PMID: 37552575 PMCID: PMC11606513 DOI: 10.1021/acs.molpharmaceut.3c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
One of the most aggressive forms of breast cancer involves the overexpression of human epidermal growth factor receptor 2 (HER2). HER2 is overexpressed in ∼25% of all breast cancers and is associated with increased proliferation, increased rates of metastasis, and poor prognosis. Treatment for HER2-positive breast cancer has vastly improved since the development of the monoclonal antibody trastuzumab (Herceptin) as well as other biological constructs. However, patients still commonly develop resistance, illustrating the need for newer therapies. Nanobodies have become an important focus for potential development as HER2-targeting imaging agents and therapeutics. Nanobodies have many favorable characteristics, including high stability in heat and nonphysiological pH, while maintaining their low-nanomolar affinity for their designed targets. Specifically, the 2Rs15d nanobody has been developed for targeting HER2 and has been evaluated as a diagnostic imaging agent for single-photon emission computed tomography (SPECT) and positron emission tomography (PET). While a construct of 2Rs15d with the positron emitter 68Ga is currently in phase I clinical trials, the only PET images acquired in preclinical or clinical research have been within 3 h postinjection. We evaluated our in-house produced 2Rs15d nanobody, conjugated with the chelator deferoxamine (DFO), and radiolabeled with 89Zr for PET imaging up to 72 h postinjection. [89Zr]Zr-DFO-2Rs15d demonstrated high stability in both phosphate-buffered saline (PBS) and human serum. Cell binding studies showed high binding and specificity for HER2, as well as prominent internalization. Our in vivo PET imaging confirmed high-quality visualization of HER2-positive tumors up to 72 h postinjection, whereas HER2-negative tumors were not visualized. Subsequent biodistribution studies quantitatively supported the significant HER2-positive tumor uptake compared to the negative control. Our studies fill an important gap in understanding the imaging and binding properties of the 2Rs15d nanobody at extended time points. As many therapeutic radioisotopes have single or multiday half-lives, this information will directly benefit the potential of the radiotherapy development of 2Rs15d for HER2-positive breast cancer patients.
Collapse
Affiliation(s)
- Maxwell Ducharme
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Lucinda Hall
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Whitney Eckenroad
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Shelbie J Cingoranelli
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Hailey A Houson
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Luke Jaskowski
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Chanelle Hunter
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Benjamin M Larimer
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| |
Collapse
|
16
|
Husarova T, MacCuaig WM, Dennahy IS, Sanderson EJ, Edil BH, Jain A, Bonds MM, McNally MW, Menclova K, Pudil J, Zaruba P, Pohnan R, Henson CE, Grizzle WE, McNally LR. Intraoperative Imaging in Hepatopancreatobiliary Surgery. Cancers (Basel) 2023; 15:3694. [PMID: 37509355 PMCID: PMC10377919 DOI: 10.3390/cancers15143694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Hepatopancreatobiliary surgery belongs to one of the most complex fields of general surgery. An intricate and vital anatomy is accompanied by difficult distinctions of tumors from fibrosis and inflammation; the identification of precise tumor margins; or small, even disappearing, lesions on currently available imaging. The routine implementation of ultrasound use shifted the possibilities in the operating room, yet more precision is necessary to achieve negative resection margins. Modalities utilizing fluorescent-compatible dyes have proven their role in hepatopancreatobiliary surgery, although this is not yet a routine practice, as there are many limitations. Modalities, such as photoacoustic imaging or 3D holograms, are emerging but are mostly limited to preclinical settings. There is a need to identify and develop an ideal contrast agent capable of differentiating between malignant and benign tissue and to report on the prognostic benefits of implemented intraoperative imaging in order to navigate clinical translation. This review focuses on existing and developing imaging modalities for intraoperative use, tailored to the needs of hepatopancreatobiliary cancers. We will also cover the application of these imaging techniques to theranostics to achieve combined diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Tereza Husarova
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - William M. MacCuaig
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Isabel S. Dennahy
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Emma J. Sanderson
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Barish H. Edil
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Ajay Jain
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Morgan M. Bonds
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Molly W. McNally
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Katerina Menclova
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - Jiri Pudil
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - Pavel Zaruba
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - Radek Pohnan
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - Christina E. Henson
- Department of Radiation Oncology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lacey R. McNally
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
17
|
Natarajan A, Khan S, Liang X, Nguyen H, Das N, Anders D, Malik N, Oderinde OM, Chin FT, Rosenthal E, Pratx G. Preclinical Evaluation of 89Zr-Panitumumab for Biology-Guided Radiation Therapy. Int J Radiat Oncol Biol Phys 2023; 116:927-934. [PMID: 36669541 PMCID: PMC11290461 DOI: 10.1016/j.ijrobp.2023.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/27/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023]
Abstract
PURPOSE Biology-guided radiation therapy (BgRT) uses real-time line-of-response data from on-board positron emission tomography (PET) detectors to guide beamlet delivery during therapeutic radiation. The current workflow requires 18F-fluorodeoxyglucose (FDG) administration daily before each treatment fraction. However, there are advantages to reducing the number of tracer injections by using a PET tracer with a longer decay time. In this context, we investigated 89Zr-panitumumab (89Zr-Pan), an antibody PET tracer with a half-life of 78 hours that can be imaged for up to 9 days using PET. METHODS AND MATERIALS The BgRT workflow was evaluated preclinically in mouse colorectal cancer xenografts (HCT116) using small-animal positron emission tomography/computed tomography (PET/CT) for imaging and image-guided kilovoltage conformal irradiation for therapy. Mice (n = 5 per group) received 7 MBq of 89Zr-Pan as a single dose 2 weeks after tumor induction, with or without fractionated radiation therapy (RT; 6 × 6.6 Gy) to the tumor region. The mice were imaged longitudinally to assess the kinetics of the tracer over 9 days. PET images were then analyzed to determine the stability of the PET signal in irradiated tumors over time. RESULTS Mice in the treatment group experienced complete tumor regression, whereas those in the control group were killed because of tumor burden. PET imaging of 89Zr-Pan showed well-delineated tumors with minimal background in both groups. On day 9 postinjection, tumor uptake of 89Zr-Pan was 7.2 ± 1.7 in the control group versus 5.2 ± 0.5 in the treatment group (mean percentage of injected dose per gram of tissue [%ID/g] ± SD; P = .07), both significantly higher than FDG uptake (1.1 ± 0.5 %ID/g) 1 hour postinjection. To assess BgRT feasibility, the clinical eligibility criteria was computed using human-equivalent uptake values that were extrapolated from preclinical PET data. Based on this semiquantitative analysis, BgRT may be feasible for 5 consecutive days after a single 740-MBq injection of 89Zr-Pan. CONCLUSIONS This study indicates the potential of long-lived antibody-based PET tracers for guiding clinical BgRT.
Collapse
Affiliation(s)
| | - Syamantak Khan
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Xuanwei Liang
- Department of Physics, Foothill College, Los Altos, California
| | - Hieu Nguyen
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Neeladrisingha Das
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - David Anders
- Department of Radiology, Stanford University, Stanford, California
| | - Noeen Malik
- Department of Radiology, Stanford University, Stanford, California
| | | | - Frederick T Chin
- Department of Radiology, Stanford University, Stanford, California
| | - Eben Rosenthal
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Guillem Pratx
- Department of Radiation Oncology, Stanford University, Stanford, California.
| |
Collapse
|
18
|
Mulgaonkar A, Udayakumar D, Yang Y, Harris S, Öz OK, Ramakrishnan Geethakumari P, Sun X. Current and potential roles of immuno-PET/-SPECT in CAR T-cell therapy. Front Med (Lausanne) 2023; 10:1199146. [PMID: 37441689 PMCID: PMC10333708 DOI: 10.3389/fmed.2023.1199146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/15/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have evolved as breakthrough treatment options for the management of hematological malignancies and are also being developed as therapeutics for solid tumors. However, despite the impressive patient responses from CD19-directed CAR T-cell therapies, ~ 40%-60% of these patients' cancers eventually relapse, with variable prognosis. Such relapses may occur due to a combination of molecular resistance mechanisms, including antigen loss or mutations, T-cell exhaustion, and progression of the immunosuppressive tumor microenvironment. This class of therapeutics is also associated with certain unique toxicities, such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and other "on-target, off-tumor" toxicities, as well as anaphylactic effects. Furthermore, manufacturing limitations and challenges associated with solid tumor infiltration have delayed extensive applications. The molecular imaging modalities of immunological positron emission tomography and single-photon emission computed tomography (immuno-PET/-SPECT) offer a target-specific and highly sensitive, quantitative, non-invasive platform for longitudinal detection of dynamic variations in target antigen expression in the body. Leveraging these imaging strategies as guidance tools for use with CAR T-cell therapies may enable the timely identification of resistance mechanisms and/or toxic events when they occur, permitting effective therapeutic interventions. In addition, the utilization of these approaches in tracking the CAR T-cell pharmacokinetics during product development and optimization may help to assess their efficacy and accordingly to predict treatment outcomes. In this review, we focus on current challenges and potential opportunities in the application of immuno-PET/-SPECT imaging strategies to address the challenges encountered with CAR T-cell therapies.
