1
|
Rebeck ON, Wallace MJ, Prusa J, Ning J, Evbuomwan EM, Rengarajan S, Habimana-Griffin L, Kwak S, Zahrah D, Tung J, Liao J, Mahmud B, Fishbein SRS, Ramirez Tovar ES, Mehta R, Wang B, Gorelik MG, Helmink BA, Dantas G. A yeast-based oral therapeutic delivers immune checkpoint inhibitors to reduce intestinal tumor burden. Cell Chem Biol 2025; 32:98-110.e7. [PMID: 39571582 PMCID: PMC11741927 DOI: 10.1016/j.chembiol.2024.10.013] [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: 03/15/2024] [Revised: 08/09/2024] [Accepted: 10/28/2024] [Indexed: 12/13/2024]
Abstract
Engineered probiotics are an emerging platform for in situ delivery of therapeutics to the gut. Herein, we developed an orally administered, yeast-based therapeutic delivery system to deliver next-generation immune checkpoint inhibitor (ICI) proteins directly to gastrointestinal tumors. We engineered Saccharomyces cerevisiae var. boulardii (Sb), a probiotic yeast with high genetic tractability and innate anticancer activity, to secrete "miniature" antibody variants that target programmed death ligand 1 (Sb_haPD-1). When tested in an ICI-refractory colorectal cancer (CRC) mouse model, Sb_haPD-1 significantly reduced intestinal tumor burden and resulted in significant shifts to the immune cell profile and microbiome composition. This oral therapeutic platform is modular and highly customizable, opening new avenues of targeted drug delivery that can be applied to treat a myriad of gastrointestinal malignancies.
Collapse
Affiliation(s)
- Olivia N Rebeck
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Miranda J Wallace
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jerome Prusa
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Esse M Evbuomwan
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Sunaina Rengarajan
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Dermatology, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, St. Louis MO 63110, USA
| | - LeMoyne Habimana-Griffin
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suryang Kwak
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David Zahrah
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jason Tung
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James Liao
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bejan Mahmud
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erick S Ramirez Tovar
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rehan Mehta
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark G Gorelik
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beth A Helmink
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
2
|
Binder U, Skerra A. Strategies for extending the half-life of biotherapeutics: successes and complications. Expert Opin Biol Ther 2025; 25:93-118. [PMID: 39663567 DOI: 10.1080/14712598.2024.2436094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION Engineering of the drug half-life in vivo has become an integral part of modern biopharmaceutical development due to the fact that many proteins/peptides with therapeutic potential are quickly cleared by kidney filtration after injection and, thus, circulate only a few hours in humans (or just minutes in mice). AREAS COVERED Looking at the growing list of clinically approved biologics that have been modified for prolonged activity, and also the plethora of such drugs under preclinical and clinical development, it is evident that not one solution fits all needs, owing to the vastly different structural features and functional properties of the pharmacologically active entities. This article provides an overview of established half-life extension strategies, as well as of emerging novel concepts for extending the in vivo stability of biologicals, and their pros and cons. EXPERT OPINION Beyond the classical and still dominating technologies for improving drug pharmacokinetics and bioavailability, Fc fusion and PEGylation, various innovative approaches that offer advantages in different respects have entered the clinical stage. While the Fc fusion partner may be gradually superseded by engineered albumin-binding domains, chemical PEGylation may be replaced by biodegradable recombinant amino-acid polymers like PASylation, thus also offering a purely biotechnological manufacturing route.
