1
|
Martinez-Orengo N, Shah S, Lai J, Basuli F, Lyndaker A, Turner ML, Peiravi M, Sourabh S, Sampson K, Zhang P, Swenson RE, Lusso P, Maldarelli F, Nath A, Lau CY, Hammoud DA. PET imaging of HIV-1 envelope protein gp120 using 18F-labeled nanobodies. iScience 2025; 28:111795. [PMID: 39917021 PMCID: PMC11800091 DOI: 10.1016/j.isci.2025.111795] [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: 08/02/2024] [Revised: 11/02/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Abstract
Radiolabeled antibodies against the HIV-1 envelope protein, gp120, have been previously tested in animal models and in people with HIV (PWH). Nanobodies offer advantages over antibodies, including smaller size and faster clearance, which allow labeling with fluorine-18. In this study, three nanobodies (J3, 3E3, B9) chosen based on their binding properties to the conserved CD4-binding site of gp120 were labeled with fluorine-18 and used for PET imaging in mice bearing wild-type (WT) and/or gp120-expressing (Env+) tumors. [18F]J3 and [18F]3E3 selectively targeted Env+ tumors and not WT tumors, with minimal background signal. Switching from non-site-specific radiolabeling method to sortase A-mediated site-specific conjugation at the C-terminus improved binding to Env+ tumors for all nanobodies. Site-specifically 18F-labeled J3 nanobody is the most promising candidate with the highest level of binding. These results establish an Env+ imaging method that will enable next stage testing in an HIV-1 preclinical infection model and potentially in PWH.
Collapse
Affiliation(s)
- Neysha Martinez-Orengo
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Swati Shah
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Jianhao Lai
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Rockville, MD, USA
| | - Anna Lyndaker
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Mitchell L. Turner
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Morteza Peiravi
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Suman Sourabh
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Kevon Sampson
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA
| | - Peng Zhang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Rolf E. Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Rockville, MD, USA
| | - Paolo Lusso
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
| | - Avindra Nath
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA
| | - Chuen-Yen Lau
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Srinivasula S, Kim I, Jang H, Degrange P, Brown H, Dalton V, Badralmaa Y, Natarajan V, Long B, Carrasquillo JA, Di Mascio M. Whole-body PET imaging of simian immunodeficiency virus using gp120-targeting probes fails to reveal regions of specific uptake in rhesus macaques. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07110-8. [PMID: 39888424 DOI: 10.1007/s00259-025-07110-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
PURPOSE Following the initial reports demonstrating the feasibility of immunoPET imaging of simian immunodeficiency virus (SIV) using gp120-targeting monoclonal antibodies in non-human primates, replication efforts of the imaging system in human immunodeficiency virus (HIV)-infected individuals have yielded conflicting results. Herein, we used two anti-gp120 antibodies, 7D3 and ITS103.01LS-F(ab')2, to interrogate the reproducibility of gp120-targeting probes for immunoPET imaging of SIV in rhesus macaques. METHODS The binding affinity estimates of 89Zr radiolabeled 7D3 and ITS103.01LS-F(ab')2 to SIV gp120, and the in-vitro and ex-vivo binding specificities of [89Zr]Zr-7D3 and [89Zr]Zr-ITS103.01LS-F(ab')2 to SIV Env expressing cells, primary cells, and tissue sections from uninfected and SIV-infected macaques were obtained through competition assays. The biodistributions of [89Zr]Zr-7D3 and [89Zr]Zr-ITS103.01LS-F(ab')2 were performed with static PET scans up to 6 days post-injection in 20 rhesus macaques and the standardized uptake values in various tissues were compared between SIV-infected and uninfected controls. RESULTS Despite the demonstrated nanomolar affinity of [89Zr]Zr-7D3 and [89Zr]Zr-ITS103.01LS-F(ab')2 to SIV gp120, and strong binding specificity to SIV gp120 cell lines, we observed no discernible differences in their binding in primary cells, tissue sections of secondary lymphoid organs, in-vivo probe uptake between SIV-infected and uninfected macaques, or ex-vivo validation necropsies. While the probes remained stable in-vivo, only [89Zr]Zr-ITS103.01LS-F(ab')2 in chronic plasma retained its binding specificity to SIV gp120, with [89Zr]Zr-7D3 experiencing a > 97% reduction in binding to gp120 due to competition from endogenous antibodies at the 7D3 binding site. CONCLUSION The overall absence of specific uptake suggests inadequate binding potential (ligand affinity x target molarity) for these probes to effectively image SIV or HIV in-vivo, warranting further investigation into the lack of reproducibility observed with earlier non-human primate SIV imaging and conflicting human studies.
Collapse
Affiliation(s)
- Sharat Srinivasula
- AIDS Imaging Research Section, Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Insook Kim
- AIDS Imaging Research Section, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Hyukjin Jang
- AIDS Imaging Research Section, Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Paula Degrange
- AIDS Imaging Research Section, Laulima Government Solutions, Integrated Research Facility, NIAID, NIH, Frederick, MD, USA
| | - Heather Brown
- AIDS Imaging Research Section, Laulima Government Solutions, Integrated Research Facility, NIAID, NIH, Frederick, MD, USA
| | - Viviana Dalton
- AIDS Imaging Research Section, Laulima Government Solutions, Integrated Research Facility, NIAID, NIH, Frederick, MD, USA
| | - Yunden Badralmaa
- Laboratory of Molecular Cell Biology, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ven Natarajan
- Laboratory of Molecular Cell Biology, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brad Long
- AIDS Imaging Research Section, Laulima Government Solutions, Integrated Research Facility, NIAID, NIH, Frederick, MD, USA
| | - Jorge A Carrasquillo
- Molecular Imaging Program, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Michele Di Mascio
- AIDS Imaging Research Section, Division of Clinical Research, NIAID, NIH, Poolesville, MD, USA.
| |
Collapse
|
3
|
Kelly B, Boudreau JE, Beyea S, Brewer K. Molecular imaging of viral pathogenesis and opportunities for the future. NPJ IMAGING 2025; 3:3. [PMID: 39872292 PMCID: PMC11761071 DOI: 10.1038/s44303-024-00056-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/24/2024] [Indexed: 01/30/2025]
Abstract
Molecular imaging is used in clinical and research settings. Since tools to study viral pathogenesis longitudinally and systemically are limited, molecular imaging is an attractive and largely unexplored tool. This review discusses molecular imaging probes and techniques for studying viruses, particularly those currently used in oncology that are applicable to virology. Expanding the repertoire of probes to better detect viral disease may make imaging even more valuable in (pre-)clinical settings.