Collapse
Affiliation(s)
- Aditi Mulgaonkar
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Durga Udayakumar
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yaxing Yang
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Shelby Harris
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Orhan K. Öz
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Praveen Ramakrishnan Geethakumari
- Section of Hematologic Malignancies/Transplant and Cell Therapy, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
19
|
Morito T, Harada R, Iwata R, Ishikawa Y, Okamura N, Kudo Y, Furumoto S, Yanai K, Tashiro M. Evaluation of 18F labeled glial fibrillary acidic protein binding nanobody and its brain shuttle peptide fusion proteins using a neuroinflammation rat model. PLoS One 2023; 18:e0287047. [PMID: 37315033 DOI: 10.1371/journal.pone.0287047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 05/27/2023] [Indexed: 06/16/2023] Open
Abstract
Astrogliosis is a crucial feature of neuroinflammation and is characterized by the significant upregulation of glial fibrillary acidic protein (GFAP) expression. Hence, visualizing GFAP in the living brain of patients with damaged central nervous system using positron emission tomography (PET) is of great importance, and it is expected to depict neuroinflammation more directly than existing neuroinflammation imaging markers. However, no PET radiotracers for GFAP are currently available. Therefore, neuroimaging with antibody-like affinity proteins could be a viable strategy for visualizing imaging targets that small molecules rarely recognize, such as GFAP, while we need to overcome the challenges of slow clearance and low brain permeability. The E9 nanobody, a small-affinity protein with high affinity and selectivity for GFAP, was utilized in this study. E9 was engineered by fusing a brain shuttle peptide that facilitates blood-brain barrier permeation via two different types of linker domains: E9-GS-ApoE (EGA) and E9-EAK-ApoE (EEA). E9, EGA and EEA were radiolabeled with fluorine-18 using cell-free protein radiosynthesis. In vitro autoradiography showed that all radiolabeled proteins exhibited a significant difference in neuroinflammation in the brain sections created from a rat model constructed by injecting lipopolysaccharide (LPS) into the unilateral striatum of wildtype rats, and an excess competitor displaced their binding. However, exploratory in vivo PET imaging and ex vivo biodistribution studies in the rat model failed to distinguish neuroinflammatory lesions within 3 h of 18F-EEA intravenous injection. This study contributes to a better understanding of the characteristics of small-affinity proteins fused with a brain shuttle peptide for further research into the use of protein molecules as PET tracers for imaging neuropathology.
Collapse
Affiliation(s)
- Takahiro Morito
- Division of Cyclotron Nuclear Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Ren Iwata
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Miyagi, Japan
| | - Yoichi Ishikawa
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Miyagi, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Yukitsuka Kudo
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Shozo Furumoto
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Miyagi, Japan
| | - Kazuhiko Yanai
- Division of Cyclotron Nuclear Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Manabu Tashiro
- Division of Cyclotron Nuclear Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
20
|
Xia W, Singh N, Goel S, Shi S. Molecular Imaging of Innate Immunity and Immunotherapy. Adv Drug Deliv Rev 2023; 198:114865. [PMID: 37182699 DOI: 10.1016/j.addr.2023.114865] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/17/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
The innate immune system plays a key role as the first line of defense in various human diseases including cancer, cardiovascular and inflammatory diseases. In contrast to tissue biopsies and blood biopsies, in vivo imaging of the innate immune system can provide whole body measurements of immune cell location and function and changes in response to disease progression and therapy. Rationally developed molecular imaging strategies can be used in evaluating the status and spatio-temporal distributions of the innate immune cells in near real-time, mapping the biodistribution of novel innate immunotherapies, monitoring their efficacy and potential toxicities, and eventually for stratifying patients that are likely to benefit from these immunotherapies. In this review, we will highlight the current state-of-the-art in noninvasive imaging techniques for preclinical imaging of the innate immune system particularly focusing on cell trafficking, biodistribution, as well as pharmacokinetics and dynamics of promising immunotherapies in cancer and other diseases; discuss the unmet needs and current challenges in integrating imaging modalities and immunology and suggest potential solutions to overcome these barriers.
Collapse
Affiliation(s)
- Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States.
| |
Collapse
|
21
|
Bai JW, Qiu SQ, Zhang GJ. Molecular and functional imaging in cancer-targeted therapy: current applications and future directions. Signal Transduct Target Ther 2023; 8:89. [PMID: 36849435 PMCID: PMC9971190 DOI: 10.1038/s41392-023-01366-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 01/19/2023] [Accepted: 02/14/2023] [Indexed: 03/01/2023] Open
Abstract
Targeted anticancer drugs block cancer cell growth by interfering with specific signaling pathways vital to carcinogenesis and tumor growth rather than harming all rapidly dividing cells as in cytotoxic chemotherapy. The Response Evaluation Criteria in Solid Tumor (RECIST) system has been used to assess tumor response to therapy via changes in the size of target lesions as measured by calipers, conventional anatomically based imaging modalities such as computed tomography (CT), and magnetic resonance imaging (MRI), and other imaging methods. However, RECIST is sometimes inaccurate in assessing the efficacy of targeted therapy drugs because of the poor correlation between tumor size and treatment-induced tumor necrosis or shrinkage. This approach might also result in delayed identification of response when the therapy does confer a reduction in tumor size. Innovative molecular imaging techniques have rapidly gained importance in the dawning era of targeted therapy as they can visualize, characterize, and quantify biological processes at the cellular, subcellular, or even molecular level rather than at the anatomical level. This review summarizes different targeted cell signaling pathways, various molecular imaging techniques, and developed probes. Moreover, the application of molecular imaging for evaluating treatment response and related clinical outcome is also systematically outlined. In the future, more attention should be paid to promoting the clinical translation of molecular imaging in evaluating the sensitivity to targeted therapy with biocompatible probes. In particular, multimodal imaging technologies incorporating advanced artificial intelligence should be developed to comprehensively and accurately assess cancer-targeted therapy, in addition to RECIST-based methods.