Collapse
Affiliation(s)
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Freising, Germany
| |
Collapse
|
3
|
Kastner AL, Marx AF, Dimitrova M, Abreu-Mota T, Ertuna YI, Bonilla WV, Stauffer K, Künzli M, Wagner I, Kreutzfeldt M, Merkler D, Pinschewer DD. Durable lymphocyte subset elimination upon a single dose of AAV-delivered depletion antibody dissects immune control of chronic viral infection. Immunity 2024:S1074-7613(24)00536-3. [PMID: 39719711 DOI: 10.1016/j.immuni.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/02/2024] [Accepted: 11/26/2024] [Indexed: 12/26/2024]
Abstract
To interrogate the role of specific immune cells in infection, cancer, and autoimmunity, immunologists commonly use monoclonal depletion antibodies (depletion-mAbs) or genetically engineered mouse models (GEMMs). To generate a tool that combines specific advantages and avoids select drawbacks of the two methods, we engineered adeno-associated viral vectors expressing depletion mAbs (depletion-AAVs). Single-dose depletion-AAV administration durably eliminated lymphocyte subsets in mice and avoided accessory deficiencies of GEMMs, such as marginal zone defects in B cell-deficient animals. Depletion-AAVs can be used in animals of different genetic backgrounds, and multiple depletion-AAVs can readily be combined. Exploiting depletion-AAV technology, we showed that B cells were required for unimpaired CD4+ and CD8+ T cell responses to chronic lymphocytic choriomeningitis virus (LCMV) infection. Upon B cell depletion, CD8+ T cells failed to suppress viremia, and they only helped resolve chronic infection when antibodies dampened viral loads. Our study positions depletion-AAVs as a versatile tool for immunological research.
Collapse
Affiliation(s)
- Anna Lena Kastner
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | | | - Mirela Dimitrova
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Tiago Abreu-Mota
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Yusuf I Ertuna
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Weldy V Bonilla
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Karsten Stauffer
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Marco Künzli
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1206 Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1206 Geneva, Switzerland
| | | |
Collapse
|
4
|
Crawford MW, Abdelwahab WM, Siram K, Parkins CJ, Harrison HF, Osman SR, Schweitzer D, Evans JT, Burkhart DJ, Pinto AK, Brien JD, Smith JL, Hirsch AJ. The TLR7/8 agonist INI-4001 enhances the immunogenicity of a Powassan virus-like-particle vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.28.625832. [PMID: 39677812 PMCID: PMC11642962 DOI: 10.1101/2024.11.28.625832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Powassan virus (POWV) is a pathogenic tick-borne flavivirus that causes fatal neuroinvasive disease in humans. There are currently no approved therapies or vaccines for POWV infection. Here, we develop a POW virus-like-particle (POW-VLP) based vaccine adjuvanted with the novel synthetic Toll-like receptor 7/8 agonist INI-4001. We demonstrate that INI-4001 outperforms both alum and the Toll-like receptor 4 agonist INI-2002 in enhancing the immunogenicity of a dose-sparing POW-VLP vaccine in mice. INI-4001 increases the magnitude and breadth of the antibody response as measured by whole-virus ELISA, induces neutralizing antibodies measured by FRNT, reduces viral burden in the brain of infected mice measured by RT qPCR, and confers 100% protection from lethal challenge with both lineages of POWV. We show that the antibody response induced by INI-4001 is more durable than standard alum, and 80% of mice remain protected from lethal challenge 9-months post-vaccination. Lastly, we show that the protection elicited by INI-4001 adjuvanted POW-VLP vaccine is unaffected by either CD4+ or CD8+ T cell depletion and can be passively transferred to unvaccinated mice indicating that protection is mediated through humoral immunity. This study highlights the utility of novel synthetic adjuvants in VLP-based vaccines.
Collapse
Affiliation(s)
- Michael W. Crawford
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Walid M. Abdelwahab
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
- Center for Translational Medicine – Adjuvant Research Team, University of Montana, Missoula, MT, USA
| | - Karthik Siram
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
- Center for Translational Medicine – Adjuvant Research Team, University of Montana, Missoula, MT, USA
| | - Christopher J. Parkins
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Henry F. Harrison
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Samantha R. Osman
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Dillon Schweitzer
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
- Center for Translational Medicine – Adjuvant Research Team, University of Montana, Missoula, MT, USA
| | - Jay T. Evans
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
- Center for Translational Medicine – Adjuvant Research Team, University of Montana, Missoula, MT, USA
| | - David J. Burkhart
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
- Center for Translational Medicine – Adjuvant Research Team, University of Montana, Missoula, MT, USA
| | - Amelia K. Pinto
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - James D. Brien
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jessica L. Smith
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Alec J. Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