Collapse
Affiliation(s)
- Brianna Kelly
- Biomedical MRI Research Laboratory (BMRL), IWK Health Centre, Halifax, NS Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS Canada
| | - Jeanette E. Boudreau
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS Canada
- Department of Pathology, Dalhousie University, Halifax, NS Canada
- Beatrice Hunter Cancer Research Institute (BHCRI), Halifax, NS Canada
| | - Steven Beyea
- IWK Health Centre, Halifax, NS Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS Canada
- Department of Physics & Atmospheric Science, Dalhousie University, Halifax, NS Canada
| | - Kimberly Brewer
- Biomedical MRI Research Laboratory (BMRL), IWK Health Centre, Halifax, NS Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS Canada
- IWK Health Centre, Halifax, NS Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS Canada
- Department of Physics & Atmospheric Science, Dalhousie University, Halifax, NS Canada
| |
Collapse
|
4
|
Hu K, O’Neil TR, Baharlou H, Austin PJ, Karrasch JF, Sarkawt L, Li Y, Bertram KM, Cunningham AL, Patrick E, Harman AN. The spatial biology of HIV infection. PLoS Pathog 2025; 21:e1012888. [PMID: 39854613 PMCID: PMC11760614 DOI: 10.1371/journal.ppat.1012888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025] Open
Abstract
HIV infection implicates a spectrum of tissues in the human body starting with viral transmission in the anogenital tract and subsequently persisting in lymphoid tissues and brain. Though studies using isolated cells have contributed significantly towards our understanding of HIV infection, the tissue microenvironment is characterised by a complex interplay of a range of factors, all of which can influence the course of infection but are otherwise missed in ex vivo studies. To address this knowledge gap, it is necessary to investigate the dynamics of infection and the host immune response in situ using imaging-based approaches. Over the last decade, emerging imaging techniques have continually redefined the limits of detection, both in terms of the scope and the scale of the targets. In doing so, this has opened up new questions that can be answered by in situ studies. This review discusses the high-dimensional imaging modalities that are now available and their application towards understanding the spatial biology of HIV infection.
Collapse
Affiliation(s)
- Kevin Hu
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Thomas R. O’Neil
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Heeva Baharlou
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul J. Austin
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, School of Medical of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Jackson F. Karrasch
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, School of Medical of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Lara Sarkawt
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Yuchen Li
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kirstie M. Bertram
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anthony L. Cunningham
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ellis Patrick
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew N. Harman
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
5
|
Lemos MP, Astronomo RD, Huang Y, Narpala S, Prabhakaran M, Mann P, Paez CA, Lu Y, Mize GJ, Glantz H, Westerberg K, Colegrove H, Smythe KS, Lin M, Pierce RH, Hutter J, Frank I, Mascola JR, McDermott AB, Bekker LG, McElrath MJ. Enhanced and sustained biodistribution of HIV-1 neutralizing antibody VRC01LS in human genital and rectal mucosa. Nat Commun 2024; 15:10332. [PMID: 39609400 PMCID: PMC11604655 DOI: 10.1038/s41467-024-54580-9] [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: 01/17/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
To prevent sexually-acquired HIV-1 infection by immunoprophylaxis, effective concentrations of broadly neutralizing antibodies are likely needed at mucosal sites of exposure. Here, we examine the biodistribution of monoclonal antibody VRC01 and its extended half-life variant, VRC01LS, in colorectal and genitourinary tracts of healthy adults 1-52 weeks after intravenous infusion. At 1-2 weeks, VRC01LS levels are ~3-4 times higher than VRC01 in serum (p = 0.048), rectal (p = 0.067), vaginal (p = 0.003) and cervical tissues (p = 0.003); these differences increase over time. Both antibodies primarily localize within rectal lamina propria and cervicovaginal stroma, with limited and variable epithelial distribution. Although 8-28% of serum mAb levels reach mucosal tissues, <3% are in seminal and rectal secretions. Elimination half-lives in mucosal tissues are 20-28 days for VRC01 and 51-68 days for VRC01LS. Thus, VRC01LS infusion achieves higher, sustained concentrations in human mucosal tissues than VRC01, supporting the future investigation of potent, long-acting LS-modified antibodies to prevent HIV-1.
Collapse
Affiliation(s)
- Maria P Lemos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Rena D Astronomo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Madhu Prabhakaran
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Philipp Mann
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Carmen A Paez
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Gregory J Mize
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hayley Glantz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Katharine Westerberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hunter Colegrove
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kimberly S Smythe
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Minggang Lin
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Robert H Pierce
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Julia Hutter
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ian Frank
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Linda-Gail Bekker
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Medicine, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
6
|
Nakamoto Y, Inui Y, Hotta M, Wakabayashi H, Hanaoka H. Recent advancements in new tracers from first-in-human studies. Ann Nucl Med 2024; 38:877-883. [PMID: 39325320 PMCID: PMC11489164 DOI: 10.1007/s12149-024-01979-5] [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: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/27/2024]
Abstract
Recent advancements in the development of positron emission tomography (PET) tracers have significantly enhanced our ability to image neuroinflammatory processes and neurotransmitter systems, which are vital for understanding and treating neurodegenerative and psychiatric disorders. Similarly, innovative tracers in oncology provide detailed images of the metabolic and molecular characteristics of tumors, which are crucial for tailoring targeted therapies and monitoring responses, including radiotherapy. Notable advancements include programmed death ligand 1 (PD-L1)-targeting agents for lung cancer, prostate-specific membrane antigen-based tracers for prostate cancer, chemokine receptor-targeting agents for hematological malignancies, human epidermal growth factor receptor 2 (HER2)-targeting tracers for various cancers, Claudin 18 based tracers for epithelial tumors, glutamine tracers for colorectal cancer, and ascorbic acid analogs for assessing cancer metabolism and therapy efficacy. Additionally, novel tracers have been developed for non-neurological and non-oncological applications, including adrenal imaging, amyloidosis, and human immunodeficiency virus (HIV) infection. This overview focuses on the newly developed tracers, particularly those used in neurology and oncology.