Collapse
Affiliation(s)
- Jing-Wen Bai
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Department of Medical Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Si-Qi Qiu
- Diagnosis and Treatment Center of Breast Diseases, Clinical Research Center, Shantou Central Hospital, 515041, Shantou, China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, 515041, Shantou, China
| | - Guo-Jun Zhang
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
| |
Collapse
|
22
|
Du W, Jiang P, Li Q, Wen H, Zheng M, Zhang J, Guo Y, Yang J, Feng W, Ye S, Kamara S, Jiang P, Chen J, Li W, Zhu S, Zhang L. Novel Affibody Molecules Specifically Bind to SARS-CoV-2 Spike Protein and Efficiently Neutralize Delta and Omicron Variants. Microbiol Spectr 2023; 11:e0356222. [PMID: 36511681 PMCID: PMC9927262 DOI: 10.1128/spectrum.03562-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has been an unprecedented public health disaster in human history, and its spike (S) protein is the major target for vaccines and antiviral drug development. Although widespread vaccination has been well established, the viral gene is prone to rapid mutation, resulting in multiple global spread waves. Therefore, specific antivirals are needed urgently, especially those against variants. In this study, the domain of the receptor binding motif (RBM) and fusion peptide (FP) (amino acids [aa] 436 to 829; denoted RBMFP) of the SARS-CoV-2 S protein was expressed as a recombinant RBMFP protein in Escherichia coli and identified as being immunogenic and antigenically active. Then, the RBMFP proteins were used for phage display to screen the novel affibody. After prokaryotic expression and selection, four novel affibody molecules (Z14, Z149, Z171, and Z327) were obtained. Through surface plasmon resonance (SPR) and pseudovirus neutralization assay, we showed that affibody molecules specifically bind to the RBMFP protein with high affinity and neutralize against SARS-CoV-2 pseudovirus infection. Especially, Z14 and Z171 displayed strong neutralizing activities against Delta and Omicron variants. Molecular docking predicted that affibody molecule interaction sites with RBM overlapped with ACE2. Thus, the novel affibody molecules could be further developed as specific neutralization agents against SARS-CoV-2 variants. IMPORTANCE SARS-CoV-2 and its variants are threatening the whole world. Although a full dose of vaccine injection showed great preventive effects and monoclonal antibody reagents have also been used for a specific treatment, the global pandemic persists. So, developing new vaccines and specific agents are needed urgently. In this work, we expressed the recombinant RBMFP protein as an antigen, identified its antigenicity, and used it as an antigen for affibody phage-display selection. After the prokaryotic expression, the specific affibody molecules were obtained and tested for pseudovirus neutralization. Results showed that the serum antibody induced by RBMFP neutralized Omicron variants. The screened affibody molecules specifically bound the RBMFP of SARS-CoV-2 with high affinity and neutralized the Delta and Omicron pseudovirus in vitro. So, the RBMFP induced serum provides neutralizing effects against pseudovirus in vitro, and the affibodies have the potential to be developed into specific prophylactic agents for SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Wangqi Du
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peipei Jiang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingfeng Li
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - He Wen
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Maolin Zheng
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanru Guo
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia Yang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weixu Feng
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sisi Ye
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Saidu Kamara
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Pengfei Jiang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Chen
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenshu Li
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shanli Zhu
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lifang Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
23
|
ImmunoPET Directed to the Brain: A New Tool for Preclinical and Clinical Neuroscience. Biomolecules 2023; 13:biom13010164. [PMID: 36671549 PMCID: PMC9855881 DOI: 10.3390/biom13010164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Immuno-positron emission tomography (immunoPET) is a non-invasive in vivo imaging method based on tracking and quantifying radiolabeled monoclonal antibodies (mAbs) and other related molecules, such as antibody fragments, nanobodies, or affibodies. However, the success of immunoPET in neuroimaging is limited because intact antibodies cannot penetrate the blood-brain barrier (BBB). In neuro-oncology, immunoPET has been successfully applied to brain tumors because of the compromised BBB. Different strategies, such as changes in antibody properties, use of physiological mechanisms in the BBB, or induced changes to BBB permeability, have been developed to deliver antibodies to the brain. These approaches have recently started to be applied in preclinical central nervous system PET studies. Therefore, immunoPET could be a new approach for developing more specific PET probes directed to different brain targets.
Collapse
|
24
|
Heo GS, Diekmann J, Thackeray JT, Liu Y. Nuclear Methods for Immune Cell Imaging: Bridging Molecular Imaging and Individualized Medicine. Circ Cardiovasc Imaging 2023; 16:e014067. [PMID: 36649445 PMCID: PMC9858352 DOI: 10.1161/circimaging.122.014067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Inflammation is a key mechanistic contributor to the progression of cardiovascular disease, from atherosclerosis through ischemic injury and overt heart failure. Recent evidence has identified specific roles of immune cell subpopulations in cardiac pathogenesis that diverges between individual patients. Nuclear imaging approaches facilitate noninvasive and serial quantification of inflammation severity, offering the opportunity to predict eventual outcome, stratify patient risk, and guide novel targeted molecular therapies against specific leukocyte subpopulations. Here, we will discuss the established and emerging nuclear imaging methods to label and track exogenous and endogenous immune cells, with a particular focus on clinical situations in which targeted molecular inflammation imaging would be advantageous. The expanding options for imaging inflammation provide the foundation to bridge between molecular imaging and individual therapy.
Collapse
Affiliation(s)
- Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO (G.S.H., Y. L.)
| | - Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany (J.D., J.T.T.)
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany (J.D., J.T.T.)
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO (G.S.H., Y. L.)
| |
Collapse
|
25
|
Freidel L, Li S, Choffart A, Kuebler L, Martins AF. Imaging Techniques in Pharmacological Precision Medicine. Handb Exp Pharmacol 2023; 280:213-235. [PMID: 36907970 DOI: 10.1007/164_2023_641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Biomedical imaging is a powerful tool for medical diagnostics and personalized medicines. Examples of commonly used imaging modalities include Positron Emission Tomography (PET), Ultrasound (US), Single Photon Emission Computed Tomography (SPECT), and hybrid imaging. By combining these modalities, scientists can gain a comprehensive view and better understand physiology and pathology at the preclinical, clinical, and multiscale levels. This can aid in the accuracy of medical diagnoses and treatment decisions. Moreover, biomedical imaging allows for evaluating the metabolic, functional, and structural details of living tissues. This can be particularly useful for the early diagnosis of diseases such as cancer and for the application of personalized medicines. In the case of hybrid imaging, two or more modalities are combined to produce a high-resolution image with enhanced sensitivity and specificity. This can significantly improve the accuracy of diagnosis and offer more detailed treatment plans. In this book chapter, we showcase how continued advancements in biomedical imaging technology can potentially revolutionize medical diagnostics and personalized medicine.
Collapse
Affiliation(s)
- Lucas Freidel
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| | - Sixing Li
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| | - Anais Choffart
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| | - Laura Kuebler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - André F Martins
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany.
- German Cancer Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
26
|
Conformation specific antagonistic high affinity antibodies to the RON receptor kinase for imaging and therapy. Sci Rep 2022; 12:22564. [PMID: 36581692 PMCID: PMC9800565 DOI: 10.1038/s41598-022-26404-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 12/14/2022] [Indexed: 12/31/2022] Open
Abstract
The RON receptor tyrosine kinase is an exceptionally interesting target in oncology and immunology. It is not only overexpressed in a wide variety of tumors but also has been shown to be expressed on myeloid cells associated with tumor infiltration, where it serves to dampen tumour immune responses and reduce the efficacy of anti-CTLA4 therapy. Potent and selective inhibitory antibodies to RON might therefore both inhibit tumor cell growth and stimulate immune rejection of tumors. We derived cloned and sequenced a new panel of exceptionally avid anti-RON antibodies with picomolar binding affinities that inhibit MSP-induced RON signaling and show remarkable potency in antibody dependent cellular cytotoxicity. Antibody specificity was validated by cloning the antibody genes and creating recombinant antibodies and by the use of RON knock out cell lines. When radiolabeled with 89-Zirconium, the new antibodies 3F8 and 10G1 allow effective immuno-positron emission tomography (immunoPET) imaging of RON-expressing tumors and recognize universally exposed RON epitopes at the cell surface. The 10G1 was further developed into a novel bispecific T cell engager with a 15 pM EC50 in cytotoxic T cell killing assays.
Collapse
|
27
|
Synthesis and evaluation of a para-carboxylated benzyl-DOTA for labeling peptides and polypeptides. Nucl Med Biol 2022; 114-115:18-28. [PMID: 36088873 DOI: 10.1016/j.nucmedbio.2022.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/02/2022] [Accepted: 08/17/2022] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Radiolabeled peptides and low-molecular-weight (LMW) polypeptides show high and persistent radioactivity levels in the kidney. To develop a DOTA-based bifunctional chelating agent that provides a radiometabolite with a rapid elimination rate from the kidney, a para-carboxyl Bn-DOTA (p-COOH-Bn-DOTA) was designed, synthesized, and evaluated. METHODS A precursor compound, p-COOH-Bn-DOTA(tBu)4, was synthesized in 9 steps using N-Boc-p-iodo-L-phenylalanine as the starting material. A synthetic somatostatin analog (TOC) was used as a representative peptide metabolized in the renal lysosomes. p-COOH-Bn-DOTA-conjugated TOC (DOTA-Bn-TOC) was synthesized by the conventional solid-phase peptide synthesis using p-COOH-Bn-DOTA(tBu)4. DOTA-tris(tBu ester) was also conjugated with TOC to prepare DOTATOC. 111In-labeling of the peptides was conducted under similar conditions. The radiochemical conversions, stability against apo-transferrin (apoTf), and in vivo behaviors were compared. RESULTS [111In]In-DOTA-Bn-TOC was obtained with higher radiochemical conversions than [111In]In-DOTATOC. Both 111In-labeled TOC derivatives remained stable against apoTf. In biodistribution studies, [111In]In-DOTA-Bn-TOC exhibited higher initial uptake in the kidney, followed by a faster elimination rate of radioactivity into the urine than [111In]In-DOTATOC. The metabolic studies showed that the shorter residence time of the radiometabolite from [111In]In-DOTA-Bn-TOC was responsible for the renal radioactivity decline. CONCLUSION p-COOH-Bn-DOTA provides stable 111In-labeled peptides in high yields at low peptide concentrations. p-COOH-Bn-DOTA also provides a radiometabolite with a short residence time in the kidney. Such characteristics would render p-COOH-Bn-DOTA useful to the future application to radiolabeled LMW polypeptides with low renal radioactivity levels.