5
|
Xiang Y, Xu J, McGovern BL, Ranzenigo A, Huang W, Sang Z, Shen J, Diaz-Tapia R, Pham ND, Teunissen AJP, Rodriguez ML, Benjamin J, Taylor DJ, van Leent MMT, White KM, García-Sastre A, Zhang P, Shi Y. Adaptive multi-epitope targeting and avidity-enhanced nanobody platform for ultrapotent, durable antiviral therapy. Cell 2024; 187:6966-6980.e23. [PMID: 39447570 PMCID: PMC11748749 DOI: 10.1016/j.cell.2024.09.043] [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: 02/26/2024] [Revised: 07/30/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
Pathogens constantly evolve and can develop mutations that evade host immunity and treatment. Addressing these escape mechanisms requires targeting evolutionarily conserved vulnerabilities, as mutations in these regions often impose fitness costs. We introduce adaptive multi-epitope targeting with enhanced avidity (AMETA), a modular and multivalent nanobody platform that conjugates potent bispecific nanobodies to a human immunoglobulin M (IgM) scaffold. AMETA can display 20+ nanobodies, enabling superior avidity binding to multiple conserved and neutralizing epitopes. By leveraging multi-epitope SARS-CoV-2 nanobodies and structure-guided design, AMETA constructs exponentially enhance antiviral potency, surpassing monomeric nanobodies by over a million-fold. These constructs demonstrate ultrapotent, broad, and durable efficacy against pathogenic sarbecoviruses, including Omicron sublineages, with robust preclinical results. Structural analysis through cryoelectron microscopy and modeling has uncovered multiple antiviral mechanisms within a single construct. At picomolar to nanomolar concentrations, AMETA efficiently induces inter-spike and inter-virus cross-linking, promoting spike post-fusion and striking viral disarmament. AMETA's modularity enables rapid, cost-effective production and adaptation to evolving pathogens.
Collapse
Affiliation(s)
- Yufei Xiang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jialu Xu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Briana L McGovern
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anna Ranzenigo
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Wei Huang
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Zhe Sang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Juan Shen
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Randy Diaz-Tapia
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ngoc Dung Pham
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abraham J P Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - M Luis Rodriguez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jared Benjamin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Derek J Taylor
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Mandy M T van Leent
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kris M White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Peijun Zhang
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK; Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK; Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK.
| | - Yi Shi
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
6
|
York ES, Dratch BD, Ito J, Horwitz SM, Emamian S, Ambarian JA, Gill S, Jones J, Chonat S, Lollar P, Meeks SL, Davis KM, Batsuli G. Persistent splenic-derived IgMs preferentially recognize factor VIII A2 and C2 domain epitopes but do not alter antibody production. J Thromb Haemost 2024:S1538-7836(24)00633-0. [PMID: 39476969 DOI: 10.1016/j.jtha.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND The most significant treatment complication for patients with hemophilia A is the development of neutralizing immunoglobins (Igs) G, termed inhibitors, against factor VIII (FVIII), which prevent FVIII replacement therapy. Low titers of FVIII-specific IgMs have been identified in hemophilia A patients with and without inhibitors, as well as in healthy individuals. However, the duration and influence of IgMs on the immune response to FVIII remains unclear. OBJECTIVES To characterize the binding interactions of persistently secreted FVIII-specific IgMs in hemophilia A mice and assess their effect on IgG antibody development. METHODS Splenic-derived monoclonal antibodies (mAbs) from immunized FVIII knockout mice were isolated and purified using hybridoma technology. Binding interactions were assessed utilizing a novel fluid-phase enzyme-linked immunosorbent assay and computational modeling with High Ambiguity-Driven protein-protein DOCKing to account for weak IgM binding. RESULTS Sixteen porcine cross-reactive and noninhibitory FVIII-specific IgM mAbs were identified. RNA sequencing of FVIII-specific IgMs revealed 13 unique variable, diversity, and joining (VDJ)/variable and joining (VJ) sequences indicating derivation from 13 unique B cell clones. The IgMs demonstrated polyclonal and polyreactive binding to FVIII in vitro and in silico. Molecular docking studies with reconstructed IgM variable, diversity, and joining/variable and joining regions identified frequent IgM interactions with amino acid residues K376, T381, K437, R2215, or K2249 within the FVIII A2 and C2 domains. Injections of individual IgMs prior to FVIII exposure and co-injection of FVIII/IgM immune complexes did not affect de novo FVIII antibody production. CONCLUSION Persistent FVIII-specific IgMs are polyclonal but preferentially bind the A2 and C2 domains. FVIII/IgM immune complex formation does not significantly alter inhibitor development.