Collapse
Affiliation(s)
- Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| | - Yoshitaka Inui
- Department of Radiology, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
| | - Masatoshi Hotta
- Department of Radiology and Nuclear Medicine, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hirofumi Hanaoka
- Division of Fundamental Technology Development, Near InfraRed Photo-ImmunoTherapy Research Institute at Kansai Medical University, 2-5-1, Shin-Machi, Hirakata, 573-1010, Japan
| |
Collapse
|
7
|
Suliman S, Maison DP, Henrich TJ. The promise and reality of new immune profiling technologies. Nat Immunol 2024; 25:1765-1769. [PMID: 39242838 DOI: 10.1038/s41590-024-01948-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Affiliation(s)
- Sara Suliman
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - David P Maison
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Sun H, Huang Y, Hu D, Hong X, Salimi Y, Lv W, Chen H, Zaidi H, Wu H, Lu L. Artificial intelligence-based joint attenuation and scatter correction strategies for multi-tracer total-body PET. EJNMMI Phys 2024; 11:66. [PMID: 39028439 PMCID: PMC11264498 DOI: 10.1186/s40658-024-00666-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 07/04/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Low-dose ungated CT is commonly used for total-body PET attenuation and scatter correction (ASC). However, CT-based ASC (CT-ASC) is limited by radiation dose risks of CT examinations, propagation of CT-based artifacts and potential mismatches between PET and CT. We demonstrate the feasibility of direct ASC for multi-tracer total-body PET in the image domain. METHODS Clinical uEXPLORER total-body PET/CT datasets of [18F]FDG (N = 52), [18F]FAPI (N = 46) and [68Ga]FAPI (N = 60) were retrospectively enrolled in this study. We developed an improved 3D conditional generative adversarial network (cGAN) to directly estimate attenuation and scatter-corrected PET images from non-attenuation and scatter-corrected (NASC) PET images. The feasibility of the proposed 3D cGAN-based ASC was validated using four training strategies: (1) Paired 3D NASC and CT-ASC PET images from three tracers were pooled into one centralized server (CZ-ASC). (2) Paired 3D NASC and CT-ASC PET images from each tracer were individually used (DL-ASC). (3) Paired NASC and CT-ASC PET images from one tracer ([18F]FDG) were used to train the networks, while the other two tracers were used for testing without fine-tuning (NFT-ASC). (4) The pre-trained networks of (3) were fine-tuned with two other tracers individually (FT-ASC). We trained all networks in fivefold cross-validation. The performance of all ASC methods was evaluated by qualitative and quantitative metrics using CT-ASC as the reference. RESULTS CZ-ASC, DL-ASC and FT-ASC showed comparable visual quality with CT-ASC for all tracers. CZ-ASC and DL-ASC resulted in a normalized mean absolute error (NMAE) of 8.51 ± 7.32% versus 7.36 ± 6.77% (p < 0.05), outperforming NASC (p < 0.0001) in [18F]FDG dataset. CZ-ASC, FT-ASC and DL-ASC led to NMAE of 6.44 ± 7.02%, 6.55 ± 5.89%, and 7.25 ± 6.33% in [18F]FAPI dataset, and NMAE of 5.53 ± 3.99%, 5.60 ± 4.02%, and 5.68 ± 4.12% in [68Ga]FAPI dataset, respectively. CZ-ASC, FT-ASC and DL-ASC were superior to NASC (p < 0.0001) and NFT-ASC (p < 0.0001) in terms of NMAE results. CONCLUSIONS CZ-ASC, DL-ASC and FT-ASC demonstrated the feasibility of providing accurate and robust ASC for multi-tracer total-body PET, thereby reducing the radiation hazards to patients from redundant CT examinations. CZ-ASC and FT-ASC could outperform DL-ASC for cross-tracer total-body PET AC.
Collapse
Affiliation(s)
- Hao Sun
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, China
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva 4, Switzerland
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, China
| | - Yanchao Huang
- Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Debin Hu
- Department of Medical Engineering, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Xiaotong Hong
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, China
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva 4, Switzerland
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, China
| | - Yazdan Salimi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva 4, Switzerland
| | - Wenbing Lv
- Department of Electronic Engineering, Information School, Yunnan University, Kunming, 650091, China
| | - Hongwen Chen
- Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva 4, Switzerland
| | - Hubing Wu
- Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China.
| | - Lijun Lu
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, China.
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, China.
- Pazhou Lab, Guangzhou, 510330, China.
| |
Collapse
|
9
|
Mahomed S. Broadly neutralizing antibodies for HIV prevention: a comprehensive review and future perspectives. Clin Microbiol Rev 2024; 37:e0015222. [PMID: 38687039 PMCID: PMC11324036 DOI: 10.1128/cmr.00152-22] [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: 05/02/2024] Open
Abstract
SUMMARYThe human immunodeficiency virus (HIV) epidemic remains a formidable global health concern, with 39 million people living with the virus and 1.3 million new infections reported in 2022. Despite anti-retroviral therapy's effectiveness in pre-exposure prophylaxis, its global adoption is limited. Broadly neutralizing antibodies (bNAbs) offer an alternative strategy for HIV prevention through passive immunization. Historically, passive immunization has been efficacious in the treatment of various diseases ranging from oncology to infectious diseases. Early clinical trials suggest bNAbs are safe, tolerable, and capable of reducing HIV RNA levels. Although challenges such as bNAb resistance have been noted in phase I trials, ongoing research aims to assess the additive or synergistic benefits of combining multiple bNAbs. Researchers are exploring bispecific and trispecific antibodies, and fragment crystallizable region modifications to augment antibody efficacy and half-life. Moreover, the potential of other antibody isotypes like IgG3 and IgA is under investigation. While promising, the application of bNAbs faces economic and logistical barriers. High manufacturing costs, particularly in resource-limited settings, and logistical challenges like cold-chain requirements pose obstacles. Preliminary studies suggest cost-effectiveness, although this is contingent on various factors like efficacy and distribution. Technological advancements and strategic partnerships may mitigate some challenges, but issues like molecular aggregation remain. The World Health Organization has provided preferred product characteristics for bNAbs, focusing on optimizing their efficacy, safety, and accessibility. The integration of bNAbs in HIV prophylaxis necessitates a multi-faceted approach, considering economic, logistical, and scientific variables. This review comprehensively covers the historical context, current advancements, and future avenues of bNAbs in HIV prevention.