Collapse
|
28
|
Niessen VJA, Wenker STM, Lam MGEH, van Noesel MM, Poot AJ. Biologicals as theranostic vehicles in paediatric oncology. Nucl Med Biol 2022; 114-115:58-64. [PMID: 36126433 DOI: 10.1016/j.nucmedbio.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 12/27/2022]
Abstract
Biologicals, such as antibodies or antibody-fragments e.g. nanobodies, have changed the landscape of cancer therapy and can be used in combination with traditional cancer treatments. They have been demonstrated to be excellent vehicles for molecular imaging. Several biologicals for nuclear imaging of adult cancer may be used in combination with (nuclear) therapy. Though it's great potential, molecular imaging using biologicals is rarely applied in paediatric oncology. This paper describes the current status of biologicals as radiopharmaceuticals for childhood cancer. Furthermore, the importance and potential for developing additional biological theranostics as opportunity to image and treat childhood cancer is discussed.
Collapse
Affiliation(s)
- Veerle J A Niessen
- Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands; Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands.
| | - Sylvia T M Wenker
- Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands; Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands.
| | - Marnix G E H Lam
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands.
| | - Max M van Noesel
- Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands.
| | - Alex J Poot
- Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands; Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands.
| |
Collapse
|
29
|
Baurand PE, Balland J, Reynas C, Ramseyer M, Vivier D, Bellaye PS, Collin B, Paul C, Denat F, Asgarov K, Pallandre JR, Ringenbach L. Development of Anti-LRRC15 Small Fragments for Imaging Purposes Using a Phage-Display ScFv Approach. Int J Mol Sci 2022; 23:ijms232012677. [PMID: 36293532 PMCID: PMC9604383 DOI: 10.3390/ijms232012677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
The human leucine-rich repeat-containing protein 15 (LRRC15) is a membrane protein identified as a marker of CAF (cancer-associated fibroblast) cells whose overexpression is positively correlated with cancer grade and outcome. Nuclear molecular imaging (i.e., SPECT and PET) to track LRRC15 expression could be very useful in guiding further therapeutic strategies. In this study, we developed an ScFv mouse phage-display library to obtain small fragment antibodies against human LRRC15 for molecular imaging purposes. Mice were immunized with recombinant human LRRC15 (hLRRC15), and lymph node cells were harvested for ScFv (single-chain variable fragment) phage-display analysis. The built library was used for panning on cell lines with constitutive or induced expression after transfection. The choice of best candidates was performed by screening various other cell lines, using flow cytometry. The selected candidates were reformatted into Cys-ScFv or Cys-diabody by addition of cysteine, and cloned in mammalian expression vectors to obtain batches of small fragments that were further used in site-specific radiolabeling tests. The obtained library was 1.2 × 107 cfu/µg with an insertion rate >95%. The two panning rounds performed on cells permittedenrichment of 2 × 10−3. Screening with flow cytometry allowed us to identify 28 specific hLRRC15 candidates. Among these, two also recognized murine LRCC15 and were reformatted into Cys-ScFv and Cys-diabody. They were expressed transiently in a mammalian system to obtain 1.0 to 4.5 mg of Cys fragments ready for bioconjugation and radiolabeling. Thus, in this paper, we demonstrate the relevance of the phage-display ScFv library approach for the fast-track development of small antibodies for imaging and/or immunotherapy purposes.
Collapse
Affiliation(s)
- Pierre-Emmanuel Baurand
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
- Correspondence:
| | - Jérémy Balland
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
| | - Chloé Reynas
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
| | - Mélanie Ramseyer
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
| | - Delphine Vivier
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS 6302, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Pierre-Simon Bellaye
- Plateforme D’imagerie et de Radiothérapie Précliniques (PIRP), Service de Médecine Nucléaire, Centre Georges-François Leclerc, 1 Rue du Pr Marion, 21000 Dijon, France
| | - Bertrand Collin
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS 6302, Université de Bourgogne Franche-Comté, 21000 Dijon, France
- Plateforme D’imagerie et de Radiothérapie Précliniques (PIRP), Service de Médecine Nucléaire, Centre Georges-François Leclerc, 1 Rue du Pr Marion, 21000 Dijon, France
| | - Catherine Paul
- Laboratoire d’Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Franck Denat
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS 6302, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Kamal Asgarov
- INSERM, EFS BFC, UMR1098, RIGHT, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, 25000 Besançon, France
- Clinical Investigation Center in Biotherapy, INSERM CIC-BT1431, Besançon University Hospital, 25000 Besançon, France
- ITAC Platform, University of Bourgogne Franche-Comté, 25000 Besançon, France
| | - Jean-René Pallandre
- INSERM, EFS BFC, UMR1098, RIGHT, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, 25000 Besançon, France
| | - Laurence Ringenbach
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
| |
Collapse
|
30
|
Positron Emission Tomography Probes for Imaging Cytotoxic Immune Cells. Pharmaceutics 2022; 14:pharmaceutics14102040. [PMID: 36297474 PMCID: PMC9610635 DOI: 10.3390/pharmaceutics14102040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022] Open
Abstract
Non-invasive positron emission tomography (PET) imaging of immune cells is a powerful approach for monitoring the dynamics of immune cells in response to immunotherapy. Despite the clinical success of many immunotherapeutic agents, their clinical efficacy is limited to a subgroup of patients. Conventional imaging, as well as analysis of tissue biopsies and blood samples do not reflect the complex interaction between tumour and immune cells. Consequently, PET probes are being developed to capture the dynamics of such interactions, which may improve patient stratification and treatment evaluation. The clinical efficacy of cancer immunotherapy relies on both the infiltration and function of cytotoxic immune cells at the tumour site. Thus, various immune biomarkers have been investigated as potential targets for PET imaging of immune response. Herein, we provide an overview of the most recent developments in PET imaging of immune response, including the radiosynthesis approaches employed in their development.
Collapse
|
31
|
Clausen AS, Christensen C, Christensen E, Cold S, Kristensen LK, Hansen AE, Kjaer A. Development of a 64Cu-labeled CD4+ T cell targeting PET tracer: evaluation of CD4 specificity and its potential use in collagen-induced arthritis. EJNMMI Res 2022; 12:62. [PMID: 36114433 PMCID: PMC9481863 DOI: 10.1186/s13550-022-00934-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background CD4+ T cells are central inflammatory mediators in the pathogenesis of autoimmune rheumatoid arthritis (RA), as they are one of the dominating cell types in synovial inflammation. Molecular imaging of CD4+ T cells has potential role for early detection and monitoring of RA. Here, we developed a new radiotracer for in vivo immunoPET imaging of murine CD4+ T cells and tested it in the collagen-induced arthritis (CIA) mouse model of human RA. Results The tracer, [64Cu]Cu-NOTA-CD4-F(ab)’2 ([64Cu]Cu-NOTA-CD4), was generated from F(ab)’2 fragments of R-anti-mouse CD4 antibodies conjugated to the 2-S-(isothiocyanatbenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA) chelator and radiolabeled with copper-64. Accumulation of the tracer and isotype control was evaluated in the CIA model and mice receiving whole-body irradiation (WBI) (5 Gy). The potential of [64Cu]Cu-NOTA-CD4 for response assessment was evaluated in CIA induced mice treated with dexamethasone (DXM). Imaging data were compared with flow cytometry and immunohistochemistry (IHC) of inflammatory cells including CD4+ T cells. [64Cu]Cu-NOTA-CD4 showed increased accumulation in T cell-rich tissues compared with isotype control (p < 0.0001). In addition, reduced accumulation of [64Cu]Cu-NOTA-CD4 was observed in T cell-depleted tissue (p < 0.0001). Flow cytometry and IHC confirmed the increased infiltration of CD4+ T cells in CIA mice. Conclusions We developed and evaluated a new radiotracer, [64Cu]Cu-NOTA-CD4, for immunoPET imaging of murine CD4+ T cells. [64Cu]Cu-NOTA-CD4 was successfully synthesized by F(ab)’2 fragments of R-anti-mouse CD4 antibodies conjugated to a chelator and radiolabeled with copper-64. We found that our novel CD4 PET tracer can be used for noninvasive visualization of murine CD4+ T cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-022-00934-7.
