1
|
Santana JG, Shewarega A, Nam D, Duncan J, Madoff DC, Hyder F, Coman D, Chapiro J. Molecular MRI of T-cell immune response to cryoablation in immunologically hot vs. cold hepatocellular carcinoma. JHEP Rep 2025; 7:101294. [PMID: 40028344 PMCID: PMC11870164 DOI: 10.1016/j.jhepr.2024.101294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 03/05/2025] Open
Abstract
Background & Aims Increasing enthusiasm around integrating locoregional therapy with systemic immunotherapy in primary liver cancer underscores the need for non-invasive imaging biomarkers. In this study, we aimed to establish advanced molecular MRI tools for monitoring T-cell responses to cryoablation in murine models, distinguishing between immunologically "hot" and "cold" hepatocellular carcinoma (HCC). Methods Immunocompetent 7-10-week-old C57BL/6J and BALB/cJ mice (n = 18 each) received carbon tetrachloride for 12 weeks to induce cirrhosis. Intrinsically immunogenic Hepa1-6 ("hot") and non-immunogenic TiB75 ("cold") cells were orthotopically implanted into C57BL/6 or BALB/c mice, respectively, to generate focal HCC lesions. After one week, animals were randomly assigned to (A) partial cryoablation (pCryo) (1.2 mm cryoprobe, -40 °C) or (B) no treatment (n = 8 per group and tumor type). Gadolinium 160 (160Gd)-labeled CD8+ antibody was administered intravenously either 1 week after tumor induction (control) or 1-week post (pCryo) (treatment). T1-weighted MRI scans were performed using a 9.4 T MRI scanner. Radiological-pathological correlation included imaging mass cytometry and immunohistochemistry. Results pCryo-treated Hepa1-6 tumors displayed peritumoral ring enhancement on T1-weighted MRI with 160Gd-CD8, correlating with imaging mass cytometry signal patterns. Untreated Hepa1-6 tumors lacked such enhancement. Radiological-pathological correlation confirmed significantly increased tumor-infiltrating CD8+ T lymphocytes in pCryo Hepa1-6 tumors compared with untreated tumors (p <0.001), and a stronger local response compared with systemic lymph nodes (p = 0.0415). Increased T-lymphocyte infiltration was not observed in TiB75 tumors, as indicated by MRI and histopathology. Conclusion pCryo induced increased T-cell infiltration in Hepa1-6 tumors compared to TiB75 tumors. T1-weighted MRI, following 160Gd-CD8 antibody administration, reproducibly detected the ablation-induced changes. These findings encourage further investigation of MRI-based molecular imaging biomarkers to assess immune responses to local tumor therapies. Impact and implications This study successfully established reliable MR-based molecular imaging tools to visualize CD8+ anti-tumor specific T-cell infiltration following partial cryoablation (pCryo) in murine tumor models. The study's significance lies in advancing our understanding of immune responses within induced cirrhosis and distinguishing between "hot" and "cold" tumor phenotypes. The findings not only build upon previous proof-of-principle data but also extend this technology to include different immune cell types in hepatocellular carcinoma. The study reveals that pCryo may exert specific effects on the tumor microenvironment, augmenting the anti-tumor immune response in immunogenic tumors while displaying a weaker local effect in non-immunogenic tumors.
Collapse
Affiliation(s)
- Jessica Gois Santana
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Annabella Shewarega
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, 10117 Berlin, Germany
| | - David Nam
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - James Duncan
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - David Craig Madoff
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
- Department of Medicine, Section of Medical Oncology, 333 Cedar Street, New Haven, CT 06510 United States
- Department of Surgery, Section of Surgical Oncology, 333 Cedar Street, New Haven, CT 06510, United States
| | - Fahmeed Hyder
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - Daniel Coman
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - Julius Chapiro
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
2
|
Zwanenburg A, Price G, Löck S. Artificial intelligence for response prediction and personalisation in radiation oncology. Strahlenther Onkol 2025; 201:266-273. [PMID: 39212687 PMCID: PMC11839704 DOI: 10.1007/s00066-024-02281-z] [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: 05/10/2024] [Accepted: 07/14/2024] [Indexed: 09/04/2024]
Abstract
Artificial intelligence (AI) systems may personalise radiotherapy by assessing complex and multifaceted patient data and predicting tumour and normal tissue responses to radiotherapy. Here we describe three distinct generations of AI systems, namely personalised radiotherapy based on pretreatment data, response-driven radiotherapy and dynamically optimised radiotherapy. Finally, we discuss the main challenges in clinical translation of AI systems for radiotherapy personalisation.
Collapse
Affiliation(s)
- Alex Zwanenburg
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstr. 74, PF 41, 01307, Dresden, Germany.
- National Center for Tumor Diseases Dresden (NCT/UCC), Germany:, German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany.
| | - Gareth Price
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| | - Steffen Löck
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstr. 74, PF 41, 01307, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| |
Collapse
|
3
|
Ning YD, Song YX, He YQ, Li H, Liu SY. Discordant Responses Between Imaging Examination and Surgical Pathology of Head and Heck Squamous Cell Carcinoma After Neoadjuvant Immunotherapy Combined With Chemotherapy. World J Oncol 2025; 16:59-69. [PMID: 39850520 PMCID: PMC11750755 DOI: 10.14740/wjon1973] [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: 09/18/2024] [Accepted: 11/21/2024] [Indexed: 01/25/2025] Open
Abstract
Background We here investigated the value of imaging examination in evaluating tumor remission-based surgery in patients with head and neck squamous cell carcinoma (HNSCC), who had undergone neoadjuvant immunotherapy combined with chemotherapy (NICC). Methods HNSCC patients who underwent NICC and surgery from May 2021 to September 2023 were retrospectively analyzed. All patients had to undergo imaging examination evaluation, including enhanced computed tomography (CT) and enhanced magnetic resonance (MR) imaging before and after NICC. Data related to clinical parameters, complete response of the primary site (PrCR), complete response of the primary site and the lymph node (PLCR), complete response of the lymph node (LCR), and tumor response (TR), were gathered. The paired Chi-square test and t-test were conducted to analyze the differences in responses between imaging examination and pathology. Binary logistic regression was applied to analyze the relevant clinical factors of differences in responses. Results In total, data of 41 patients were included in this study. Significant discordant responses were observed between enhanced CT, magnetic resonance imaging (MRI), and pathology in PrCR (4.9%, 7.3% vs. 41.5%), LCR (12.2%, 7.3% vs. 53.7%), PLCR (0%, 0% vs. 31.7%), and TR (severe 29.3%,17.1% vs. 25.61%) (P < 0.05). Patients with hypopharyngeal cancer (odds ratio (OR): 7.04), oral cancer (OR: 3.64), higher neutrophil to lymphocyte ratio (NLR) (OR: 2.05), and earlier T stage (OR: 0.71) exhibited a larger response difference between enhanced CT and pathology. Patients with younger age (OR: 0.79) hypopharyngeal cancer (OR: 22.81), oral cancer (OR: 2.65), higher NLR (OR: 19.47), and earlier T stage (OR: 0.29) exhibited a larger response difference between enhanced MR and pathology. Conclusions Discordant responses were noted between the imaging examination and surgical pathology of HNSCC after NICC. Hypopharyngeal cancer, higher NLR, and earlier T stage may predict a higher response difference.
Collapse
Affiliation(s)
- Yu Dong Ning
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- These authors contributed equally to this study
| | - Yi Xuan Song
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- These authors contributed equally to this study
| | - Yu Qin He
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Li
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shao Yan Liu
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Yamamoto K, Torigoe S, Tsujimura Y, Asaka MN, Okumura K, Ato M. In vivo imaging identified efficient antimicrobial treatment against Mycobacterium marinum infection in mouse footpads. Sci Rep 2024; 14:24343. [PMID: 39420066 PMCID: PMC11487254 DOI: 10.1038/s41598-024-75207-5] [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: 05/20/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Mycobacterium marinum (M. marinum) is the most common causative bacteria of cutaneous non-tuberculous mycobacterial (NTM) infections, including fish tank granuloma. Treating M. marinum-caused infection takes longer than other NTM diseases because M. marinum is less susceptible to antimicrobial agents. A standard treatment regimen for M. marinum infection has not been established yet, and few in vivo experiments have been performed in mammals to evaluate the bactericidal effects of antimicrobials. In this study, we developed a noninvasive in vivo imaging method to assess the therapeutic efficacy of antimicrobials against M. marinum infection. The data obtained using fluorescent protein or bioluminescence from luciferase will offer valuable insights into bacteria visualization across various bacterial infections. Furthermore, through this imaging technique, we demonstrated that combining clarithromycin, rifampicin, ethambutol, and minocycline effectively cleared M. marinum from the footpad. Granulomas with necrotic abscesses formed on the footpad of M. marinum-infected mice, primarily due to neutrophils involved in the host's cell-mediated immune response. Inflammatory cytokine and chemokine levels significantly increased 7 days post-infection, aligning with the footpad swelling and granuloma formation observed in the untreated group. Interestingly, immune mediators and cells induced by M. marinum footpad infection were crucial factors associated with hypersensitivity and granuloma formation, as seen in pulmonary tuberculosis. This novel imaging analysis using a cutaneous NTM mouse model might be a powerful tool for the comprehensive analysis of mycobacterial infections.