Collapse
Affiliation(s)
- Sharana Mahomed
- Centre for the AIDS
Programme of Research in South Africa (CAPRISA), Doris Duke Medical
Research Institute, Nelson R Mandela School of Medicine, University of
KwaZulu-Natal, Durban,
South Africa
| |
Collapse
|
10
|
García-Varela L, Codesido J, Perez-Pedrosa A, Muñoz-González M, Ramos-Docampo E, Rey-Bretal D, García-Otero X, Gómez-Lado N, Turrero A, Beiroa D, Rodríguez-Perez AI, Vidal A, Fernández-Ferreiro A, Pubul V, Aguiar P. Biodistribution and pharmacokinetics of [ 89Zr]-anti-VEGF mAbs using PET in glioblastoma rat models. Int J Pharm 2024; 652:123795. [PMID: 38224761 DOI: 10.1016/j.ijpharm.2024.123795] [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: 08/24/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/17/2024]
Abstract
INTRODUCTION Glioblastomas present intensive angiogenesis, thus anti-Vascular Endothelial Growth Factor (VEGF) antibodies (mAbs) have been proposed as promising therapies. However, the results of clinical trials reported moderate toxicity and limited effectiveness. This study evaluates the in vivo pharmacokinetics and biodistribution of these mAbs in a growing model of glioblastoma in rats using Positron Emission Tomography (PET). MATERIAL &Methods: mAbs were radiolabeled with zirconium-89. Four days after the model induction, animals were injected with 2.33 ± 1.3 MBq of [89Zr]-DFO-bevacizumab (n = 8) or 2.35 ± 0.26 MBq of [89Zr]-DFO-aflibercept (n = 6). PETs were performed at 0H, 48H, 168H, 240H, and 336H post-injection. Tumor induction was confirmed using [18F]-Fluorodeoxyglucose-PET and immunohistochemistry. Radiotracer uptake was estimated in all pre-defined Volumes-of-Interest. RESULTS Anti-VEGF mAbs showed 100 % Radiochemical-Purity. [89Zr]-DFO-bevacizumab showed a significantly higher bioavailability in whole-blood. A significant increase in the tumor uptake was detectable at 168H PET with [89Zr]-DFO-bevacizumab meanwhile with [89Zr]-DFO-aflibercept it was only detectable at 336H. [89Zr]-DFO-bevacizumab tumor uptake was significantly higher than that of [89Zr]-DFO-aflibercept in all the scans. Tumor induction was confirmed in all animal models. CONCLUSION MAbs detect VEGF-expression in glioblastoma models. Tumors were earlier targeted by Bevacizumab. The lower blood availability of aflibercept resulted in a lower tumor uptake than bevacizumab in all the scans.
Collapse
Affiliation(s)
- Lara García-Varela
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Jessica Codesido
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Pharmacy Dept & Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | | | - María Muñoz-González
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Emma Ramos-Docampo
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - David Rey-Bretal
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Xurxo García-Otero
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Noemí Gómez-Lado
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Angela Turrero
- Cell Cycle and Oncology Group (CiCLOn), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain
| | - Daniel Beiroa
- Centro de Biomedicina Experimental (CEBEGA), University of Santiago de Compostela, Spain
| | - Ana Isabel Rodríguez-Perez
- Cell and Molecular Neurobiology of Parkinson's disease, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain. Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Anxo Vidal
- Cell Cycle and Oncology Group (CiCLOn), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain
| | - Anxo Fernández-Ferreiro
- Pharmacy Dept & Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Virginia Pubul
- Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Pablo Aguiar
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| |
Collapse
|
11
|
Grasberger P, Sondrini AR, Clayton KL. Harnessing immune cells to eliminate HIV reservoirs. Curr Opin HIV AIDS 2024; 19:62-68. [PMID: 38167784 PMCID: PMC10908255 DOI: 10.1097/coh.0000000000000840] [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: 01/05/2024]
Abstract
PURPOSE OF REVIEW Despite decades of insights about how CD8 + T cells and natural killer (NK) cells contribute to natural control of infection, additional hurdles (mutational escape from cellular immunity, sequence diversity, and hard-to-access tissue reservoirs) will need to be overcome to develop a cure. In this review, we highlight recent findings of novel mechanisms of antiviral cellular immunity and discuss current strategies for therapeutic deisgn. RECENT FINDINGS Of note are the apparent converging roles of viral antigen-specific MHC-E-restricted CD8 + T cells and NK cells, interleukin (IL)-15 biologics to boost cytotoxicity, and broadly neutralizing antibodies in their native form or as anitbody fragments to neutralize virus and engage cellular immunity, respectively. Finally, renewed interest in myeloid cells as relevant viral reservoirs is an encouraging sign for designing inclusive therapeutic strategies. SUMMARY Several studies have shown promise in many preclinical models of disease, including simian immunodeficiency virus (SIV)/SHIV infection in nonhuman primates and HIV infection in humanized mice. However, each model comes with its own limitations and may not fully predict human responses. We eagerly await the results of clinical trails assessing the efficacy of these strategies to achieve reductions in viral reservoirs, delay viral rebound, or ultimately elicit immune based control of infection without combination antiretroviral therapy (cART).
Collapse
Affiliation(s)
- Paula Grasberger
- Department of Pathology, University of Massachusetts Chan Medical School
| | | | - Kiera L. Clayton
- Department of Pathology, University of Massachusetts Chan Medical School
| |
Collapse
|
12
|
Mohr P, van Sluis J, Lub-de Hooge MN, Lammertsma AA, Brouwers AH, Tsoumpas C. Advances and challenges in immunoPET methodology. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1360710. [PMID: 39355220 PMCID: PMC11440922 DOI: 10.3389/fnume.2024.1360710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/05/2024] [Indexed: 10/03/2024]
Abstract
Immuno-positron emission tomography (immunoPET) enables imaging of specific targets that play a role in targeted therapy and immunotherapy, such as antigens on cell membranes, targets in the disease microenvironment, or immune cells. The most common immunoPET applications use a monoclonal antibody labeled with a relatively long-lived positron emitter such as 89Zr (T 1/2 = 78.4 h), but smaller antibody-based constructs labeled with various other positron emitting radionuclides are also being investigated. This molecular imaging technique can thus guide the development of new drugs and may have a pivotal role in selecting patients for a particular therapy. In early phase immunoPET trials, multiple imaging time points are used to examine the time-dependent biodistribution and to determine the optimal imaging time point, which may be several days after tracer injection due to the slow kinetics of larger molecules. Once this has been established, usually only one static scan is performed and semi-quantitative values are reported. However, total PET uptake of a tracer is the sum of specific and nonspecific uptake. In addition, uptake may be affected by other factors such as perfusion, pre-/co-administration of the unlabeled molecule, and the treatment schedule. This article reviews imaging methodologies used in immunoPET studies and is divided into two parts. The first part summarizes the vast majority of clinical immunoPET studies applying semi-quantitative methodologies. The second part focuses on a handful of studies applying pharmacokinetic models and includes preclinical and simulation studies. Finally, the potential and challenges of immunoPET quantification methodologies are discussed within the context of the recent technological advancements provided by long axial field of view PET/CT scanners.