Collapse
|
32
|
Mulero F. ImmunoPET in oncology. Rev Esp Med Nucl Imagen Mol 2022; 41:332-339. [PMID: 35961857 DOI: 10.1016/j.remnie.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 01/14/2023]
Abstract
Due to increase of immunotherapy in oncology, it is essential to have a biological characterization of tumors. Knowing which antigens are expressed both on the surface of the tumor cell and at tumor microenvironment in order to predict the tretment response different therapeutic antibodies, has become a need. ImmunoPET is a non-invasive diagnostic imaging tool that combines the high specificity of antibodies against antigens with the high sensitivity, resolution and quantification capacity of PET imaging. With ImmunoPET we obtain a virtual biopsy of tumors, it has a big present and future in preclinical-clinical research, being already a reality in predicting and monitoring the response to treatments with monoclonal antibodies, allowing a selection of patients and therapies reaching a personalized medicine contributing to improve clinical decisions.
Collapse
Affiliation(s)
- Francisca Mulero
- Unidad de Imagen Molecular, Centro Nacional de Investigaciones Oncológicas, Melchor Fernández Almagro, 3, Madrid, Spain.
| |
Collapse
|
33
|
InmunoPET en oncología. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
34
|
Tran VL, Bouleau A, Nozach H, Richard M, Chevaleyre C, Dubois S, Kereselidze D, Kuhnast B, Evans MJ, Specklin S, Truillet C. Impact of Radiolabeling Strategies on the Pharmacokinetics and Distribution of an Anti-PD-L1 PET Ligand. Mol Pharm 2022; 19:3673-3680. [PMID: 35998011 DOI: 10.1021/acs.molpharmaceut.2c00497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Molecular imaging with PET offers an alternative method to quantify programmed-death-ligand 1 (PD-L1) to accurately select patients for immunotherapies. More and more clinical and preclinical trials involve radiolabeling of antibody fragments for their desirably fast clearance and high tumor penetration. As the radiolabeling strategy can significantly impact pharmacokinetics and biodistribution, we explored in this work a site-specific radiofluorination strategy on an anti-PD-L1 fragment antigen-binding (Fab) and compared the pharmacokinetic and biodistribution properties with the same Fab labeled using stochastic radiolabeling chemistry. We applied an enzymatic bioconjugation mediated by a variant of the lipoic acid ligase (LplA) that promotes the formation of an amide bond between a short peptide cloned onto the C terminus of the Fab. A synthetic analogue of the enzyme natural substrate, lipoic acid, was radiolabeled with fluorine-18 for site-specific conjugation by LplA. We compared the biodistribution of the site-specifically labeled Fab with a stochastically labeled Fab on lysine side chains in tumor-bearing mice. The two methods of fluorination demonstrate a comparable whole-body biodistribution. The 89Zr-labeled Fab had different biodistribution compared to either 18F-labeled Fab. We attribute the difference to [89Zr] metabolism. Fab-LAP-[18F]FPyOctA therefore reflects better the true pharmacokinetic profile of the Fab.
Collapse
Affiliation(s)
- Vu Long Tran
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Alizée Bouleau
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Hervé Nozach
- Université Paris-Saclay, CEA, DMTS, SIMoS, CEA-Saclay, Gif-sur-Yvette CEDEX 91191, France
| | - Mylène Richard
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Céline Chevaleyre
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Steven Dubois
- Université Paris-Saclay, CEA, DMTS, SIMoS, CEA-Saclay, Gif-sur-Yvette CEDEX 91191, France
| | - Dimitri Kereselidze
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Bertrand Kuhnast
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California 94107, United States
| | - Simon Specklin
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| |
Collapse
|
35
|
Xiong Y, Zhang Y, Li Z, Reza MS, Li X, Tian X, Ai HW. Engineered Amber-Emitting Nano Luciferase and Its Use for Immunobioluminescence Imaging In Vivo. J Am Chem Soc 2022; 144:14101-14111. [PMID: 35913786 PMCID: PMC9425369 DOI: 10.1021/jacs.2c02320] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The NanoLuc luciferase (NLuc) and its furimazine (FRZ) substrate have revolutionized bioluminescence (BL) assays and imaging. However, the use of the NLuc-FRZ luciferase-luciferin pair for mammalian tissue imaging is hindered by the low tissue penetration of the emitting blue photons. Here, we present the development of an NLuc mutant, QLuc, which catalyzes the oxidation of a synthetic QTZ luciferin for bright and red-shifted emission peaking at ∼585 nm. Compared to other small single-domain NLuc mutants, this amber-light-emitting luciferase exhibited improved performance for imaging deep-tissue targets in live mice. Leveraging this novel bioluminescent reporter, we further pursued in vivo immunobioluminescence imaging (immunoBLI), which used a fusion protein of a single-chain variable antibody fragment (scFv) and QLuc for molecular imaging of tumor-associated antigens in a xenograft mouse model. As one of the most red-shifted NLuc variants, we expect QLuc to find broad applications in noninvasive mammalian imaging. Moreover, the immunoBLI method complements immunofluorescence imaging and immuno-positron emission tomography (immunoPET), serving as a convenient and nonradioactive molecular imaging tool for animal models in basic and preclinical research.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Yiyu Zhang
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Zefan Li
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Md Shamim Reza
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Xinyu Li
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Xiaodong Tian
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Hui-wang Ai
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- The UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22908, USA
| |
Collapse
|
36
|
Current state and upcoming opportunities for immunoPET biomarkers in lung cancer. Lung Cancer 2022; 169:84-93. [DOI: 10.1016/j.lungcan.2022.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 11/21/2022]
|
37
|
Manafi-Farid R, Ataeinia B, Ranjbar S, Jamshidi Araghi Z, Moradi MM, Pirich C, Beheshti M. ImmunoPET: Antibody-Based PET Imaging in Solid Tumors. Front Med (Lausanne) 2022; 9:916693. [PMID: 35836956 PMCID: PMC9273828 DOI: 10.3389/fmed.2022.916693] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Immuno-positron emission tomography (immunoPET) is a molecular imaging modality combining the high sensitivity of PET with the specific targeting ability of monoclonal antibodies. Various radioimmunotracers have been successfully developed to target a broad spectrum of molecules expressed by malignant cells or tumor microenvironments. Only a few are translated into clinical studies and barely into clinical practices. Some drawbacks include slow radioimmunotracer kinetics, high physiologic uptake in lymphoid organs, and heterogeneous activity in tumoral lesions. Measures are taken to overcome the disadvantages, and new tracers are being developed. In this review, we aim to mention the fundamental components of immunoPET imaging, explore the groundbreaking success achieved using this new technique, and review different radioimmunotracers employed in various solid tumors to elaborate on this relatively new imaging modality.