Collapse
Affiliation(s)
- Kentaro Yamamoto
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan.
| | - Shota Torigoe
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan
- Research Center for Biosafety, Laboratory Animal and Pathogen Bank, National Institute of Infectious Diseases, Toyama, Shinjuku, Tokyo, Japan
| | - Yusuke Tsujimura
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan
| | - Masamitsu N Asaka
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan
| | - Kayo Okumura
- Research Center for Biosafety, Laboratory Animal and Pathogen Bank, National Institute of Infectious Diseases, Toyama, Shinjuku, Tokyo, Japan
| | - Manabu Ato
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Aoba-cho, Higashimurayama, Tokyo, Japan
| |
Collapse
|
5
|
Ashbery D, Baez HC, Kanarr RE, Kunala K, Power D, Chu CJ, Schallek J, McGregor JE. In Vivo Visualization of Intravascular Patrolling Immune Cells in the Primate Eye. Invest Ophthalmol Vis Sci 2024; 65:23. [PMID: 39283618 PMCID: PMC11407476 DOI: 10.1167/iovs.65.11.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Purpose Insight into the immune status of the living eye is essential as we seek to understand ocular disease and develop new treatments. The nonhuman primate (NHP) is the gold standard preclinical model for therapeutic development in ophthalmology, owing to the similar visual system and immune landscape in the NHP relative to the human. Here, we demonstrate the utility of phase-contrast adaptive optics scanning light ophthalmoscope (AOSLO) to visualize immune cell dynamics on the cellular scale, label-free in the NHP. Methods Phase-contrast AOSLO was used to image preselected areas of retinal vasculature in five NHP eyes. Images were registered to correct for eye motion, temporally averaged, and analyzed for immune cell activity. Cell counts, dimensions, velocities, and frequency per vessel were determined manually and compared between retinal arterioles and venules. Based on cell appearance and circularity index, cells were divided into three morphologies: ovoid, semicircular, and flattened. Results Immune cells were observed migrating along vascular endothelium with and against blood flow. Cell velocity did not significantly differ between morphology or vessel type and was independent of blow flood. Venules had a significantly higher cell frequency than arterioles. A higher proportion of cells resembled "flattened" morphology in arterioles. Based on cell speeds, morphologies, and behaviors, we identified these cells as nonclassical patrolling monocytes (NCPMs). Conclusions Phase-contrast AOSLO has the potential to reveal the once hidden behaviors of single immune cells in retinal circulation and can do so without the requirement of added contrast agents that may disrupt immune cell behavior.
Collapse
Affiliation(s)
- Drew Ashbery
- University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
- Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Hector C Baez
- Center for Visual Science, University of Rochester, Rochester, New York, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, United States
| | - Rye E Kanarr
- Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Karteek Kunala
- Byers Eye Institute, Stanford University, Palo Alto, California, United States
| | - Derek Power
- Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Colin J Chu
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Jesse Schallek
- Center for Visual Science, University of Rochester, Rochester, New York, United States
- Flaum Eye Institute, University of Rochester, Rochester, New York, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, United States
- Department of Neuroscience, University of Rochester, Rochester, New York, United States
| | - Juliette E McGregor
- Center for Visual Science, University of Rochester, Rochester, New York, United States
- Flaum Eye Institute, University of Rochester, Rochester, New York, United States
| |
Collapse
|
6
|
Li X, Sun X, Chen H, Wang Y, Chen H, Gao Y. Boron Dipyrromethene-Based Nanotheranostic System for Sonophotoassisted Therapy and Simultaneous Monitoring of Tumor Immune Microenvironment Reprogramming. ACS NANO 2024; 18:18230-18245. [PMID: 38950337 DOI: 10.1021/acsnano.4c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Therapy-induced modulation of the tumor microenvironment (TME) to overcome the immunosuppressive TME is considered to be an opportunity for cancer treatment. However, monitoring of TME modulation during the therapeutic process to accurately determine immune responses and adjust treatment plans in a timely manner remains to be challenging. Herein, we report a carrier-free nanotheranostic system (CANPs) assembled by two boron dipyrromethene (BODIPY) dyes, a sonophotosensitizer C-BDP, and a nitric oxide (NO) probe amino-BODIPY (A-BDP). CANPs can exert combined sonophototherapeutic effects of C-BDP under ultrasound and light irradiation and simultaneously induce inflammatory TME, as well as emit bright fluorescence via A-BDP by monitoring tumor-associated macrophages (TAMs) repolarization through the released NO in vitro and in vivo. Of note, transforming growth factor-β (TGF-β) could be the key cytokine involved in the sonophototherapy-induced TME reprogramming. By virtue of high physiological stability, good biocompatibility, and effective tumor targetability, CANPs could be a potential nanotheranostic system for the simultaneous induction and detection of TME reprogramming triggered by sonophototherapy.
Collapse
Affiliation(s)
- Xudong Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| | - Xianbin Sun
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| | - Hui Chen
- College of Chemistry, Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), Fuzhou University, Fuzhou, Fujian 350116, China
| | - Ya Wang
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| | - Haijun Chen
- College of Chemistry, Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), Fuzhou University, Fuzhou, Fujian 350116, China
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
7
|
Lemdani K, Marlin R, Mayet C, Perkov V, Pascal Q, Ripoll M, Relouzat F, Dhooge N, Bossevot L, Dereuddre-Bosquet N, Dargazanli G, Thibaut-Duprey K, Haensler J, Chapon C, Prost C, Le Grand R. Distinct dynamics of mRNA LNPs in mice and nonhuman primates revealed by in vivo imaging. NPJ Vaccines 2024; 9:113. [PMID: 38902327 PMCID: PMC11189915 DOI: 10.1038/s41541-024-00900-5] [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: 12/07/2023] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
The characterization of vaccine distribution to relevant tissues after in vivo administration is critical to understanding their mechanisms of action. Vaccines based on mRNA lipid nanoparticles (LNPs) are now being widely considered against infectious diseases and cancer. Here, we used in vivo imaging approaches to compare the trafficking of two LNP formulations encapsulating mRNA following intramuscular administration: DLin-MC3-DMA (MC3) and the recently developed DOG-IM4. The mRNA formulated in DOG-IM4 LNPs persisted at the injection site, whereas mRNA formulated in MC3 LNPs rapidly migrated to the draining lymph nodes. Furthermore, MC3 LNPs induced the fastest increase in blood neutrophil counts after injection and greater inflammation, as shown by IL-1RA, IL-15, CCL-1, and IL-6 concentrations in nonhuman primate sera. These observations highlight the influence of the nature of the LNP on mRNA vaccine distribution and early immune responses.
Collapse
Affiliation(s)
- Katia Lemdani
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
- Sanofi, Marcy-L'étoile, France
| | - Romain Marlin
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Céline Mayet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | - Quentin Pascal
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | - Francis Relouzat
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Nina Dhooge
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Laetitia Bossevot
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | | | | | - Catherine Chapon
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France.
| |
Collapse
|
8
|
Racca NM, Dontu A, Riley K, Yolcu ES, Shirwan H, Coronel MM. Bending the Rules: Amplifying PD-L1 Immunoregulatory Function Through Flexible Polyethylene Glycol Synthetic Linkers. Tissue Eng Part A 2024; 30:299-313. [PMID: 38318841 DOI: 10.1089/ten.tea.2023.0274] [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: 02/07/2024] Open
Abstract
Immune checkpoint signaling, such as programmed cell death protein-1 (PD-1), is a key target for immunotherapy due to its role in dampening immune responses. PD-1 signaling in T cells is regulated by complex physicochemical and mechanical cues. However, how these mechanical forces are integrated with biochemical responses remains poorly understood. Our previous work demonstrated that the use of an immobilizing polyethylene glycol (PEG) linker on synthetic microgels for the presentation of a chimeric form of PD-L1, SA-PD-L1, lead to local regulatory responses capable of abrogating allograft rejection in a model of cell-based transplantation. We herein provide evidence that enhanced immune regulating function can be obtained when presentation of SA-PD-L1 is achieved through a longer more flexible PEG chain. Presentation of SA-PD-L1 through a linker of high molecular weight, and thus longer length (10 kDa, 60 nm in length), led to enhance conversion of naive T cells into T regulatory cells (Tregs) in vitro. In addition, using a subcutaneous implant model and protein tethered through three different linker sizes (6, 30, and 60 nm) to the surface of PEG hydrogels, we demonstrated that longer linkers promoted PD-1 immunomodulatory role in vivo through three main functions: (1) augmenting immune cell recruitment at the transplant site; (2) promoting the accumulation of naive Tregs expressing migratory markers; and (3) dampening CD8+ cytolytic molecule production while augmenting expression of exhaustion phenotypes locally. Notably, accumulation of Treg cells at the implant site persisted for over 30 days postimplantation, an effect not observed when protein was presented with the shorter version of the linkers (6 and 30 nm). Collectively, these studies reveal a facile approach by which PD-L1 function can be modulated through external tuning of synthetic presenting linkers. Impact statement Recently, there has been a growing interest in immune checkpoint molecules as potential targets for tolerance induction, including programmed cell death protein-1 (PD-1). However, how the mechanics of ligand binding to PD-1 receptor affect downstream activation signaling pathways remains unresolved. By taking advantage of the effect of polyethylene glycol chain length on molecule kinetics in an aqueous solution, we herein show that PD-L1 function can be amplified by adjusting the length of the grafting linker. Our results uncover a potential facile mechanism that can be exploited to advance the role of immune checkpoint ligands, in particular PD-L1, in tolerance induction for immunosuppression-free cell-based therapies.