Collapse
Affiliation(s)
- Philipp Mohr
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
13
|
Nakamura T, Okumura M, Takamune N, Hirotsu T, Sugiura M, Yasunaga J, Nakata H. Conversion of raltegravir carrying a 1,3,4-oxadiazole ring to a hydrolysis product upon pH changes decreases its antiviral activity. PNAS NEXUS 2024; 3:pgad446. [PMID: 38170115 PMCID: PMC10758923 DOI: 10.1093/pnasnexus/pgad446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024]
Abstract
Raltegravir (RAL), a human immunodeficiency virus (HIV)-1 integrase inhibitor, has been administered as part of antiretroviral therapy. Studies in patients with HIV-1 have shown high variability in the pharmacokinetics of RAL, and in healthy volunteers, coadministration of proton-pump inhibitors has been shown to increase the plasma RAL concentrations. Here, we found that RAL containing a 1,3,4-oxadiazole ring is converted to a hydrolysis product (H-RAL) with a cleaved 1,3,4-oxadiazole ring at pH 1.0 and 13.0 conditions in vitro, thereby reducing the anti-HIV activity of the drug. The inclusion of cyclodextrins (beta-cyclodextrin [βCD], random methyl-βCD [RAM-βCD], and hydroxypropyl-βCD [HP-βCD]) can protect RAL from pH-induced changes. The conversion of RAL to H-RAL was detected by using various mass spectrometry analyses. The chromatogram of H-RAL increased in a time-dependent manner similar to another 1,3,4-oxadiazole-containing drug, zibotentan, using high-performance liquid chromatography. Oral bioavailability and target protein interactions of H-RAL were predicted to be lower than those of RAL. Moreover, H-RAL exhibited significantly reduced anti-HIV-1 activity, whereas combinations with βCD, RAM-βCD, and HP-βCD attenuated this effect in cell-based assays. These findings suggest that βCDs can potentially protect against the conversion of RAL to H-RAL under acidic conditions in the stomach, thereby preserving the anti-HIV-1 effect of RAL. Although clinical trials are needed for evaluation, we anticipate that protective devices such as βCDs may improve the pharmacokinetics of RAL, leading to better treatment outcomes, including reduced dosing, long-term anti-HIV-1 activity, and deeper HIV-1 suppression.
Collapse
Affiliation(s)
- Tomofumi Nakamura
- Department of Hematology, Rheumatology, and Infectious Diseases, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Honjyo 1-1-1, Chuo-ku, Kumamoto 860-8556, Japan
- Department of Laboratory Medicine, Kumamoto University Hospital, Honjyo 1-1-1, Chuo-ku, Kumamoto 860-8556, Japan
| | - Mayu Okumura
- Department of Hematology, Rheumatology, and Infectious Diseases, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Honjyo 1-1-1, Chuo-ku, Kumamoto 860-8556, Japan
| | - Nobutoki Takamune
- Kumamoto Innovative Development Organization, Kumamoto University, Kurokami 2-39-1, Chuo-ku, Kumamoto 860-0862, Japan
| | - Tatsunori Hirotsu
- CyDing Company Limited, Oehonmachi 5-1, Chuo-ku, Kumamoto 862-0973, Japan
| | - Masaharu Sugiura
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan
| | - Junichiro Yasunaga
- Department of Hematology, Rheumatology, and Infectious Diseases, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Honjyo 1-1-1, Chuo-ku, Kumamoto 860-8556, Japan
| | - Hirotomo Nakata
- Department of Hematology, Rheumatology, and Infectious Diseases, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Honjyo 1-1-1, Chuo-ku, Kumamoto 860-8556, Japan
| |
Collapse
|
14
|
Dadachova E, Rangel DEN. Recent Advancements in Radiopharmaceuticals for Infection Imaging. Methods Mol Biol 2024; 2813:205-217. [PMID: 38888780 DOI: 10.1007/978-1-0716-3890-3_14] [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: 06/20/2024]
Abstract
COVID-19 pandemic has heightened the interest toward diagnosis and treatment of infectious diseases. Nuclear medicine, with its powerful scintigraphic, single photon emission computer tomography (SPECT), and positron emission tomography (PET) imaging modalities, has always played an important role in diagnosis of infections and distinguishing them from the sterile inflammation. In addition to the clinically available radiopharmaceuticals, there has been a decades-long effort to develop more specific imaging agents with some examples being radiolabeled antibiotics and antimicrobial peptides for bacterial imaging, radiolabeled antifungals for fungal infections imaging, radiolabeled pathogen-specific antibodies, and molecular engineered constructs. In this chapter, we discuss some examples of the work published in the last decade on developing nuclear imaging agents for bacterial, fungal, and viral infections to generate more interest among nuclear medicine community toward conducting clinical trials of these novel probes, as well as toward developing novel radiotracers for imaging infections.
Collapse
Affiliation(s)
- Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada.
| | | |
Collapse
|
15
|
Cossarini F, Aberg JA, Chen BK, Mehandru S. Viral Persistence in the Gut-Associated Lymphoid Tissue and Barriers to HIV Cure. AIDS Res Hum Retroviruses 2023; 40:54-65. [PMID: 37450338 PMCID: PMC10790554 DOI: 10.1089/aid.2022.0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
More than 40 years after the first reported cases of what then became known as acquired immunodeficiency syndrome (AIDS), tremendous progress has been achieved in transforming the disease from almost universally fatal to a chronic manageable condition. Nonetheless, the efforts to find a preventative vaccine or a cure for the underlying infection with Human Immunodeficiency Virus (HIV) remain largely unsuccessful. Many challenges intrinsic to the virus characteristics and host response need to be overcome for either goal to be achieved. This article will review the obstacles to an effective HIV cure, specifically the steps involved in the generation of HIV latency, focusing on the role of the gut-associated lymphoid tissue, which has received less attention compared with the peripheral blood, despite being the largest repository of lymphoid tissue in the human body, and a large site for HIV persistence.
Collapse
Affiliation(s)
- Francesca Cossarini
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
| | - Judith A. Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
| | - Benjamin K. Chen
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
- Division of Gastroenterology, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
| |
Collapse
|
16
|
Sun W, Rassadkina Y, Gao C, Collens SI, Lian X, Solomon IH, Mukerji SS, Yu XG, Lichterfeld M. Persistence of intact HIV-1 proviruses in the brain during antiretroviral therapy. eLife 2023; 12:RP89837. [PMID: 37938115 PMCID: PMC10631759 DOI: 10.7554/elife.89837] [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: 11/09/2023] Open
Abstract
HIV-1 reservoir cells that circulate in peripheral blood during suppressive antiretroviral therapy (ART) have been well characterized, but little is known about the dissemination of HIV-1-infected cells across multiple anatomical tissues, especially the CNS. Here, we performed single-genome, near full-length HIV-1 next-generation sequencing to evaluate the proviral landscape in distinct anatomical compartments, including multiple CNS tissues, from 3 ART-treated participants at autopsy. While lymph nodes and, to a lesser extent, gastrointestinal and genitourinary tissues represented tissue hotspots for the persistence of intact proviruses, we also observed intact proviruses in CNS tissue sections, particularly in the basal ganglia. Multi-compartment dissemination of clonal intact and defective proviral sequences occurred across multiple anatomical tissues, including the CNS, and evidence for the clonal proliferation of HIV-1-infected cells was found in the basal ganglia, in the frontal lobe, in the thalamus and in periventricular white matter. Deep analysis of HIV-1 reservoirs in distinct tissues will be informative for advancing HIV-1 cure strategies.