Collapse
Affiliation(s)
- Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahar Ataeinia
- Department of Radiology, Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shaghayegh Ranjbar
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Zahra Jamshidi Araghi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mobin Moradi
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
38
|
Krutzek F, Kopka K, Stadlbauer S. Development of Radiotracers for Imaging of the PD-1/PD-L1 Axis. Pharmaceuticals (Basel) 2022; 15:ph15060747. [PMID: 35745666 PMCID: PMC9228425 DOI: 10.3390/ph15060747] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/24/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has emerged as a major treatment option for a variety of cancers. Among the immune checkpoints addressed, the programmed death receptor 1 (PD-1) and its ligand PD-L1 are the key targets for an ICI. PD-L1 has especially been proven to be a reproducible biomarker allowing for therapy decisions and monitoring therapy success. However, the expression of PD-L1 is not only heterogeneous among and within tumor lesions, but the expression is very dynamic and changes over time. Immunohistochemistry, which is the standard diagnostic tool, can only inadequately address these challenges. On the other hand, molecular imaging techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) provide the advantage of a whole-body scan and therefore fully address the issue of the heterogeneous expression of checkpoints over time. Here, we provide an overview of existing PET, SPECT, and optical imaging (OI) (radio)tracers for the imaging of the upregulation levels of PD-1 and PD-L1. We summarize the preclinical and clinical data of the different molecule classes of radiotracers and discuss their respective advantages and disadvantages. At the end, we show possible future directions for developing new radiotracers for the imaging of PD-1/PD-L1 status in cancer patients.
Collapse
Affiliation(s)
- Fabian Krutzek
- Department of Translational TME Ligands, Institute of Radiopharmaceutical Cancer Research, Helmholtz Center Dresden-Rossendorf, 01328 Dresden, Germany; (F.K.); (K.K.)
| | - Klaus Kopka
- Department of Translational TME Ligands, Institute of Radiopharmaceutical Cancer Research, Helmholtz Center Dresden-Rossendorf, 01328 Dresden, Germany; (F.K.); (K.K.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01069 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, University Cancer Cancer (UCC), 01307 Dresden, Germany
| | - Sven Stadlbauer
- Department of Translational TME Ligands, Institute of Radiopharmaceutical Cancer Research, Helmholtz Center Dresden-Rossendorf, 01328 Dresden, Germany; (F.K.); (K.K.)
- Correspondence:
| |
Collapse
|
39
|
van der Geest KSM, Sandovici M, Nienhuis PH, Slart RHJA, Heeringa P, Brouwer E, Jiemy WF. Novel PET Imaging of Inflammatory Targets and Cells for the Diagnosis and Monitoring of Giant Cell Arteritis and Polymyalgia Rheumatica. Front Med (Lausanne) 2022; 9:902155. [PMID: 35733858 PMCID: PMC9207253 DOI: 10.3389/fmed.2022.902155] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/13/2022] [Indexed: 12/26/2022] Open
Abstract
Giant cell arteritis (GCA) and polymyalgia rheumatica (PMR) are two interrelated inflammatory diseases affecting patients above 50 years of age. Patients with GCA suffer from granulomatous inflammation of medium- to large-sized arteries. This inflammation can lead to severe ischemic complications (e.g., irreversible vision loss and stroke) and aneurysm-related complications (such as aortic dissection). On the other hand, patients suffering from PMR present with proximal stiffness and pain due to inflammation of the shoulder and pelvic girdles. PMR is observed in 40-60% of patients with GCA, while up to 21% of patients suffering from PMR are also affected by GCA. Due to the risk of ischemic complications, GCA has to be promptly treated upon clinical suspicion. The treatment of both GCA and PMR still heavily relies on glucocorticoids (GCs), although novel targeted therapies are emerging. Imaging has a central position in the diagnosis of GCA and PMR. While [18F]fluorodeoxyglucose (FDG)-positron emission tomography (PET) has proven to be a valuable tool for diagnosis of GCA and PMR, it possesses major drawbacks such as unspecific uptake in cells with high glucose metabolism, high background activity in several non-target organs and a decrease of diagnostic accuracy already after a short course of GC treatment. In recent years, our understanding of the immunopathogenesis of GCA and, to some extent, PMR has advanced. In this review, we summarize the current knowledge on the cellular heterogeneity in the immunopathology of GCA/PMR and discuss how recent advances in specific tissue infiltrating leukocyte and stromal cell profiles may be exploited as a source of novel targets for imaging. Finally, we discuss prospective novel PET radiotracers that may be useful for the diagnosis and treatment monitoring in GCA and PMR.
Collapse
Affiliation(s)
- Kornelis S. M. van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Pieter H. Nienhuis
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Riemer H. J. A. Slart
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - William F. Jiemy
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
40
|
Drozd M, Duszczyk A, Ivanova P, Pietrzak M. Interactions of proteins with metal-based nanoparticles from a point of view of analytical chemistry - Challenges and opportunities. Adv Colloid Interface Sci 2022; 304:102656. [PMID: 35367856 DOI: 10.1016/j.cis.2022.102656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 11/01/2022]
Abstract
Interactions of proteins with nanomaterials draw attention of many research groups interested in fundamental phenomena. However, alongside with valuable information regarding physicochemical aspects of such processes and their mechanisms, they more and more often prove to be useful from a point of view of bioanalytics. Deliberate use of processes based on adsorption of proteins on nanoparticles (or vice versa) allows for a development of new analytical methods and improvement of the existing ones. It also leads to obtaining of nanoparticles of desired properties and functionalities, which can be used as elements of analytical tools for various applications. Due to interactions with nanoparticles, proteins can also gain new functionalities or lose their interfering potential, which from perspective of bioanalytics seems to be very inviting and attractive. In the framework of this article we will discuss the bioanalytical potential of interactions of proteins with a chosen group of nanoparticles, and implementation of so driven processes for biosensing. Moreover, we will show both positive and negative (opportunities and challenges) aspects resulting from the presence of proteins in media/samples containing metal-based nanoparticles or their precursors.
Collapse
|
41
|
Gong J, Zhao L, Yang J, Zhu M, Zhao J. [99mTc]Tc-Labeled Plectin-Targeting Peptide as a Novel SPECT Probe for Tumor Imaging. Pharmaceutics 2022; 14:pharmaceutics14050996. [PMID: 35631582 PMCID: PMC9146797 DOI: 10.3390/pharmaceutics14050996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
Certain receptors are often overexpressed during tumor occurrence and development and closely correlate with carcinogenesis. Owing to its overexpression on the cell membrane and cytoplasm of various tumors, plectin, which is involved in tumor proliferation, migration, and invasion, has been viewed as a promising target for cancer imaging. Hence, plectin-targeting agents have great potential as imaging probes for tumor diagnosis. In this study, we developed a [99mTc]Tc-labeled plectin-targeted peptide (PTP) as a novel single-photon emission computed tomography (SPECT) probe for tumor imaging and investigated its pharmacokinetics, biodistribution, and targeting ability in several types of tumor-bearing mouse models. The PTP had good biocompatibility and targeting ability to tumor cells in vitro and could be readily labeled with [99mTc]Tc after modification with the bifunctional chelator 6-hydrazino nicotinamide (HYNIC). Furthermore, the prepared [99mTc]Tc-labeled PTP ([99mTc]Tc-HYNIC-PTP) showed high radiochemical purity and excellent stability in vitro. In addition, favorable biodistribution, fast blood clearance, and clear accumulation of [99mTc]Tc-HYNIC-PTP in several types of tumors were observed, with a good correlation between tumor uptake and plectin expression levels. These results indicate the potential of [99mTc]Tc-HYNIC-PTP as a novel SPECT probe for tumor imaging.
Collapse
Affiliation(s)
- Jiali Gong
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.G.); (L.Z.)
| | - Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.G.); (L.Z.)
| | - Jiqin Yang
- Department of Nuclear Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
- Correspondence: (J.Y.); (M.Z.); (J.Z.); Tel.: +86-21-3779-8352 (J.Z.)
| | - Meilin Zhu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
- Correspondence: (J.Y.); (M.Z.); (J.Z.); Tel.: +86-21-3779-8352 (J.Z.)
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.G.); (L.Z.)
- Correspondence: (J.Y.); (M.Z.); (J.Z.); Tel.: +86-21-3779-8352 (J.Z.)
| |
Collapse
|
42
|
Van Fossen EM, Bednar RM, Jana S, Franklin R, Beckman J, Karplus PA, Mehl RA. Nanobody assemblies with fully flexible topology enabled by genetically encoded tetrazine amino acids. SCIENCE ADVANCES 2022; 8:eabm6909. [PMID: 35522749 PMCID: PMC9075797 DOI: 10.1126/sciadv.abm6909] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/23/2022] [Indexed: 06/14/2023]
Abstract
Assembling nanobodies (Nbs) into polyvalent multimers is a powerful strategy for improving the effectiveness of Nb-based therapeutics and biotechnological tools. However, generally effective approaches to Nb assembly are currently restricted to the amino or carboxyl termini, greatly limiting the diversity of Nb multimer topologies that can be produced. Here, we show that reactive tetrazine groups-site-specifically inserted by genetic code expansion at Nb surface sites-are compatible with Nb folding and function, enabling Nb assembly at any desired point. Using two anti-SARS-CoV-2 Nbs with viral neutralization ability, we created Nb homo- and heterodimers with improved properties compared with conventionally linked Nb homodimers, which, in the case of our tetrazine-conjugated trimer, translated into enhanced viral neutralization. Thus, this tetrazine-based approach is a generally applicable strategy that greatly increases the accessible range of Nb assembly topologies, and thereby adds the optimization of topology as an effective avenue to generate Nb assemblies with improved efficacy.