Collapse
Affiliation(s)
- Nicole M Racca
- Department of Biomedical Engineering and Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
- Elizabeth Caswell Diabetes Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexander Dontu
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kayle Riley
- Department of Biomedical Engineering and Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
- Elizabeth Caswell Diabetes Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Esma S Yolcu
- Department of Pediatrics and University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Haval Shirwan
- Department of Pediatrics and University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
- Associate Director, Immunomodulation and Regenerative Medicine Program, Ellis Fischel Cancer Center, Columbia, Missouri, USA
| | - María M Coronel
- Department of Biomedical Engineering and Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
- Elizabeth Caswell Diabetes Institute, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
Kazim M, Yoo E. Recent Advances in the Development of Non-Invasive Imaging Probes for Cancer Immunotherapy. Angew Chem Int Ed Engl 2024; 63:e202310694. [PMID: 37843426 DOI: 10.1002/anie.202310694] [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: 07/26/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/17/2023]
Abstract
The last two decades have witnessed a major revolution in the field of tumor immunology including clinical progress using various immunotherapy strategies. These advances have highlighted the potential for approaches that harness the power of the immune system to fight against cancer. While cancer immunotherapies have shown significant clinical successes, patient responses vary widely due to the complex and heterogeneous nature of tumors and immune responses, calling for reliable biomarkers and therapeutic strategies to maximize the benefits of immunotherapy. Especially, stratifying responding individuals from non-responders during the early stages of treatment could help avoid long-term damage and tailor personalized treatments. In efforts to develop non-invasive means for accurately evaluating and predicting tumor response to immunotherapy, multiple affinity-based agents targeting immune cell markers and checkpoint molecules have been developed and advanced to clinical trials. In addition, researchers have recently turned their attention to substrate and activity-based imaging probes that can provide real-time, functional assessment of immune response to treatment. Here, we highlight some of those recently designed probes that image functional proteases as biomarkers of cancer immunotherapy with a focus on their chemical design and detection modalities and discuss challenges and opportunities for the development of imaging tools utilized in cancer immunotherapy.
Collapse
Affiliation(s)
- Muhammad Kazim
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Euna Yoo
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
10
|
Cawthorne CJ, Volpe A, Fruhwirth GO. The Basics of Visualizing, Analyzing, and Reporting Preclinical PET/CT Imaging Data. Methods Mol Biol 2024; 2729:195-220. [PMID: 38006498 DOI: 10.1007/978-1-0716-3499-8_12] [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: 11/27/2023]
Abstract
Positron emission tomography (PET) has transformed medical imaging, and while first developed and applied to the human setting, it has found widespread application at the preclinical level over the past two decades. Its strength is that it offers noninvasive 3D tomographic imaging in a quantitative manner at very high sensitivity. Paired with the right molecular probes, invaluable insights into physiology and pathophysiology have been accessible and therapeutic development has been enhanced through preclinical PET imaging. PET imaging is now often routinely combined with either computed tomography (CT) or magnetic resonance imaging (MRI) to provide additional anatomical context. All these developments were accompanied by the provision of ever more complex and powerful analysis software enabling users to visualize and quantify signals from PET imaging data. Aside from experimental complexities, there are also various pitfalls in PET image data analysis, which can negatively impact on reporting and reproducibility.Here, we provide a protocol intended to guide the inexperienced user through PET/CT data analysis. We describe the general principles and workflows required for PET/CT image data visualization and quantitative analysis using various software packages popular in the field. Moreover, we present recommendations for reporting of preclinical PET/CT data including examples of good and poor practice.
Collapse
Affiliation(s)
- Christopher J Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit Leuven, Leuven, Belgium.
| | - Alessia Volpe
- Molecular Imaging Group, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gilbert O Fruhwirth
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK.
| |
Collapse
|
11
|
Cella L, Monti S, Pacelli R, Palma G. Modeling frameworks for radiation induced lymphopenia: A critical review. Radiother Oncol 2024; 190:110041. [PMID: 38042499 DOI: 10.1016/j.radonc.2023.110041] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/17/2023] [Accepted: 11/25/2023] [Indexed: 12/04/2023]
Abstract
Radiation-induced lymphopenia (RIL) is a frequent, and often considered unavoidable, side effect of radiation therapy (RT), whether or not chemotherapy is included. However, in the last few years several studies have demonstrated the detrimental effect of RIL on therapeutic outcomes, with conflicting findings concerning possible inferior patient survival. In addition, since immunotherapeutic treatment has become an integral part of cancer therapy, preserving the immune system is recognized as crucial. Given this background, various research groups have reported on different frameworks for modelling RIL, frequently based on different definitions of RIL itself, and discordant results have been reported. Our aim is to critically review the current literature on RIL modelling and summarize the different approaches recently proposed to improve the prediction of RIL after RT and aimed at immunity-sparing RT. A detailed description of these approaches will be outlined and illustrated through their applications as found in the literature from the last five years. Such a critical analysis represents the necessary starting step to develop an effective strategy that ultimately could harmonize the diverse modelling methods.
Collapse
Affiliation(s)
- Laura Cella
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy.
| | - Serena Monti
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Roberto Pacelli
- Department of Advanced Biomedical Sciences, Federico II School of Medicine, Naples, Italy
| | - Giuseppe Palma
- Institute of Nanotechnology, National Research Council, Lecce, Italy
| |
Collapse
|
12
|
Mellid-Carballal R, Gutierrez-Gutierrez S, Rivas C, Garcia-Fuentes M. Viral protein-based nanoparticles (part 2): Pharmaceutical applications. Eur J Pharm Sci 2023; 189:106558. [PMID: 37567394 DOI: 10.1016/j.ejps.2023.106558] [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: 02/17/2023] [Revised: 07/10/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Viral protein nanoparticles (ViP NPs) such as virus-like particles and virosomes are structures halfway between viruses and synthetic nanoparticles. The biological nature of ViP NPs endows them with the biocompatibility, biodegradability, and functional properties that many synthetic nanoparticles lack. At the same time, the absence of a viral genome avoids the safety concerns of viruses. Such characteristics of ViP NPs offer a myriad of opportunities for theirapplication at several points across disease development: from prophylaxis to diagnosis and treatment. ViP NPs present remarkable immunostimulant properties, and thus the vaccination field has benefited the most from these platforms capable of overcoming the limitations of both traditional and subunit vaccines. This was reflected in the marketing authorization of several VLP- and virosome-based vaccines. Besides, ViP NPs inherit the ability of viruses to deliver their cargo to target cells. Because of that, ViP NPs are promising candidates as vectors for drug and gene delivery, and for diagnostic applications. In this review, we analyze the pharmaceutical applications of ViP NPs, describing the products that are commercially available or under clinical evaluation, but also the advances that scientists are making toward the implementation of ViP NPs in other areas of major pharmaceutical interest.
Collapse
Affiliation(s)
- Rocio Mellid-Carballal
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Sara Gutierrez-Gutierrez
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Carmen Rivas
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain; Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CNB)-CSIC, Spain
| | - Marcos Garcia-Fuentes
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain.
| |
Collapse
|
13
|
Laaker C, Baenen C, Kovács KG, Sandor M, Fabry Z. Immune cells as messengers from the CNS to the periphery: the role of the meningeal lymphatic system in immune cell migration from the CNS. Front Immunol 2023; 14:1233908. [PMID: 37662908 PMCID: PMC10471710 DOI: 10.3389/fimmu.2023.1233908] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
In recent decades there has been a large focus on understanding the mechanisms of peripheral immune cell infiltration into the central nervous system (CNS) in neuroinflammatory diseases. This intense research led to several immunomodulatory therapies to attempt to regulate immune cell infiltration at the blood brain barrier (BBB), the choroid plexus (ChP) epithelium, and the glial barrier. The fate of these infiltrating immune cells depends on both the neuroinflammatory environment and their type-specific interactions with innate cells of the CNS. Although the fate of the majority of tissue infiltrating immune cells is death, a percentage of these cells could become tissue resident immune cells. Additionally, key populations of immune cells can possess the ability to "drain" out of the CNS and act as messengers reporting signals from the CNS toward peripheral lymphatics. Recent data supports that the meningeal lymphatic system is involved not just in fluid homeostatic functions in the CNS but also in facilitating immune cell migration, most notably dendritic cell migration from the CNS to the meningeal borders and to the draining cervical lymph nodes. Similar to the peripheral sites, draining immune cells from the CNS during neuroinflammation have the potential to coordinate immunity in the lymph nodes and thus influence disease. Here in this review, we will evaluate evidence of immune cell drainage from the brain via the meningeal lymphatics and establish the importance of this in animal models and humans. We will discuss how targeting immune cells at sites like the meningeal lymphatics could provide a new mechanism to better provide treatment for a variety of neurological conditions.
Collapse
Affiliation(s)
- Collin Laaker
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI, United States
| | - Cameron Baenen
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Kristóf G. Kovács
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| |
Collapse
|
14
|
Kakaei N, Amirian R, Azadi M, Mohammadi G, Izadi Z. Perfluorocarbons: A perspective of theranostic applications and challenges. Front Bioeng Biotechnol 2023; 11:1115254. [PMID: 37600314 PMCID: PMC10436007 DOI: 10.3389/fbioe.2023.1115254] [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: 12/03/2022] [Accepted: 03/15/2023] [Indexed: 08/22/2023] Open
Abstract
Perfluorocarbon (PFC) are biocompatible compounds, chemically and biologically inert, and lacks toxicity as oxygen carriers. PFCs nanoemulsions and nanoparticles (NPs) are highly used in diagnostic imaging and enable novel imaging technology in clinical imaging modalities to notice and image pathological and physiological alterations. Therapeutics with PFCs such as the innovative approach to preventing thrombus formation, PFC nanodroplets utilized in ultrasonic medication delivery in arthritis, or PFC-based NPs such as Perfluortributylamine (PFTBA), Pentafluorophenyl (PFP), Perfluorohexan (PFH), Perfluorooctyl bromide (PFOB), and others, recently become renowned for oxygenating tumors and enhancing the effects of anticancer treatments as oxygen carriers for tumor hypoxia. In this review, we will discuss the recent advancements that have been made in PFC's applications in theranostic (therapeutics and diagnostics) as well as assess the benefits and drawbacks of these applications.