Collapse
Affiliation(s)
- Weiwei Sun
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
| | | | - Ce Gao
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
| | | | - Xiaodong Lian
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
| | - Isaac H Solomon
- Department of Pathology, Brigham and Women’s HospitalBostonUnited States
| | - Shibani S Mukerji
- Department of Neurology, Massachusetts General HospitalBostonUnited States
| | - Xu G Yu
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- Infectious Disease Division, Brigham and Women’s HospitalBostonUnited States
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- Infectious Disease Division, Brigham and Women’s HospitalBostonUnited States
| |
Collapse
|
17
|
Hammoud DA, Clifford Lane H, Jain SK. Molecular Imaging of Infections: Advancing the Search for the Hidden Enemy. J Infect Dis 2023; 228:S233-S236. [PMID: 37788496 PMCID: PMC10547366 DOI: 10.1093/infdis/jiad079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023] Open
Abstract
Even before the coronavirus disease 2019 pandemic, infections were a major threat to human health, as the third leading cause of death and the leading cause of morbidity among all human diseases. Although conventional imaging studies are routinely used for patients with infections, they provide structural or anatomic information only. Molecular imaging technologies enable noninvasive visualization of molecular processes at the cellular level within intact living subjects, including patients, and hold great potential for infections. We hope that this supplement will spur interest in the field and establish new collaborations to develop and translate novel molecular imaging approaches to the clinic.
Collapse
Affiliation(s)
- Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, NIH Clinical Center, Bethesda, Maryland, USA
| | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Lau CY, Martinez-Orengo N, Lyndaker A, Flavahan K, Johnson RF, Shah S, Hammoud DA. Advances and Challenges in Molecular Imaging of Viral Infections. J Infect Dis 2023; 228:S270-S280. [PMID: 37788495 PMCID: PMC10547465 DOI: 10.1093/infdis/jiad247] [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: 10/05/2023] Open
Abstract
Molecular imaging of viral infection, using a variety of advanced imaging techniques such as optical and nuclear imaging, can and has been used for direct visualization of the virus as well as assessment of virus-host interactions. Unlike imaging of other pathogens such as bacteria and fungi, challenging aspects of imaging viral infections include the small size of viruses, the complexity of viral infection animal models (eg, species dependence), and the high-level containment needs for many high-consequence pathogens, among others. In this review, using representative viral infections, we discuss how molecular imaging can reveal real-time infection dynamics, improve our understanding of disease pathogenesis, and guide optimization of treatment and prevention strategies. Key findings from human and animal studies are highlighted.
Collapse
Affiliation(s)
- Chuen-Yen Lau
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Neysha Martinez-Orengo
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Anna Lyndaker
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Kelly Flavahan
- Center for Infection and Inflammation Imaging Research, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Reed F Johnson
- SARS-CoV-2 Virology Core, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Swati Shah
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Dima A Hammoud
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Shah S, Turner ML, Chen X, Ances BM, Hammoud DA, Tucker EW. The Promise of Molecular Imaging: Focus on Central Nervous System Infections. J Infect Dis 2023; 228:S311-S321. [PMID: 37788502 PMCID: PMC11009511 DOI: 10.1093/infdis/jiad223] [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: 10/05/2023] Open
Abstract
Central nervous system (CNS) infections can lead to high mortality and severe morbidity. Diagnosis, monitoring, and assessing response to therapy of CNS infections is particularly challenging with traditional tools, such as microbiology, due to the dangers associated with invasive CNS procedures (ie, biopsy or surgical resection) to obtain tissues. Molecular imaging techniques like positron emission tomography (PET) and single-photon emission computed tomography (SPECT) imaging have long been used to complement anatomic imaging such as computed tomography (CT) and magnetic resonance imaging (MRI), for in vivo evaluation of disease pathophysiology, progression, and treatment response. In this review, we detail the use of molecular imaging to delineate host-pathogen interactions, elucidate antimicrobial pharmacokinetics, and monitor treatment response. We also discuss the utility of pathogen-specific radiotracers to accurately diagnose CNS infections and strategies to develop radiotracers that would cross the blood-brain barrier.
Collapse
Affiliation(s)
- Swati Shah
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Mitchell L Turner
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Xueyi Chen
- Department of Pediatrics, Center for Infection and Inflammation Imaging Research, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Beau M Ances
- Department of Neurology, Washington University, St Louis, Missouri, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth W Tucker
- Department of Anesthesiology and Critical Care Medicine, Center for Infection and Inflammation Imaging Research, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
20
|
Kleynhans J, Sathekge MM, Ebenhan T. Preclinical Research Highlighting Contemporary Targeting Mechanisms of Radiolabelled Compounds for PET Based Infection Imaging. Semin Nucl Med 2023; 53:630-643. [PMID: 37012169 DOI: 10.1053/j.semnuclmed.2023.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 04/04/2023]
Abstract
It is important to constantly monitor developments in the preclinical imaging arena of infection. Firstly, novel radiopharmaceuticals with the correct characteristics must be identified to funnel into the clinic. Secondly, it must be evaluated if enough innovative research is being done and adequate resources are geared towards the development of radiopharmaceuticals that could feed into the Nuclear Medicine Clinic in the near future. It is proposed that the ideal infection imaging agent will involve PET combined with CT but more ideally MRI. The radiopharmaceuticals currently presented in preclinical literature have a wide selection of vectors and targets. Ionic formulations of PET-radionuclides such 64CuCl2 and 68GaCl2 are evaluated for bacterial infection imaging. Many small molecule based radiopharmaceuticals are being investigated with the most prominent targets being cell wall synthesis, maltodextrin transport (such as [18F]F-maltotriose), siderophores (bacterial and fungal infections), the folate synthesis pathway (such as [18F]F-PABA) and protein synthesis (radiolabelled puromycin). Mycobacterial specific antibiotics, antifungals and antiviral agents are also under investigation as infection imaging agents. Peptide based radiopharmaceuticals are developed for bacterial, fungal and viral infections. The radiopharmaceutical development could even react quickly enough on a pandemic to develop a SARS-CoV-2 imaging agent in a timely fashion ([64Cu]Cu-NOTA-EK1). New immuno-PET agents for the imaging of viruses have recently been published, specifically for HIV persistence but also for SARS-CoV2. A very promising antifungal immuno-PET agent (hJ5F) is also considered. Future technologies could include the application of aptamers and bacteriophages and even going as far as the design of theranostic infection. Another possibility would be the application of nanobodies for immuno-PET applications. Standardization and optimization of the preclinical evaluation of radiopharmaceuticals could enhance clinical translation and reduce time spent in pursuing less than optimal candidates.