Collapse
Affiliation(s)
- Elise M. Van Fossen
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Riley M. Bednar
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Subhashis Jana
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Rachel Franklin
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Joseph Beckman
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
- e-MSion, Inc., 2121 NE Jack London Drive, Corvallis, OR 97330, USA
| | - P. Andrew Karplus
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Ryan A. Mehl
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
43
|
Gosmann D, Russelli L, Weber WA, Schwaiger M, Krackhardt AM, D'Alessandria C. Promise and challenges of clinical non-invasive T-cell tracking in the era of cancer immunotherapy. EJNMMI Res 2022; 12:5. [PMID: 35099641 PMCID: PMC8804060 DOI: 10.1186/s13550-022-00877-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
In the last decades, our understanding of the role of the immune system in cancer has significantly improved and led to the discovery of new immunotherapeutic targets and tools, which boosted the advances in cancer immunotherapy to fight a growing number of malignancies. Approved immunotherapeutic approaches are currently mainly based on immune checkpoint inhibitors, antibody-derived targeted therapies, or cell-based immunotherapies. In essence, these therapies induce or enhance the infiltration and function of tumor-reactive T cells within the tumors, ideally resulting in complete tumor eradication. While the clinical application of immunotherapies has shown great promise, these therapies are often accompanied either by a variety of side effects as well as partial or complete unresponsiveness of a number of patients. Since different stages of disease progression elicit different local and systemic immune responses, the ability to longitudinally interrogate the migration and expansion of immune cells, especially T cells, throughout the whole body might greatly facilitate disease characterization and understanding. Furthermore, it can serve as a tool to guide development as well as selection of appropriate treatment regiments. This review provides an overview about a variety of immune-imaging tools available to characterize and study T-cell responses induced by anti-cancer immunotherapy. Moreover, challenges are discussed that must be taken into account and overcome to use immune-imaging tools as predictive and surrogate markers to enhance assessment and successful application of immunotherapies.
Collapse
Affiliation(s)
- Dario Gosmann
- Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Lisa Russelli
- Klinik und Poliklinik für Nuklearmedizin, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Wolfgang A Weber
- Klinik und Poliklinik für Nuklearmedizin, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Markus Schwaiger
- Klinik und Poliklinik für Nuklearmedizin, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Angela M Krackhardt
- Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany. .,German Cancer Consortium (DKTK), Partner-Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Calogero D'Alessandria
- Klinik und Poliklinik für Nuklearmedizin, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| |
Collapse
|
44
|
Rafidi H, Rajan S, Urban K, Shatz-Binder W, Hui K, Ferl GZ, Kamath AV, Boswell CA. Effect of molecular size on interstitial pharmacokinetics and tissue catabolism of antibodies. MAbs 2022; 14:2085535. [PMID: 35867780 PMCID: PMC9311319 DOI: 10.1080/19420862.2022.2085535] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Advances in antibody engineering have enabled the construction of novel molecular formats in diverse shapes and sizes, providing new opportunities for biologic therapies and expanding the need to understand how various structural aspects affect their distribution properties. To assess the effect of antibody size on systemic pharmacokinetics (PK) and tissue distribution with or without neonatal Fc receptor (FcRn) binding, we evaluated a series of non-mouse-binding anti-glycoprotein D monoclonal antibody formats, including IgG [~150 kDa], one-armed IgG [~100 kDa], IgG-HAHQ (attenuated FcRn binding) [~150 kDa], F(ab')2 [~100 kDa], and F(ab) [~50 kDa]. Tissue-specific concentration-time profiles were corrected for blood content based on vascular volumes and normalized based on interstitial volumes to allow estimation of interstitial concentrations and interstitial:serum concentration ratios. Blood correction demonstrated that the contribution of circulating antibody on total uptake was greatest at early time points and for highly vascularized tissues. Tissue interstitial PK largely mirrored serum exposure profiles. Similar interstitial:serum ratios were obtained for the two FcRn-binding molecules, IgG and one-armed IgG, which reached pseudo-steady-state kinetics in most tissues. For non-FcRn-binding molecules, interstitial:serum ratios changed over time, suggesting that these molecules did not reach steady-state kinetics during the study. Furthermore, concentration-time profiles of both intact and catabolized molecule were measured by a dual tracer approach, enabling quantification of tissue catabolism and demonstrating that catabolism levels were highest for IgG-HAHQ. Overall, these data sets provide insight into factors affecting preclinical distribution and may be useful in estimating interstitial concentrations and/or catabolism in human tissues.
Collapse
Affiliation(s)
- Hanine Rafidi
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Sharmila Rajan
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Konnie Urban
- Safety Assessment, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Whitney Shatz-Binder
- Protein Chemistry, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Keliana Hui
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Gregory Z Ferl
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA.,Biomedical Imaging, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Amrita V Kamath
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - C Andrew Boswell
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA.,Biomedical Imaging, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
45
|
Madden PJ, Arif MS, Becker ME, McRaven MD, Carias AM, Lorenzo-Redondo R, Xiao S, Midkiff CC, Blair RV, Potter EL, Martin-Sancho L, Dodson A, Martinelli E, Todd JPM, Villinger FJ, Chanda SK, Aye PP, Roy CJ, Roederer M, Lewis MG, Veazey RS, Hope TJ. Development of an In Vivo Probe to Track SARS-CoV-2 Infection in Rhesus Macaques. Front Immunol 2021; 12:810047. [PMID: 35003140 PMCID: PMC8739270 DOI: 10.3389/fimmu.2021.810047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/10/2021] [Indexed: 01/02/2023] Open
Abstract
Infection with the novel coronavirus, SARS-CoV-2, results in pneumonia and other respiratory symptoms as well as pathologies at diverse anatomical sites. An outstanding question is whether these diverse pathologies are due to replication of the virus in these anatomical compartments and how and when the virus reaches those sites. To answer these outstanding questions and study the spatiotemporal dynamics of SARS-CoV-2 infection a method for tracking viral spread in vivo is needed. We developed a novel, fluorescently labeled, antibody-based in vivo probe system using the anti-spike monoclonal antibody CR3022 and demonstrated that it could successfully identify sites of SARS-CoV-2 infection in a rhesus macaque model of COVID-19. Our results showed that the fluorescent signal from our antibody-based probe could differentiate whole lungs of macaques infected for 9 days from those infected for 2 or 3 days. Additionally, the probe signal corroborated the frequency and density of infected cells in individual tissue blocks from infected macaques. These results provide proof of concept for the use of in vivo antibody-based probes to study SARS-CoV-2 infection dynamics in rhesus macaques.
Collapse
Affiliation(s)
- Patrick J. Madden
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Muhammad S. Arif
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Mark E. Becker
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Michael D. McRaven
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ann M. Carias
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Institute for Global Health, Chicago, IL, United States
| | - Sixia Xiao
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Cecily C. Midkiff
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Robert V. Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Elizabeth Lake Potter
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Laura Martin-Sancho
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | | | - Elena Martinelli
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - John-Paul M. Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Francois J. Villinger
- New Iberia Research Center, University of Louisiana-Lafayette, New Iberia, LA, United States
| | - Sumit K. Chanda
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Pyone Pyone Aye
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Chad J. Roy
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | - Ronald S. Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Thomas J. Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
46
|
Lin B, Du H, Fan J, Huang D, Gao F, Li J, Zhang Y, Feng G, Dai T, Du X. Radioimmunotherapy Combined With Low-Intensity Ultrasound and Microbubbles: A Potential Novel Strategy for Treatment of Solid Tumors. Front Oncol 2021; 11:750741. [PMID: 34745976 PMCID: PMC8570127 DOI: 10.3389/fonc.2021.750741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/06/2021] [Indexed: 11/17/2022] Open
Abstract
The prognosis of advanced malignant tumors is very poor, and effective treatment is limited. Radioimmunotherapy (RIT) is a novel treatment method. However, its anti-tumor effect is relatively low in solid tumors, which is mainly due to the blood-tumor barrier preventing RIT from penetrating the tumor, resulting in an insufficient dose. Low-intensity ultrasound with microbubbles (USMB) has proven capable of opening the blood-tumor barrier. The combination of the two technologies may overcome the poor anti-tumor effect of RIT and promote the clinical application of RIT in solid tumors. In this article, we reviewed the current research status of RIT in the treatment of solid tumors and the opportunities and challenges of USMB combined with RIT.