Collapse
Affiliation(s)
- Nasrin Kakaei
- Student Research Committee, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roshanak Amirian
- Student Research Committee, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehdi Azadi
- Student Research Committee, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghobad Mohammadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zhila Izadi
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
15
|
Jacob JB, Wei KC, Bepler G, Reyes JD, Cani A, Polin L, White K, Kim S, Viola N, McGrath J, Guastella A, Yin C, Mi QS, Kidder BL, Wagner KU, Ratner S, Phillips V, Xiu J, Parajuli P, Wei WZ. Identification of actionable targets for breast cancer intervention using a diversity outbred mouse model. iScience 2023; 26:106320. [PMID: 36968078 PMCID: PMC10034465 DOI: 10.1016/j.isci.2023.106320] [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: 08/29/2022] [Revised: 01/16/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
HER2-targeted therapy has improved breast cancer survival, but treatment resistance and disease prevention remain major challenges. Genes that enable HER2/Neu oncogenesis are the next intervention targets. A bioinformatics discovery platform of HER2/Neu-expressing Diversity Outbred (DO) F1 Mice was established to identify cancer-enabling genes. Quantitative Trait Loci (QTL) associated with onset ages and growth rates of spontaneous mammary tumors were sought. Twenty-six genes in 3 QTL contain sequence variations unique to the genetic backgrounds that are linked to aggressive tumors and 21 genes are associated with human breast cancer survival. Concurrent identification of TSC22D3, a transcription factor, and its target gene LILRB4, a myeloid cell checkpoint receptor, suggests an immune axis for regulation, or intervention, of disease. We also investigated TIEG1 gene that impedes tumor immunity but suppresses tumor growth. Although not an actionable target, TIEG1 study revealed genetic regulation of tumor progression, forming the basis of the genetics-based discovery platform.
Collapse
Affiliation(s)
- Jennifer B. Jacob
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kuang-Chung Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Gerold Bepler
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Joyce D. Reyes
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Andi Cani
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lisa Polin
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kathryn White
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Seongho Kim
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Nerissa Viola
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Julie McGrath
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - Anthony Guastella
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - CongCong Yin
- Department of Immunology, Henry Ford Health System, Detroit, MI48202, USA
| | - Qing-Shen Mi
- Department of Immunology, Henry Ford Health System, Detroit, MI48202, USA
| | - Benjamin L. Kidder
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kay-Uwe Wagner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Stuart Ratner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Victoria Phillips
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Joanne Xiu
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - Prahlad Parajuli
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Wei-Zen Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
16
|
Schuurman AR, Sloot PMA, Wiersinga WJ, van der Poll T. Embracing complexity in sepsis. Crit Care 2023; 27:102. [PMID: 36906606 PMCID: PMC10007743 DOI: 10.1186/s13054-023-04374-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 02/19/2023] [Indexed: 03/13/2023] Open
Abstract
Sepsis involves the dynamic interplay between a pathogen, the host response, the failure of organ systems, medical interventions and a myriad of other factors. This together results in a complex, dynamic and dysregulated state that has remained ungovernable thus far. While it is generally accepted that sepsis is very complex indeed, the concepts, approaches and methods that are necessary to understand this complexity remain underappreciated. In this perspective we view sepsis through the lens of complexity theory. We describe the concepts that support viewing sepsis as a state of a highly complex, non-linear and spatio-dynamic system. We argue that methods from the field of complex systems are pivotal for a fuller understanding of sepsis, and we highlight the progress that has been made over the last decades in this respect. Still, despite these considerable advancements, methods like computational modelling and network-based analyses continue to fly under the general scientific radar. We discuss what barriers contribute to this disconnect, and what we can do to embrace complexity with regards to measurements, research approaches and clinical applications. Specifically, we advocate a focus on longitudinal, more continuous biological data collection in sepsis. Understanding the complexity of sepsis will require a huge multidisciplinary effort, in which computational approaches derived from complex systems science must be supported by, and integrated with, biological data. Such integration could finetune computational models, guide validation experiments, and identify key pathways that could be targeted to modulate the system to the benefit of the host. We offer an example for immunological predictive modelling, which may inform agile trials that could be adjusted throughout the trajectory of disease. Overall, we argue that we should expand our current mental frameworks of sepsis, and embrace nonlinear, system-based thinking in order to move the field forward.
Collapse
Affiliation(s)
- Alex R Schuurman
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Peter M A Sloot
- Institute for Advanced Study, University of Amsterdam, Amsterdam, The Netherlands
| | - W Joost Wiersinga
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Centre for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centres - Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Division of Infectious Diseases, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Vladimirov N, Perlman O. Molecular MRI-Based Monitoring of Cancer Immunotherapy Treatment Response. Int J Mol Sci 2023; 24:3151. [PMID: 36834563 PMCID: PMC9959624 DOI: 10.3390/ijms24043151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Immunotherapy constitutes a paradigm shift in cancer treatment. Its FDA approval for several indications has yielded improved prognosis for cases where traditional therapy has shown limited efficiency. However, many patients still fail to benefit from this treatment modality, and the exact mechanisms responsible for tumor response are unknown. Noninvasive treatment monitoring is crucial for longitudinal tumor characterization and the early detection of non-responders. While various medical imaging techniques can provide a morphological picture of the lesion and its surrounding tissue, a molecular-oriented imaging approach holds the key to unraveling biological effects that occur much earlier in the immunotherapy timeline. Magnetic resonance imaging (MRI) is a highly versatile imaging modality, where the image contrast can be tailored to emphasize a particular biophysical property of interest using advanced engineering of the imaging pipeline. In this review, recent advances in molecular-MRI based cancer immunotherapy monitoring are described. Next, the presentation of the underlying physics, computational, and biological features are complemented by a critical analysis of the results obtained in preclinical and clinical studies. Finally, emerging artificial intelligence (AI)-based strategies to further distill, quantify, and interpret the image-based molecular MRI information are discussed in terms of perspectives for the future.
Collapse
Affiliation(s)
- Nikita Vladimirov
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
18
|
Gao Y, Wang Z, Cui Y, Xu M, Weng L. Emerging Strategies of Engineering and Tracking Dendritic Cells for Cancer Immunotherapy. ACS APPLIED BIO MATERIALS 2023; 6:24-43. [PMID: 36520013 DOI: 10.1021/acsabm.2c00790] [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: 12/23/2022]
Abstract
Dendritic cells (DCs), a kind of specialized immune cells, play key roles in antitumor immune response and promotion of innate and adaptive immune responses. Recently, many strategies have been developed to utilize DCs in cancer therapy, such as delivering antigens and adjuvants to DCs and using scaffold to recruit and activate DCs. Here we outline how different DC subsets influence antitumor immunity, summarize the FDA-approved vaccines and cancer vaccines under clinical trials, discuss the strategies for engineering DCs and noninvasive tracking of DCs to improve antitumor immunotherapy, and reveal the potential of artificial neural networks for the design of DC based vaccines.
Collapse
Affiliation(s)
- Yu Gao
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Zhixuan Wang
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Ying Cui
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Miaomiao Xu
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Lixing Weng
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.,School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| |
Collapse
|
19
|
Lauri C, Varani M, Bentivoglio V, Capriotti G, Signore A. Present status and future trends in molecular imaging of lymphocytes. Semin Nucl Med 2023; 53:125-134. [PMID: 36150910 PMCID: PMC9489269 DOI: 10.1053/j.semnuclmed.2022.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 01/28/2023]
Abstract
Immune system is emerging as a crucial protagonist in a huge variety of oncologic and non-oncologic conditions including response to vaccines and viral infections (such as SARS-CoV-2). The increasing knowledge of molecular biology underlying these diseases allowed the identification of specific targets and the possibility to use tailored therapies against them. Immunotherapies and vaccines are, indeed, more and more used nowadays for treating infections, cancer and autoimmune diseases and, therefore, there is the need to identify, quantify and monitor immune cell trafficking before and after treatment. This approach will provide crucial information for therapy decision-making. Imaging of B and T-lymphocytes trafficking by using tailored radiopharmaceuticals proved to be a successful nuclear medicine tool. In this review, we will provide an overview of the state of art and future trends for "in vivo" imaging of lymphocyte trafficking and homing by mean of specific receptor-tailored radiopharmaceuticals.
Collapse
Affiliation(s)
- Chiara Lauri
- Nuclear Medicine Unit Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, "Sapienza" University of Rome, Rome, Italy.
| | | | | | | | | |
Collapse
|
20
|
Zhou M, Liang S, Liu D, Ma K, Peng Y, Wang Z. Engineered Nanoprobes for Immune Activation Monitoring. ACS NANO 2022; 16:19940-19958. [PMID: 36454191 DOI: 10.1021/acsnano.2c09743] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The activation of the immune system is critical for cancer immunotherapy and treatments of inflammatory diseases. Non-invasive visualization of immunoactivation is designed to monitor the dynamic nature of the immune response and facilitate the assessment of therapeutic outcomes, which, however, remains challenging. Conventional imaging modalities, such as positron emission tomography, computed tomography, etc., were utilized for imaging immune-related biomarkers. To explore the dynamic immune monitoring, probes with signals correlated to biomarkers of immune activation or prognosis are urgently needed. These emerging molecular probes, which turn on the signal only in the presence of the intended biomarker, can improve the detection specificity. These probes with "turn on" signals enable non-invasive, dynamic, and real-time imaging with high sensitivity and efficiency, showing significance for multifunctionality/multimodality imaging. As a result, more and more innovative engineered nanoprobes combined with diverse imaging modalities were developed to assess the activation of the immune system. In this work, we comprehensively review the recent and emerging advances in engineered nanoprobes for monitoring immune activation in cancer or other immune-mediated inflammatory diseases and discuss the potential in predicting the efficacy following treatments. Research on real-time in vivo immunoimaging is still under exploration, and this review can provide guidance and facilitate the development and application of next-generation imaging technologies.