Collapse
Affiliation(s)
- Janke Kleynhans
- Department of Pharmaceutical and Pharmacological sciences, Radiopharmaceutical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Mike Machaba Sathekge
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa; Preclinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pretoria, South Africa
| | - Thomas Ebenhan
- Preclinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pretoria, South Africa; Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
21
|
Sun W, Rassadkina Y, Gao C, Collens SI, Lian X, Solomon IH, Mukerji S, Yu XG, Lichterfeld M. Persistence of intact HIV-1 proviruses in the brain during antiretroviral therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546135. [PMID: 37425847 PMCID: PMC10327102 DOI: 10.1101/2023.06.26.546135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
HIV-1 reservoir cells that circulate in peripheral blood during suppressive antiretroviral therapy (ART) have been well characterized, but little is known about the dissemination of HIV-1-infected cells across multiple anatomical tissues, especially the central nervous system (CNS). Here, we performed single-genome, near full-length HIV-1 next-generation sequencing to evaluate the proviral landscape in distinct anatomical compartments, including multiple CNS tissues, from 3 ART-treated participants at autopsy. While lymph nodes and, to a lesser extent, gastrointestinal and genitourinary tissues represented tissue hotspots for the persistence of intact proviruses, we also observed intact proviruses in CNS tissue sections, particularly in the basal ganglia. Multi-compartment dissemination of clonal intact and defective proviral sequences occurred across multiple anatomical tissues, including the CNS, and evidence for the clonal proliferation of HIV-1-infected cells was found in the basal ganglia, in the frontal lobe, in the thalamus and in periventricular white matter. Deep analysis of HIV-1 reservoirs in distinct tissues will be informative for advancing HIV-1 cure strategies.
Collapse
Affiliation(s)
- Weiwei Sun
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA
| | | | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA
| | | | - Xiaodong Lian
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA
| | - Isaac H. Solomon
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Shibani Mukerji
- Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
22
|
Buck AM, Deveau TM, Henrich TJ, Deitchman AN. Challenges in HIV-1 Latent Reservoir and Target Cell Quantification in CAR-T Cell and Other Lentiviral Gene Modifying HIV Cure Strategies. Viruses 2023; 15:1126. [PMID: 37243212 PMCID: PMC10222761 DOI: 10.3390/v15051126] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Gene-modification therapies are at the forefront of HIV-1 cure strategies. Chimeric antigen receptor (CAR)-T cells pose a potential approach to target infected cells during antiretroviral therapy or following analytical treatment interruption (ATI). However, there are technical challenges in the quantification of HIV-1-infected and CAR-T cells in the setting of lentiviral CAR gene delivery and also in the identification of cells expressing target antigens. First, there is a lack of validated techniques to identify and characterize cells expressing the hypervariable HIV gp120 in both ART-suppressed and viremic individuals. Second, close sequence homology between lentiviral-based CAR-T gene modification vectors and conserved regions of HIV-1 creates quantification challenges of HIV-1 and lentiviral vector levels. Consideration needs to be taken into standardizing HIV-1 DNA/RNA assays in the setting of CAR-T cell and other lentiviral vector-based therapies to avoid these confounding interactions. Lastly, with the introduction of HIV-1 resistance genes in CAR-T cells, there is a need for assays with single-cell resolution to determine the competence of the gene inserts to prevent CAR-T cells from becoming infected in vivo. As novel therapies continue to arise in the HIV-1 cure field, resolving these challenges in CAR-T-cell therapy will be crucial.
Collapse
Affiliation(s)
- Amanda M. Buck
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Tyler-Marie Deveau
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Timothy J. Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Amelia N. Deitchman
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
23
|
Carvalho JLC, Dadachova E. Radioimmunotherapy for the treatment of infectious diseases: a comprehensive update. Expert Rev Anti Infect Ther 2023; 21:365-374. [PMID: 36815406 DOI: 10.1080/14787210.2023.2184345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
INTRODUCTION Corona Virus Disease of 2019 (COVID-19) pandemic has renewed interest in monoclonal antibodies for treating infectious diseases. During last two decades experimental data has been accumulated showing the potential of radioimmunotherapy (RIT) of infectious diseases. In addition, COVID-19 pandemic has created a novel landscape for opportunistic fungal infections in post-COVID-19 patients resulting from severe immune suppression. AREAS COVERED We analyze recent results on targeting "pan-antigens" shared by fungal pathogens in mouse models and in healthy dogs; on developing RIT of prosthetic joint infections (PJI); examine RIT as potential human immunodeficiency virus (HIV) cure strategy and analyze its mechanisms and safety. Literature review was performed using PubMed and Google Scholar and includes relevant articles from 2000 to 2022. EXPERT OPINION Some of the RIT of infection applications can, hopefully, be moved into the clinic earlier than others after preclinical development: (1) RIT of opportunistic fungal infections might contribute to saving lives as current antifungal drugs do not work in severely immunocompromised patients; (2) RIT of patients with PJI. Success of RIT in these patients will allow to expand the application of RIT to other similarly vulnerable patients' populations such as cancer patients with weakened immune system and organ transplant recipients.
Collapse
|
24
|
Lau CY, Mostafa MYA. Editorial: Reviews in: Radiopharmaceuticals in nuclear medicine. Front Med (Lausanne) 2023; 10:1178528. [PMID: 37007772 PMCID: PMC10061581 DOI: 10.3389/fmed.2023.1178528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Affiliation(s)
- Chuen-Yen Lau
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Chuen-Yen Lau
| | | |
Collapse
|
25
|
van Dijk B, Hooning van Duyvenbode JFF, de Vor L, Nurmohamed FRHA, Lam MGEH, Poot AJ, Ramakers RM, Koustoulidou S, Beekman FJ, van Strijp J, Rooijakkers SHM, Dadachova E, Vogely HC, Weinans H, van der Wal BCH. Evaluating the Targeting of a Staphylococcus-aureus-Infected Implant with a Radiolabeled Antibody In Vivo. Int J Mol Sci 2023; 24:ijms24054374. [PMID: 36901805 PMCID: PMC10002501 DOI: 10.3390/ijms24054374] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/06/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Implant infections caused by Staphylococcus aureus are difficult to treat due to biofilm formation, which complicates surgical and antibiotic treatment. We introduce an alternative approach using monoclonal antibodies (mAbs) targeting S. aureus and provide evidence of the specificity and biodistribution of S.-aureus-targeting antibodies in a mouse implant infection model. The monoclonal antibody 4497-IgG1 targeting wall teichoic acid in S. aureus was labeled with indium-111 using CHX-A"-DTPA as a chelator. Single Photon Emission Computed Tomography/computed tomographyscans were performed at 24, 72 and 120 h after administration of the 111In-4497 mAb in Balb/cAnNCrl mice with a subcutaneous implant that was pre-colonized with S. aureus biofilm. The biodistribution of this labelled antibody over various organs was visualized and quantified using SPECT/CT imaging, and was compared to the uptake at the target tissue with the implanted infection. Uptake of the 111In-4497 mAbs at the infected implant gradually increased from 8.34 %ID/cm3 at 24 h to 9.22 %ID/cm3 at 120 h. Uptake at the heart/blood pool decreased over time from 11.60 to 7.58 %ID/cm3, whereas the uptake in the other organs decreased from 7.26 to less than 4.66 %ID/cm3 at 120 h. The effective half-life of 111In-4497 mAbs was determined to be 59 h. In conclusion, 111In-4497 mAbs were found to specifically detect S. aureus and its biofilm with excellent and prolonged accumulation at the site of the colonized implant. Therefore, it has the potential to serve as a drug delivery system for the diagnostic and bactericidal treatment of biofilm.