Collapse
Affiliation(s)
- Binwei Lin
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Huan Du
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jinjia Fan
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Dan Huang
- Radiology Department, Mianyang Central Hospital, Mianyang, China
| | - Feng Gao
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Jie Li
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Yu Zhang
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Gang Feng
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Tangzhi Dai
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Xiaobo Du
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| |
Collapse
|
47
|
Monoclonal antibody based radiopharmaceuticals for imaging and therapy. Curr Probl Cancer 2021; 45:100796. [PMID: 34657748 DOI: 10.1016/j.currproblcancer.2021.100796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/03/2021] [Accepted: 09/02/2021] [Indexed: 11/23/2022]
Abstract
The concept of personalized medicine has been steadily growing for the past decades. Monoclonal antibodies (mAbs) are undoubtedly playing an important role in the transition away from conventional medical practice to a more tailored approach to deliver the best therapy with the highest safety margin to a specific patient. In certain instances, mAbs and antibody drug conjugates (ADCs) may represent the preferred therapeutic option for several types of cancers due to their high specificity and affinity to the antigen. Monoclonal antibodies can be labeled with specific radionuclides well-suited for PET (Positron Emission Tomography) or gamma camera scintigraphy. The use of radiolabeled mAbs allows the interrogation of specific biomarkers and assessment of tumor heterogeneity in vivo by a single diagnostic imaging scan that includes the whole-body in the field-of-view. Moreover, the same mAb can then be radiolabeled with an analogous radionuclide for the delivery of beta-minus radiation or alpha-particles as part of a radioimmunotherapy (RIT) approach. However, the path to develop, validate, and implement mAb-based radiopharmaceuticals from bench-to-bedside is complex due to the extensive pre-clinical experiments and toxicological studies required, and the necessity of labor-intensive clinical trials that often require multi-time-point imaging and blood draws for internal radiation dosimetry and pharmacokinetics. As more mAb-based radiopharmaceuticals have been developed and evaluated, the opportunities and limitations offered by mAbs have become better defined. Our aim with this manuscript is therefore to provide an overview of the recent advances in the development of mAb-based radiopharmaceuticals and their clinical applications in Oncology.
Collapse
|
48
|
Hrynchak I, Santos L, Falcão A, Gomes CM, Abrunhosa AJ. Nanobody-Based Theranostic Agents for HER2-Positive Breast Cancer: Radiolabeling Strategies. Int J Mol Sci 2021; 22:ijms221910745. [PMID: 34639086 PMCID: PMC8509594 DOI: 10.3390/ijms221910745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023] Open
Abstract
The overexpression of human epidermal growth factor 2 (HER2) in breast cancer (BC) has been associated with a more aggressive tumor subtype, poorer prognosis and shorter overall survival. In this context, the development of HER2-targeted radiotracers is crucial to provide a non-invasive assessment of HER2 expression to select patients for HER2-targeted therapies, monitor response and identify those who become resistant. Antibodies represent ideal candidates for this purpose, as they provide high contrast images for diagnosis and low toxicity in the therapeutic setting. Of those, nanobodies (Nb) are of particular interest considering their favorable kinetics, crossing of relevant biological membranes and intratumoral distribution. The purpose of this review is to highlight the unique characteristics and advantages of Nb-based radiotracers in BC imaging and therapy. Additionally, radiolabeling methods for Nb including direct labeling, indirect labeling via prosthetic group and indirect labeling via complexation will be discussed, reporting advantages and drawbacks. Furthermore, the preclinical to clinical translation of radiolabeled Nbs as promising theranostic agents will be reported.
Collapse
Affiliation(s)
- Ivanna Hrynchak
- ICNAS-Produção Unipessoal, Lda.—University of Coimbra, 3000-548 Coimbra, Portugal; (I.H.); (L.S.)
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Liliana Santos
- ICNAS-Produção Unipessoal, Lda.—University of Coimbra, 3000-548 Coimbra, Portugal; (I.H.); (L.S.)
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Amílcar Falcão
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Célia M. Gomes
- iCBR—Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
- CACC—Clinical Academic Center of Coimbra, 3000-075 Coimbra, Portugal
| | - Antero J. Abrunhosa
- ICNAS-Produção Unipessoal, Lda.—University of Coimbra, 3000-548 Coimbra, Portugal; (I.H.); (L.S.)
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal;
- Correspondence:
| |
Collapse
|
49
|
Islam A, Pishesha N, Harmand TJ, Heston H, Woodham AW, Cheloha RW, Bousbaine D, Rashidian M, Ploegh HL. Converting an Anti-Mouse CD4 Monoclonal Antibody into an scFv Positron Emission Tomography Imaging Agent for Longitudinal Monitoring of CD4 + T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:1468-1477. [PMID: 34408009 PMCID: PMC8387391 DOI: 10.4049/jimmunol.2100274] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022]
Abstract
Immuno-positron emission tomography (PET), a noninvasive imaging modality, can provide a dynamic approach for longitudinal assessment of cell populations of interest. Transformation of mAbs into single-chain variable fragment (scFv)-based PET imaging agents would allow noninvasive tracking in vivo of a wide range of possible targets. We used sortase-mediated enzymatic labeling in combination with PEGylation to develop an anti-mouse CD4 scFv-based PET imaging agent constructed from an anti-mouse CD4 mAb. This anti-CD4 scFv can monitor the in vivo distribution of CD4+ T cells by immuno-PET. We tracked CD4+ and CD8+ T cells in wild-type mice, in immunodeficient recipients reconstituted with monoclonal populations of OT-II and OT-I T cells, and in a B16 melanoma model. Anti-CD4 and -CD8 immuno-PET showed that the persistence of both CD4+ and CD8+ T cells transferred into immunodeficient mice improved when recipients were immunized with OVA in CFA. In tumor-bearing animals, infiltration of both CD4+ and CD8+ T cells increased as the tumor grew. The approach described in this study should be readily applicable to convert clinically useful Abs into the corresponding scFv PET imaging agents.
Collapse
Affiliation(s)
- Ashraful Islam
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Novalia Pishesha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Society of Fellows, Harvard University, Cambridge, MA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Thibault J Harmand
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Hailey Heston
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Andrew W Woodham
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Ross W Cheloha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Djenet Bousbaine
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Microbiology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA
| | - Mohammad Rashidian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA; and
- Department of Radiology, Harvard Medical School, Boston, MA
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA;
- Department of Pediatrics, Harvard Medical School, Boston, MA
| |
Collapse
|
50
|
Küppers J, Kürpig S, Bundschuh RA, Essler M, Lütje S. Radiolabeling Strategies of Nanobodies for Imaging Applications. Diagnostics (Basel) 2021; 11:1530. [PMID: 34573872 PMCID: PMC8471529 DOI: 10.3390/diagnostics11091530] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/30/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
Nanobodies are small recombinant antigen-binding fragments derived from camelid heavy-chain only antibodies. Due to their compact structure, pharmacokinetics of nanobodies are favorable compared to full-size antibodies, allowing rapid accumulation to their targets after intravenous administration, while unbound molecules are quickly cleared from the circulation. In consequence, high signal-to-background ratios can be achieved, rendering radiolabeled nanobodies high-potential candidates for imaging applications in oncology, immunology and specific diseases, for instance in the cardiovascular system. In this review, a comprehensive overview of central aspects of nanobody functionalization and radiolabeling strategies is provided.
Collapse
Affiliation(s)
- Jim Küppers
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.K.); (R.A.B.); (M.E.); (S.L.)
| | | | | | | | | |
Collapse
|