Collapse
Affiliation(s)
- Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| |
Collapse
|
21
|
Berz AM, Dromain C, Vietti-Violi N, Boughdad S, Duran R. Tumor response assessment on imaging following immunotherapy. Front Oncol 2022; 12:982983. [PMID: 36387133 PMCID: PMC9641095 DOI: 10.3389/fonc.2022.982983] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, various systemic immunotherapies have been developed for cancer treatment, such as monoclonal antibodies (mABs) directed against immune checkpoints (immune checkpoint inhibitors, ICIs), oncolytic viruses, cytokines, cancer vaccines, and adoptive cell transfer. While being estimated to be eligible in 38.5% of patients with metastatic solid or hematological tumors, ICIs, in particular, demonstrate durable disease control across many oncologic diseases (e.g., in melanoma, lung, bladder, renal, head, and neck cancers) and overall survival benefits. Due to their unique mechanisms of action based on T-cell activation, response to immunotherapies is characterized by different patterns, such as progression prior to treatment response (pseudoprogression), hyperprogression, and dissociated responses following treatment. Because these features are not encountered in the Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1), which is the standard for response assessment in oncology, new criteria were defined for immunotherapies. The most important changes in these new morphologic criteria are, firstly, the requirement for confirmatory imaging examinations in case of progression, and secondly, the appearance of new lesions is not necessarily considered a progressive disease. Until today, five morphologic (immune-related response criteria (irRC), immune-related RECIST (irRECIST), immune RECIST (iRECIST), immune-modified RECIST (imRECIST), and intra-tumoral RECIST (itRECIST)) criteria have been developed to accurately assess changes in target lesion sizes, taking into account the specific response patterns after immunotherapy. In addition to morphologic response criteria, 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography/computed tomography (18F-FDG-PET/CT) is a promising option for metabolic response assessment and four metabolic criteria are used (PET/CT Criteria for Early Prediction of Response to Immune Checkpoint Inhibitor Therapy (PECRIT), PET Response Evaluation Criteria for Immunotherapy (PERCIMT), immunotherapy-modified PET Response Criteria in Solid Tumors (imPERCIST5), and immune PERCIST (iPERCIST)). Besides, there is evidence that parameters on 18F-FDG-PET/CT, such as the standardized uptake value (SUV)max and several radiotracers, e.g., directed against PD-L1, may be potential imaging biomarkers of response. Moreover, the emerge of human intratumoral immunotherapy (HIT-IT), characterized by the direct injection of immunostimulatory agents into a tumor lesion, has given new importance to imaging assessment. This article reviews the specific imaging patterns of tumor response and progression and available imaging response criteria following immunotherapy.
Collapse
Affiliation(s)
- Antonia M. Berz
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
- Department of Radiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Clarisse Dromain
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Naïk Vietti-Violi
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Sarah Boughdad
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Rafael Duran
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
22
|
Li Y, Lu S, Zhang Y, Li J, Zhang J, Zhang C, Xiong L. Multifunctional Imaging of Vessels, Brown Adipose Tissue, and Bones in the Visible and Second Near-infrared Region Using Dual-Emitting Polymer Dots. ACS APPLIED MATERIALS & INTERFACES 2022; 14:37504-37513. [PMID: 35970519 DOI: 10.1021/acsami.2c10420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Dual-emitting polymer dots (dual-Pdots) in the visible and second near-infrared (NIR-II) region can facilitate the high-resolution imaging of the fine structure and improve the signal-to-noise ratio in in vivo imaging. Herein, combining high brightness of Pdots and multi-scale imaging, we synthesized dual-Pdots using a simple nano-coprecipitation method and performed multi-functional imaging of vessels, brown adipose tissue, and bones. Results showed that in vivo blood vessel imaging had a high resolution of up to 5.9 μm and bone imaging had a signal-to-noise ratio of 3.9. Moreover, dual-Pdots can accumulate in the interscapular brown adipose tissue within 2 min with a signal-to-noise ratio of 5.8. In addition, the prepared dual-Pdots can image the lymphatic valves and the frequency of contraction. Our study provides a feasible method of using Pdots as nanoprobes for multi-scale imaging in the fields of metabolic disorders, skeletal system diseases, and circulatory systems.
Collapse
Affiliation(s)
- Yuqiao Li
- Shanghai Med-X Engineering Center for Medical Equipment and Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Shuting Lu
- Shanghai Med-X Engineering Center for Medical Equipment and Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Yufan Zhang
- Shanghai Med-X Engineering Center for Medical Equipment and Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Jingru Li
- Shanghai Med-X Engineering Center for Medical Equipment and Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Juxiang Zhang
- Shanghai Med-X Engineering Center for Medical Equipment and Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Chunfu Zhang
- Shanghai Med-X Engineering Center for Medical Equipment and Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Liqin Xiong
- Shanghai Med-X Engineering Center for Medical Equipment and Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| |
Collapse
|
23
|
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: 27] [Impact Index Per Article: 9.0] [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
|
24
|
Xie J, El Rami F, Zhou K, Simonetta F, Chen Z, Zheng X, Chen M, Balakrishnan PB, Dai SY, Murty S, Alam IS, Baker J, Negrin RS, Gambhir SS, Rao J. Multiparameter Longitudinal Imaging of Immune Cell Activity in Chimeric Antigen Receptor T Cell and Checkpoint Blockade Therapies. ACS CENTRAL SCIENCE 2022; 8:590-602. [PMID: 35647285 PMCID: PMC9136971 DOI: 10.1021/acscentsci.2c00142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Indexed: 05/17/2023]
Abstract
Longitudinal multimodal imaging presents unique opportunities for noninvasive surveillance and prediction of treatment response to cancer immunotherapy. In this work we first designed a novel granzyme B activated self-assembly small molecule, G-SNAT, for the assessment of cytotoxic T lymphocyte mediated cancer cell killing. G-SNAT was found to specifically detect the activity of granzyme B within the cytotoxic granules of activated T cells and engaged cancer cells in vitro. In lymphoma tumor-bearing mice, the retention of cyanine 5 labeled G-SNAT-Cy5 correlated to CAR T cell mediated granzyme B exocytosis and tumor eradication. In colorectal tumor-bearing transgenic mice with hematopoietic cells expressing firefly luciferase, longitudinal bioluminescence and fluorescence imaging revealed that after combination treatment of anti-PD-1 and anti-CTLA-4, the dynamics of immune cell trafficking, tumor infiltration, and cytotoxic activity predicted the therapeutic outcome before tumor shrinkage was evident. These results support further development of G-SNAT for imaging early immune response to checkpoint blockade and CAR T-cell therapy in patients and highlight the utility of multimodality imaging for improved mechanistic insights into cancer immunotherapy.
Collapse
Affiliation(s)
- Jinghang Xie
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Fadi El Rami
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Kaixiang Zhou
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Federico Simonetta
- Division
of Blood and Marrow Transplantation, Department of Medicine, Stanford University Medical Center, Stanford, California 94305, United States
| | - Zixin Chen
- Department of Chemistry, Department of Bioengineering, and Department of Materials Science
& Engineering, Stanford University, Stanford, California 94305, United States
| | - Xianchuang Zheng
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Min Chen
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Preethi B. Balakrishnan
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Sheng-Yao Dai
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Surya Murty
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Chemistry, Department of Bioengineering, and Department of Materials Science
& Engineering, Stanford University, Stanford, California 94305, United States
| | - Israt S. Alam
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jeanette Baker
- Division
of Blood and Marrow Transplantation, Department of Medicine, Stanford University Medical Center, Stanford, California 94305, United States
| | - Robert S. Negrin
- Division
of Blood and Marrow Transplantation, Department of Medicine, Stanford University Medical Center, Stanford, California 94305, United States
| | - Sanjiv S. Gambhir
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Chemistry, Department of Bioengineering, and Department of Materials Science
& Engineering, Stanford University, Stanford, California 94305, United States
| | - Jianghong Rao
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Chemistry, Department of Bioengineering, and Department of Materials Science
& Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
25
|
Albers JJ, Pelka K. Listening in on Multicellular Communication in Human Tissue Immunology. Front Immunol 2022; 13:884185. [PMID: 35634333 PMCID: PMC9136009 DOI: 10.3389/fimmu.2022.884185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/14/2022] [Indexed: 11/23/2022] Open
Abstract
Immune responses in human tissues rely on the concerted action of different cell types. Inter-cellular communication shapes both the function of the multicellular interaction networks and the fate of the individual cells that comprise them. With the advent of new methods to profile and experimentally perturb primary human tissues, we are now in a position to systematically identify and mechanistically dissect these cell-cell interactions and their modulators. Here, we introduce the concept of multicellular hubs, functional modules of immune responses in tissues. We outline a roadmap to discover multicellular hubs in human tissues and discuss how emerging technologies may further accelerate progress in this field.
Collapse
Affiliation(s)
- Julian J. Albers
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Karin Pelka
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Gladstone-University of California San Francisco (UCSF) Institute of Genomic Immunology, Gladstone Institutes, San Francisco, CA, United States
| |
Collapse
|
26
|
Zambito G, Mishra G, Schliehe C, Mezzanotte L. Near-Infrared Bioluminescence Imaging of Macrophage Sensors for Cancer Detection In Vivo. Front Bioeng Biotechnol 2022; 10:867164. [PMID: 35615475 PMCID: PMC9124759 DOI: 10.3389/fbioe.2022.867164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Melanoma is an aggressive type of skin cancer with a poor prognosis after it gets metastasized. The early detection of malignant melanoma is critical for effective therapy. Because melanoma often resembles moles, routine skin check-up may help for timely identification of suspicious areas. Recently, it has been shown that the interplay of melanoma cells with the immune system can help develop efficient therapeutic strategies. Here, we leveraged engineered macrophages (BMC2) as cell-based sensors for metastatic melanoma. To perform dual-color bioluminescence imaging (BLI) in vivo, macrophages were engineered to express a green click beetle luciferase (CBG2) and a near-infrared fluorescent dye (DiR), and B16F10 melanoma cells were instead engineered to express a near-infrared click beetle luciferase (CBR2). Using real-time in vivo dual-color BLI and near-infrared fluorescence (FL) imaging, we could demonstrate that macrophages were able to sense and substantially accumulate in subcutaneous and metastatic melanoma tissues at 72 h after systemic injections. Together, we showed the potentiality to use optical imaging technologies to track circulating macrophages for the non-invasive detection of metastatic melanoma.