Collapse
Affiliation(s)
- Bruce van Dijk
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Correspondence: ; Tel.: +31-88-75-569-71
| | | | - Lisanne de Vor
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | | | - Marnix G. E. H. Lam
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Alex J. Poot
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ruud M. Ramakers
- MILabs B.V., 3584 CX Utrecht, The Netherlands
- Department of Radiation Science and Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Sofia Koustoulidou
- MILabs B.V., 3584 CX Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Freek J. Beekman
- MILabs B.V., 3584 CX Utrecht, The Netherlands
- Department of Radiation Science and Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Jos van Strijp
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Suzan H. M. Rooijakkers
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - H. Charles Vogely
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of BioMechanical Engineering, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Bart C. H. van der Wal
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
26
|
Andre M, Nair M, Raymond AD. HIV Latency and Nanomedicine Strategies for Anti-HIV Treatment and Eradication. Biomedicines 2023; 11:biomedicines11020617. [PMID: 36831153 PMCID: PMC9953021 DOI: 10.3390/biomedicines11020617] [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: 01/04/2023] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/22/2023] Open
Abstract
Antiretrovirals (ARVs) reduce Human Immunodeficiency Virus (HIV) loads to undetectable levels in infected patients. However, HIV can persist throughout the body in cellular reservoirs partly due to the inability of some ARVs to cross anatomical barriers and the capacity of HIV-1 to establish latent infection in resting CD4+ T cells and monocytes/macrophages. A cure for HIV is not likely unless latency is addressed and delivery of ARVs to cellular reservoir sites is improved. Nanomedicine has been used in ARV formulations to improve delivery and efficacy. More specifically, researchers are exploring the benefit of using nanoparticles to improve ARVs and nanomedicine in HIV eradication strategies such as shock and kill, block and lock, and others. This review will focus on mechanisms of HIV-1 latency and nanomedicine-based approaches to treat HIV.
Collapse
Affiliation(s)
- Mickensone Andre
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Madhavan Nair
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Andrea D. Raymond
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Correspondence: ; Tel.: +1-305-348-6430
| |
Collapse
|
27
|
Herschhorn A. Indirect Mechanisms of HIV-1 Evasion from Broadly Neutralizing Antibodies In Vivo. ACS Infect Dis 2023; 9:5-8. [PMID: 36512322 DOI: 10.1021/acsinfecdis.2c00573] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Alon Herschhorn
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, United States.,Institute for Molecular Virology, University of Minnesota, Minneapolis, Minnesota 55455, United States.,Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, Minnesota 55455, United States.,The College of Veterinary Medicine Graduate Program, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
28
|
Lau CY, Adan MA, Earhart J, Seamon C, Nguyen T, Savramis A, Adams L, Zipparo ME, Madeen E, Huik K, Grossman Z, Chimukangara B, Wulan WN, Millo C, Nath A, Smith BR, Ortega-Villa AM, Proschan M, Wood BJ, Hammoud DA, Maldarelli F. Imaging and biopsy of HIV-infected individuals undergoing analytic treatment interruption. Front Med (Lausanne) 2022; 9:979756. [PMID: 36072945 PMCID: PMC9441850 DOI: 10.3389/fmed.2022.979756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background HIV persistence during antiretroviral therapy (ART) is the principal obstacle to cure. Lymphoid tissue is a compartment for HIV, but mechanisms of persistence during ART and viral rebound when ART is interrupted are inadequately understood. Metabolic activity in lymphoid tissue of patients on long-term ART is relatively low, and increases when ART is stopped. Increases in metabolic activity can be detected by 18F-fluorodeoxyglucose Positron Emission Tomography (FDG-PET) and may represent sites of HIV replication or immune activation in response to HIV replication. Methods FDG-PET imaging will be used to identify areas of high and low metabolic uptake in lymphoid tissue of individuals undergoing long-term ART. Baseline tissue samples will be collected. Participants will then be randomized 1:1 to continue or interrupt ART via analytic treatment interruption (ATI). Image-guided biopsy will be repeated 10 days after ATI initiation. After ART restart criteria are met, image-guided biopsy will be repeated once viral suppression is re-achieved. Participants who continued ART will have a second FDG-PET and biopsies 12–16 weeks after the first. Genetic characteristics of HIV populations in areas of high and low FDG uptake will be assesed. Optional assessments of non-lymphoid anatomic compartments may be performed to evaluate HIV populations in distinct anatomic compartments. Anticipated results We anticipate that PET standardized uptake values (SUV) will correlate with HIV viral RNA in biopsies of those regions and that lymph nodes with high SUV will have more viral RNA than those with low SUV within a patient. Individuals who undergo ATI are expected to have diverse viral populations upon viral rebound in lymphoid tissue. HIV populations in tissues may initially be phylogenetically diverse after ATI, with emergence of dominant viral species (clone) over time in plasma. Dominant viral species may represent the same HIV population seen before ATI. Discussion This study will allow us to explore utility of PET for identification of HIV infected cells and determine whether high FDG uptake respresents areas of HIV replication, immune activation or both. We will also characterize HIV infected cell populations in different anatomic locations. The protocol will represent a platform to investigate persistence and agents that may target HIV populations. Study protocol registration Identifier: NCT05419024.
Collapse
Affiliation(s)
- Chuen-Yen Lau
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
- *Correspondence: Chuen-Yen Lau
| | - Matthew A. Adan
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Jessica Earhart
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Cassie Seamon
- Critical Care Medicine Department, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Thuy Nguyen
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Ariana Savramis
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Lindsey Adams
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Mary-Elizabeth Zipparo
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Erin Madeen
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Kristi Huik
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Zehava Grossman
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Benjamin Chimukangara
- Critical Care Medicine Department, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Wahyu Nawang Wulan
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Corina Millo
- PET Department, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Avindra Nath
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Bryan R. Smith
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Ana M. Ortega-Villa
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Michael Proschan
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Bradford J. Wood
- Interventional Radiology, Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Dima A. Hammoud
- Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| |
Collapse
|