Collapse
Affiliation(s)
- Giorgia Zambito
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Molecular Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Gunja Mishra
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Christopher Schliehe
- Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Molecular Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
27
|
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
|
28
|
Hyodo F, Eto H, Naganuma T, Koyasu N, Elhelaly AE, Noda Y, Kato H, Murata M, Akahoshi T, Hashizume M, Utsumi H, Matsuo M. In Vivo Dynamic Nuclear Polarization Magnetic Resonance Imaging for the Evaluation of Redox-Related Diseases and Theranostics. Antioxid Redox Signal 2022; 36:172-184. [PMID: 34015957 DOI: 10.1089/ars.2021.0087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Significance:In vivo molecular and metabolic imaging is an emerging field in biomedical research that aims to perform noninvasive detection of tissue metabolism in disease states and responses to therapeutic agents. The imbalance in tissue oxidation/reduction (Redox) states is related to the onset and progression of several diseases. Tissue redox metabolism provides biomarkers for early diagnosis and drug treatments. Thus, noninvasive imaging of redox metabolism could be a useful, novel diagnostic tool for diagnosis of redox-related disease and drug discovery. Recent Advances:In vivo dynamic nuclear polarization magnetic resonance imaging (DNP-MRI) is a technique that enables the imaging of free radicals in living animals. DNP enhances the MRI signal by irradiating the target tissue or solution with the free radical molecule's electron paramagnetic resonance frequency before executing pulse sequence of the MRI. In vivo DNP-MRI with redox-sensitive nitroxyl radicals as the DNP redox contrast agent enables the imaging of the redox metabolism on various diseases. Moreover, nitroxyl radicals show antioxidant effects that suppress oxidative stress. Critical Issues: To date, considerable progress has been documented preclinically in the development of animal imaging systems. Here, we review redox imaging of in vivo DNP-MRI with a focus on the recent progress of this system and its uses in patients with redox-related diseases. Future Directions: This technique could have broad applications in the study of other redox-related diseases, such as cancer, inflammation, and neurological disorders, and facilitate the evaluation of treatment response as a theranostic tool. Antioxid. Redox Signal. 36, 172-184.
Collapse
Affiliation(s)
- Fuminori Hyodo
- Department of Radiology, Frontier Science for Imaging, School of Medicine, Gifu University, Gifu, Japan
| | - Hinako Eto
- Center for Advanced Medical Open Innovation, Kyushu University, Fukuoka, Japan
| | | | | | - Abdelazim Elsayed Elhelaly
- Department of Radiology, Frontier Science for Imaging, School of Medicine, Gifu University, Gifu, Japan.,Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Hiroki Kato
- Department of Radiology, Gifu University, Gifu, Japan
| | - Masaharu Murata
- Center for Advanced Medical Open Innovation, Kyushu University, Fukuoka, Japan.,Graduate School of Medicine, Disaster and Emergency Medicine, Kyushu University, Fukuoka, Japan
| | - Tomohiko Akahoshi
- Graduate School of Medicine, Disaster and Emergency Medicine, Kyushu University, Fukuoka, Japan
| | | | - Hideo Utsumi
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | | |
Collapse
|
29
|
Chaurasiya S, Kim SI, O'Leary M, Park AK, Lu J, Kang S, Zhang Z, Yang A, Woo Y, Fong Y, Warner SG. Toward comprehensive imaging of oncolytic viroimmunotherapy. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:303-310. [PMID: 34786474 PMCID: PMC8569424 DOI: 10.1016/j.omto.2021.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Oncolytic viruses infect, replicate in, and kill cancer cells, leaving normal cells unharmed; they also recruit and activate immune cells against tumor cells. While clinical indications for viroimmunotherapy are growing, barriers to widespread treatment remain. Ensuring real-time tracking of viral replication and resulting anti-tumor immune responses will overcome some of these barriers and is thus a top priority. Clinically optimizing trackability of viral replication will promote safe dose increases, guide serial dosing, and enhance treatment effects. However, viral delivery is only half the story. Oncolytic viruses are known to upregulate immune checkpoint expression, thereby priming otherwise immunodeficient tumor immune microenvironments for treatment with checkpoint inhibitors. Novel modalities to track virus-induced changes in tumor microenvironments include non-invasive measurements of immune cell populations and responses to viroimmunotherapy such as (1) in situ use of radiotracers to track checkpoint protein expression or immune cell traffic, and (2) ex vivo labeling of immune cells followed by nuclear medicine imaging. Herein, we review clinical progress toward accurate imaging of oncolytic virus replication, and we further review the current status of functional imaging of immune responses to viroimmunotherapy.
Collapse
Affiliation(s)
- Shyambabu Chaurasiya
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Sang-In Kim
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Michael O'Leary
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Anthony K Park
- Center for Gene Therapy, Department of Hematologic and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jianming Lu
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Seonah Kang
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Zhifang Zhang
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Annie Yang
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Yanghee Woo
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Yuman Fong
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Susanne G Warner
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| |
Collapse
|
30
|
Bošnjak B, Do KTH, Förster R, Hammerschmidt SI. Imaging dendritic cell functions. Immunol Rev 2021; 306:137-163. [PMID: 34859450 DOI: 10.1111/imr.13050] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs) are crucial for the appropriate initiation of adaptive immune responses. During inflammation, DCs capture antigens, mature, and migrate to lymphoid tissues to present foreign material to naïve T cells. These cells get activated and differentiate either into pathogen-specific cytotoxic CD8+ T cells that destroy infected cells or into CD4+ T helper cells that, among other effector functions, orchestrate antibody production by B cells. DC-mediated antigen presentation is equally important in non-inflammatory conditions. Here, DCs mediate induction of tolerance by presenting self-antigens or harmless environmental antigens and induce differentiation of regulatory T cells or inactivation of self-reactive immune cells. Detailed insights into the biology of DCs are, therefore, crucial for the development of novel vaccines as well as the prevention of autoimmune diseases. As in many other life science areas, our understanding of DC biology would be extremely restricted without bioimaging, a compilation of methods that visualize biological processes. Spatiotemporal tracking of DCs relies on various imaging tools, which not only enable insights into their positioning and migration within tissues or entire organs but also allow visualization of subcellular and molecular processes. This review aims to provide an overview of the imaging toolbox and to provide examples of diverse imaging techniques used to obtain fundamental insights into DC biology.
Collapse
Affiliation(s)
- Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Kim Thi Hoang Do
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence RESIST (EXC 2155) Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Hannover, Germany
| | | |
Collapse
|
31
|
Lim TX, Ahamed M, Reutens DC. The aryl hydrocarbon receptor: A diagnostic and therapeutic target in glioma. Drug Discov Today 2021; 27:422-435. [PMID: 34624509 DOI: 10.1016/j.drudis.2021.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 07/29/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
Glioblastoma multiforme (GBM) is a deadly disease; 5-year survival rates have shown little improvement over the past 30 years. In vivo positron emission tomography (PET) imaging is an important method of identifying potential diagnostic and therapeutic molecular targets non-invasively. The aryl hydrocarbon receptor (AhR) is a transcription factor that regulates multiple genes involved in immune response modulation and tumorigenesis. The AhR is an attractive potential drug target and studies have shown that its activation by small molecules can modulate innate and adaptive immunity beneficially and prevent AhR-mediated tumour promotion in several cancer types. In this review, we provide an overview of the role of the AhR in glioma tumorigenesis and highlight its potential as an emerging biomarker for glioma therapies targeting the tumour immune response and PET diagnostics.
Collapse
Affiliation(s)
- Ting Xiang Lim
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Muneer Ahamed
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - David C Reutens
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
32
|
Liu J, Xu N, Wang X, Wang Y, Wu Q, Li X, Pan D, Wang L, Xu Y, Yan J, Li X, Yu L, Yang M. Quantitative radio-thin-layer chromatography and positron emission tomography studies for measuring streptavidin transduced chimeric antigen receptor T cells. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1182:122944. [PMID: 34592686 DOI: 10.1016/j.jchromb.2021.122944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/13/2021] [Accepted: 09/15/2021] [Indexed: 11/20/2022]
Abstract
The proliferation of chimeric antigen receptor (CAR) T cells is closely related to their efficacy, but it is still a great challenge to monitor and quantify CAR T cells in vivo. Based on the high affinity (Kd ≈ 10-15 M) of streptavidin (SA) and biotin, radiolabeled biotin may be used to quantify SA-transduced CAR T cells (SA-CAR T cells). Radio-thin-layer chromatography (radio-TLC) and positron emission tomography (PET) are highly sensitive for trace analysis. Our aim was to develop radio-TLC and PET methods to quantify SA-CAR T cells in vitro and in vivo. First, we developed [68Ga]-DOTA-biotin. Commercially available SA was used as a standard, and quantitative standard curves were established in vitro and in vivo by radio-TLC and PET. Furthermore, the feasibility of the method was verified in Raji model mice. The linear range of radio-TLC was 0.02 ∼ 0.15 pmol/μL with R2 = 0.9993 in vitro. The linear range of PET was 0.02 ∼ 0.76 pmol/μL with R2 = 0.9986 in vivo. SA in CAR T cells can also be accurately quantified in a Raji leukemia model according to PET imaging. The radio-TLC/PET method established in this study is promising for using in the dynamic monitoring and analysis of SA-CAR T cells during therapy.
Collapse
Affiliation(s)
- Jingjing Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Nan Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai 200062, China
| | - Xinyu Wang
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yan Wang
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Qiong Wu
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Xinxin Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Donghui Pan
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Lizhen Wang
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yuping Xu
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Junjie Yan
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Xiaotian Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lei Yu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai 200062, China
| | - Min Yang
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| |
Collapse
|
33
|
Kim EE, Youn H, Kang KW. Imaging in Tumor Immunology. Nucl Med Mol Imaging 2021; 55:225-236. [PMID: 34721715 PMCID: PMC8517056 DOI: 10.1007/s13139-021-00706-6] [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: 03/02/2021] [Revised: 05/31/2021] [Accepted: 06/15/2021] [Indexed: 10/20/2022] Open
Abstract
Recent advances in immune modulation have made impressive progress in cancer immunotherapy. Because dynamic nature of the immune response often makes it difficult to evaluate therapeutic outcomes, innovative imaging technologies have been developed to enable non-invasive visualization of immune cells and tumors in their microenvironment. This review summarizes the current tumor immunology and describes new innovative imaging methods with great potential to obtain non-invasive real-time insights into the complex functions of the immune system and into the management of cancer immunotherapy.
Collapse
Affiliation(s)
- Euishin Edmund Kim
- Department of Radiological Sciences, UCI Medical Center, Orange County, CA USA
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul, South Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
34
|
Pavlova TV, Pilkevich NB, Bessmertnyi DV, Pavlov IA, Atiakshin DV, Pavlova LA. Evaluation of New Immunohistochemical Approaches for the Study of Kidney Tumors in Geriatric. ARCHIVES OF RAZI INSTITUTE 2021; 76:1107-1113. [PMID: 35096347 PMCID: PMC8790976 DOI: 10.22092/ari.2021.355858.1731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/09/2021] [Indexed: 01/24/2023]
Abstract
Kidney malignancies are among the most deadly genitourinary tumors. It is more common in males and is often seen in people aged 60-70 years old. The incidence rate of kidney cancer seems to be increasing. One reason for this may be the fact that imaging techniques, such as computed tomography scans are more commonly used. These tests may lead to the accidental detection of more kidney cancers. Fortunately, kidney cancer is often detected in the early stages, when the tumor is small and confined to the kidney. The objective of this study was the development of new diagnostic immunohistochemical methods. Clinical examination material of 134 people, including 94 (70%) males and 40 (30%) females, were used in this study. Immunohistochemical staining of tryptase was carried out in compliance with the requirements using Anti-Mast Cell Tryptase antibodies. Goat anti-mouse antibodies #AS-M1-HRP were used as secondary antibodies, visualized with ImmPACTTM DAB Peroxidase Substrate Kit (#SK-4105) according to the instructions of the manufacturer. The nuclei were counterstained with Mayer's hematoxylin, and the sections were embedded in a permanent mounting medium. The immunohistochemical study showed an increase in both tryptase- and chymase-positive mast cells in the renal parenchyma, compared with the control group. The number of mast cells with tryptase expression directly in the tumor was significantly less than the peritumoral localization. A similar pattern was observed for chymase-positive mast cells as the content of the tumor was more than 10 times higher than the intratumoral arrangement. The histological and immunological characteristics did not differ in different age groups. The immunohistochemical method of research in the diagnosis of renal tumors plays an important diagnostic and prognostic value. It can assist pathologists in difficult and ambiguous cases to correctly diagnose renal tumors. This will make it possible to prescribe the correct treatment and predict the course of malignant tumor growth in patients.
Collapse
Affiliation(s)
- T V Pavlova
- Department of Pathology, Belgorod State University, Belgorod, Russian Federation
| | - N B Pilkevich
- Department of Pathology, Belgorod State University, Belgorod, Russian Federation
| | - D V Bessmertnyi
- Department of Pathology, Belgorod State University, Belgorod, Russian Federation
| | - I A Pavlov
- Urological Department, Belgorod Cancer Clinic, Belgorod, Russian Federation
| | - D V Atiakshin
- Voronezh State Medical Academy Named after N. N. Burdenko, 394036, Voronezh, Russian Federation
| | - L A Pavlova
- Department of Pathology, Belgorod State University, Belgorod, Russian Federation
| |
Collapse
|
35
|
Kiraga Ł, Kucharzewska P, Paisey S, Cheda Ł, Domańska A, Rogulski Z, Rygiel TP, Boffi A, Król M. Nuclear imaging for immune cell tracking in vivo – Comparison of various cell labeling methods and their application. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
36
|
Mueller CG, Gaiddon C, Venkatasamy A. Current Clinical and Pre-Clinical Imaging Approaches to Study the Cancer-Associated Immune System. Front Immunol 2021; 12:716860. [PMID: 34539653 PMCID: PMC8446654 DOI: 10.3389/fimmu.2021.716860] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/16/2021] [Indexed: 02/01/2023] Open
Abstract
In the light of the success and the expected growth of its arsenal, immuno-therapy may become the standard neoadjuvant procedure for many cancers in the near future. However, aspects such as the identity, organization and the activation status of the peri- and intra-tumoral immune cells would represent important elements to weigh in the decision for the appropriate treatment. While important progress in non-invasive imaging of immune cells has been made over the last decades, it falls yet short of entering the clinics, let alone becoming a standard procedure. Here, we provide an overview of the different intra-vital imaging approaches in the clinics and in pre-clinical settings and discuss their benefits and drawbacks for assessing the activity of the immune system, globally and on a cellular level. Stimulated by further research, the future is likely to see many technological advances both on signal detection and emission as well as image specificity and resolution to tackle current hurdles. We anticipate that the ability to precisely determine an immune stage of cancer will capture the attention of the oncologist and will create a change in paradigm for cancer therapy.
Collapse
Affiliation(s)
- Christopher G Mueller
- CNRS UPR 3572, University of Strasbourg, Immunologie-Immunopathologie-Chimie Thérapeutique, Strasbourg, France
| | - Christian Gaiddon
- Inserm UMR_S 1113, University of Strasbourg, Interface de Recherche Fondamentale et Appliquée en Cancérologie (IRFAC), Strasbourg, France
| | - Aïna Venkatasamy
- Inserm UMR_S 1113, University of Strasbourg, Interface de Recherche Fondamentale et Appliquée en Cancérologie (IRFAC), Strasbourg, France.,IHU-Strasbourg (Institut Hospitalo-Universitaire), Strasbourg, France
| |
Collapse
|
37
|
Olmeda D, Cerezo-Wallis D, Castellano-Sanz E, García-Silva S, Peinado H, Soengas MS. Physiological models for in vivo imaging and targeting the lymphatic system: Nanoparticles and extracellular vesicles. Adv Drug Deliv Rev 2021; 175:113833. [PMID: 34147531 DOI: 10.1016/j.addr.2021.113833] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/24/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023]
Abstract
Imaging of the lymphatic vasculature has gained great attention in various fields, not only because lymphatic vessels act as a key draining system in the body, but also for their implication in autoimmune diseases, organ transplant, inflammation and cancer. Thus, neolymphangiogenesis, or the generation of new lymphatics, is typically an early event in the development of multiple tumor types, particularly in aggressive ones such as malignant melanoma. Still, the understanding of how lymphatic endothelial cells get activated at distal (pre)metastatic niches and their impact on therapy is still unclear. Addressing these questions is of particular interest in the case of immune modulators, because endothelial cells may favor or halt inflammatory processes depending on the cellular context. Therefore, there is great interest in visualizing the lymphatic vasculature in vivo. Here, we review imaging tools and mouse models used to analyze the lymphatic vasculature during tumor progression. We also discuss therapeutic approaches based on nanomedicines to target the lymphatic system and the potential use of extracellular vesicles to track and target sentinel lymph nodes. Finally, we summarize main pre-clinical models developed to visualize the lymphatic vasculature in vivo, discussing their applications with a particular focus in metastatic melanoma.
Collapse
Affiliation(s)
- David Olmeda
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Daniela Cerezo-Wallis
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain; Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, 28029, Spain
| | - Elena Castellano-Sanz
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Susana García-Silva
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Héctor Peinado
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain.
| | - María S Soengas
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain.
| |
Collapse
|
38
|
Nakata J, Isohashi K, Oka Y, Nakajima H, Morimoto S, Fujiki F, Oji Y, Tsuboi A, Kumanogoh A, Hashimoto N, Hatazawa J, Sugiyama H. Imaging Assessment of Tumor Response in the Era of Immunotherapy. Diagnostics (Basel) 2021; 11:diagnostics11061041. [PMID: 34198874 PMCID: PMC8226723 DOI: 10.3390/diagnostics11061041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Assessment of tumor response during treatment is one of the most important purposes of imaging. Before the appearance of immunotherapy, response evaluation criteria in solid tumors (RECIST) and positron emission tomography response criteria in solid tumors (PERCIST) were, respectively, the established morphologic and metabolic response criteria, and cessation of treatment was recommended when progressive disease was detected according to these criteria. However, various types of immunotherapy have been developed over the past 20 years, which show novel false positive findings on images, as well as distinct response patterns from conventional therapies. Antitumor immune response itself causes 18F-fluorodeoxyglucose (FDG) uptake in tumor sites, known as "flare phenomenon", so that positron emission tomography using FDG can no longer accurately identify remaining tumors. Furthermore, tumors often initially increase, followed by stability or decrease resulting from immunotherapy, which is called "pseudoprogression", so that progressive disease cannot be confirmed by computed tomography or magnetic resonance imaging at a single time point. As a result, neither RECIST nor PERCIST can accurately predict the response to immunotherapy, and therefore several new response criteria fixed for immunotherapy have been proposed. However, these criteria are still controversial, and also require months for response confirmation. The establishment of optimal response criteria and the development of new imaging technologies other than FDG are therefore urgently needed. In this review, we summarize the false positive images and the revision of response criteria for each immunotherapy, in order to avoid discontinuation of a truly effective immunotherapy.
Collapse
Affiliation(s)
- Jun Nakata
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
- Correspondence: ; Tel.: +81-6-6879-3676; Fax: +81-6-6879-3677
| | - Kayako Isohashi
- Department of Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, Takatsuki City 596-8686, Osaka, Japan;
| | - Yoshihiro Oka
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
- Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita City 565-0871, Osaka, Japan;
| | - Hiroko Nakajima
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (H.N.); (F.F.); (H.S.)
| | - Soyoko Morimoto
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (S.M.); (A.T.)
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (H.N.); (F.F.); (H.S.)
| | - Yusuke Oji
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (S.M.); (A.T.)
| | - Atsushi Kumanogoh
- Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita City 565-0871, Osaka, Japan;
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan
| | - Naoya Hashimoto
- Department of Neurosurgery, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto City 602-8566, Kyoto, Japan;
| | - Jun Hatazawa
- Department of Research Center for Nuclear Physics, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (H.N.); (F.F.); (H.S.)
| |
Collapse
|
39
|
Van Tilbeurgh M, Lemdani K, Beignon AS, Chapon C, Tchitchek N, Cheraitia L, Marcos Lopez E, Pascal Q, Le Grand R, Maisonnasse P, Manet C. Predictive Markers of Immunogenicity and Efficacy for Human Vaccines. Vaccines (Basel) 2021; 9:579. [PMID: 34205932 PMCID: PMC8226531 DOI: 10.3390/vaccines9060579] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Vaccines represent one of the major advances of modern medicine. Despite the many successes of vaccination, continuous efforts to design new vaccines are needed to fight "old" pandemics, such as tuberculosis and malaria, as well as emerging pathogens, such as Zika virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vaccination aims at reaching sterilizing immunity, however assessing vaccine efficacy is still challenging and underscores the need for a better understanding of immune protective responses. Identifying reliable predictive markers of immunogenicity can help to select and develop promising vaccine candidates during early preclinical studies and can lead to improved, personalized, vaccination strategies. A systems biology approach is increasingly being adopted to address these major challenges using multiple high-dimensional technologies combined with in silico models. Although the goal is to develop predictive models of vaccine efficacy in humans, applying this approach to animal models empowers basic and translational vaccine research. In this review, we provide an overview of vaccine immune signatures in preclinical models, as well as in target human populations. We also discuss high-throughput technologies used to probe vaccine-induced responses, along with data analysis and computational methodologies applied to the predictive modeling of vaccine efficacy.
Collapse
Affiliation(s)
- Matthieu Van Tilbeurgh
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Katia Lemdani
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Anne-Sophie Beignon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Nicolas Tchitchek
- Unité de Recherche i3, Inserm UMR-S 959, Bâtiment CERVI, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France;
| | - Lina Cheraitia
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Ernesto Marcos Lopez
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Quentin Pascal
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Pauline Maisonnasse
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Caroline Manet
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| |
Collapse
|
40
|
Shao F, Long Y, Ji H, Jiang D, Lei P, Lan X. Radionuclide-based molecular imaging allows CAR-T cellular visualization and therapeutic monitoring. Am J Cancer Res 2021; 11:6800-6817. [PMID: 34093854 PMCID: PMC8171102 DOI: 10.7150/thno.56989] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy is a new and effective form of adoptive cell therapy that is rapidly entering the mainstream for the treatment of CD19-positive hematological cancers because of its impressive effect and durable responses. Huge challenges remain in achieving similar success in patients with solid tumors. The current methods of monitoring CAR-T, including morphological imaging (CT and MRI), blood tests, and biopsy, have limitations to assess whether CAR-T cells are homing to tumor sites and infiltrating into tumor bed, or to assess the survival, proliferation, and persistence of CAR-T cells in solid tumors associated with an immunosuppressive microenvironment. Radionuclide-based molecular imaging affords improved CAR-T cellular visualization and therapeutic monitoring through either a direct cellular radiolabeling approach or a reporter gene imaging strategy, and endogenous cell imaging is beneficial to reflect functional information and immune status of T cells. Focusing on the dynamic monitoring and precise assessment of CAR-T therapy, this review summarizes the current applications of radionuclide-based noninvasive imaging in CAR-T cells visualization and monitoring and presents current challenges and strategic choices.
Collapse
|
41
|
Liberini V, Laudicella R, Capozza M, Huellner MW, Burger IA, Baldari S, Terreno E, Deandreis D. The Future of Cancer Diagnosis, Treatment and Surveillance: A Systemic Review on Immunotherapy and Immuno-PET Radiotracers. Molecules 2021; 26:2201. [PMID: 33920423 PMCID: PMC8069316 DOI: 10.3390/molecules26082201] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is an effective therapeutic option for several cancers. In the last years, the introduction of checkpoint inhibitors (ICIs) has shifted the therapeutic landscape in oncology and improved patient prognosis in a variety of neoplastic diseases. However, to date, the selection of the best patients eligible for these therapies, as well as the response assessment is still challenging. Patients are mainly stratified using an immunohistochemical analysis of the expression of antigens on biopsy specimens, such as PD-L1 and PD-1, on tumor cells, on peritumoral immune cells and/or in the tumor microenvironment (TME). Recently, the use and development of imaging biomarkers able to assess in-vivo cancer-related processes are becoming more important. Today, positron emission tomography (PET) with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) is used routinely to evaluate tumor metabolism, and also to predict and monitor response to immunotherapy. Although highly sensitive, FDG-PET in general is rather unspecific. Novel radiopharmaceuticals (immuno-PET radiotracers), able to identify specific immune system targets, are under investigation in pre-clinical and clinical settings to better highlight all the mechanisms involved in immunotherapy. In this review, we will provide an overview of the main new immuno-PET radiotracers in development. We will also review the main players (immune cells, tumor cells and molecular targets) involved in immunotherapy. Furthermore, we report current applications and the evidence of using [18F]FDG PET in immunotherapy, including the use of artificial intelligence (AI).
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/therapeutic use
- Artificial Intelligence
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- Fluorodeoxyglucose F18/administration & dosage
- Fluorodeoxyglucose F18/chemistry
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immune Checkpoint Inhibitors/chemistry
- Immune Checkpoint Inhibitors/metabolism
- Immunotherapy, Adoptive/methods
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Neoplasms/diagnostic imaging
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- Positron-Emission Tomography/methods
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Radiopharmaceuticals/administration & dosage
- Radiopharmaceuticals/chemical synthesis
- Signal Transduction
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Virginia Liberini
- Department of Medical Science, Division of Nuclear Medicine, University of Torino, 10126 Torino, Italy;
| | - Riccardo Laudicella
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, 98125 Messina, Italy; (R.L.); (S.B.)
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland; (M.W.H.); (I.A.B.)
| | - Martina Capozza
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (M.C.); (E.T.)
| | - Martin W. Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland; (M.W.H.); (I.A.B.)
| | - Irene A. Burger
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland; (M.W.H.); (I.A.B.)
- Department of Nuclear Medicine, Kantonsspital Baden, 5004 Baden, Switzerland
| | - Sergio Baldari
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, 98125 Messina, Italy; (R.L.); (S.B.)
| | - Enzo Terreno
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (M.C.); (E.T.)
| | - Désirée Deandreis
- Department of Medical Science, Division of Nuclear Medicine, University of Torino, 10126 Torino, Italy;
| |
Collapse
|
42
|
Jhiang SM, Cheng P, Nabhan FA, Sipos JA, Menq CH. Personalized radioiodine therapy for thyroid cancer patients with known disease. Fac Rev 2021; 10:36. [PMID: 33977289 PMCID: PMC8103907 DOI: 10.12703/r/10-36] [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] [Indexed: 11/30/2022] Open
Abstract
Radioactive iodine (RAI) 131I is a targeted therapy for patients with RAI-avid follicular cell-derived thyroid cancer. However, the responsiveness to 131I therapy varies among thyroid cancer patients mainly owing to differential RAI uptake and RAI radiosensitivity among patients’ lesions. A personalized approach to maximize 131I therapeutic efficacy is proposed based on recent scientific advances and future opportunities.
Collapse
Affiliation(s)
- Sissy M Jhiang
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Peng Cheng
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Fadi A Nabhan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Jennifer A Sipos
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Chia-Hsiang Menq
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
43
|
Sofias AM, Combes F, Koschmieder S, Storm G, Lammers T. A paradigm shift in cancer nanomedicine: from traditional tumor targeting to leveraging the immune system. Drug Discov Today 2021; 26:1482-1489. [PMID: 33617793 DOI: 10.1016/j.drudis.2021.02.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/26/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022]
Abstract
Twenty-five years after the approval of the first anticancer nanodrug, we have to start re(de)fining tumor-targeted drug delivery alongside advances in immuno-oncology. Given that cancer is characterized by an immunological imbalance that goes beyond the primary tumor, we should focus on targeting, engaging, and modulating cancer-associated immune cells in the tumor microenvironment (TME), circulation, and immune cell-enriched tissues. When designed and applied rationally, nanomedicines will assist in restoring the immunological equilibrium at the whole-body level, which holds potential not only for cancer therapy, but also for the treatment of a range of other disorders.
Collapse
Affiliation(s)
- Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | - Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Steffen Koschmieder
- Department of Medicine (Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation), Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Gert Storm
- Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands; Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands; Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
44
|
In vivo cell tracking with viral vector mediated genetic labeling. J Neurosci Methods 2020; 350:109021. [PMID: 33316318 DOI: 10.1016/j.jneumeth.2020.109021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022]
Abstract
Cell tracking is a useful technique to monitor specific cell populations for their morphology, development, proliferation, migration, interaction, function, and other properties, both in vitro and in vivo. Using different materials and methodologies to label the target cells directly or indirectly, the dynamic biological processes in living organisms can be visualized with appropriate detection techniques. Viruses, with the unique ability to deliver exogenous genes into host cells, have been used as vectors to mediate gene transfer. Genetic labeling of target cells by viral vectors endows the cells to express reporter genes with high efficiency and specificity. In conjunction with corresponding imaging techniques, cells labeled with different genetic reporters mediated by different viral vectors can be monitored across spatial and temporal scales to fulfill various purposes and address different questions. In the present review, we introduce the basic principle of viral vectors in cell tracking and highlight the examples of cell tracking in various research areas.
Collapse
|