Collapse
Affiliation(s)
- Elizabeth S York
- Department of Pediatrics, Stanford University, Palo Alto, California, USA; Department of Pediatrics, Emory University, Atlanta, Georgia, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | - Jasmine Ito
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | - Sahand Emamian
- Department of Physics, Emory University, Atlanta, Georgia, USA
| | | | - Surinder Gill
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Jayre Jones
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Satheesh Chonat
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Pete Lollar
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Shannon L Meeks
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | - Glaivy Batsuli
- Department of Pediatrics, Stanford University, Palo Alto, California, USA; Department of Pediatrics, Emory University, Atlanta, Georgia, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.
| |
Collapse
|
7
|
Grace PS, Peters JM, Sixsmith J, Lu R, Luedeman C, Fenderson BA, Vickers A, Slein MD, Irvine EB, McKitrick T, Wei MH, Cummings RD, Wallace A, Cavacini LA, Choudhary A, Proulx MK, Sundling C, Källenius G, Reljic R, Ernst JD, Casadevall A, Locht C, Pinter A, Sasseti CM, Bryson BD, Fortune SM, Alter G. Antibody-Fab and -Fc features promote Mycobacterium tuberculosis restriction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617070. [PMID: 39416184 PMCID: PMC11482752 DOI: 10.1101/2024.10.07.617070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a leading cause of death by an infectious disease globally, with no efficacious vaccine. Antibodies are implicated in Mtb control, but the mechanisms of antibody action remain poorly understood. We assembled a library of TB monoclonal antibodies (mAb) and screened for the ability to restrict Mtb in mice, identifying protective antibodies targeting known and novel antigens. To dissect the mechanism of mAb-mediated Mtb restriction, we optimized a protective lipoarabinomannan-specific mAb through Fc-swapping. In vivo analysis of these Fc-variants revealed a critical role for Fc-effector function in Mtb restriction. Restrictive Fc-variants altered distribution of Mtb across innate immune cells. Single-cell transcriptomics highlighted distinctly activated molecular circuitry within innate immune cell subpopulations, highlighting early activation of neutrophils as a key signature of mAb-mediated Mtb restriction. Therefore, improved antibody-mediated restriction of Mtb is associated with reorganization of the tissue-level immune response to infection and depends on the collaboration of antibody Fab and Fc.
Collapse
|
8
|
Fernández-Gallego N, Castillo-González R, Moreno-Serna L, García-Cívico AJ, Sánchez-Martínez E, López-Sanz C, Fontes AL, Pimentel LL, Gradillas A, Obeso D, Neuhaus R, Ramírez-Huesca M, Ruiz-Fernández I, Nuñez-Borque E, Carrasco YR, Ibáñez B, Martín P, Blanco C, Barbas C, Barber D, Rodríguez-Alcalá LM, Villaseñor A, Esteban V, Sánchez-Madrid F, Jiménez-Saiz R. Allergic inflammation triggers dyslipidemia via IgG signalling. Allergy 2024; 79:2680-2699. [PMID: 38864116 DOI: 10.1111/all.16187] [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: 09/14/2023] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Allergic diseases begin early in life and are often chronic, thus creating an inflammatory environment that may precede or exacerbate other pathologies. In this regard, allergy has been associated to metabolic disorders and with a higher risk of cardiovascular disease, but the underlying mechanisms remain incompletely understood. METHODS We used a murine model of allergy and atherosclerosis, different diets and sensitization methods, and cell-depleting strategies to ascertain the contribution of acute and late phase inflammation to dyslipidemia. Untargeted lipidomic analyses were applied to define the lipid fingerprint of allergic inflammation at different phases of allergic pathology. Expression of genes related to lipid metabolism was assessed in liver and adipose tissue at different times post-allergen challenge. Also, changes in serum triglycerides (TGs) were evaluated in a group of 59 patients ≥14 days after the onset of an allergic reaction. RESULTS We found that allergic inflammation induces a unique lipid signature that is characterized by increased serum TGs and changes in the expression of genes related to lipid metabolism in liver and adipose tissue. Alterations in blood TGs following an allergic reaction are independent of T-cell-driven late phase inflammation. On the contrary, the IgG-mediated alternative pathway of anaphylaxis is sufficient to induce a TG increase and a unique lipid profile. Lastly, we demonstrated an increase in serum TGs in 59 patients after undergoing an allergic reaction. CONCLUSION Overall, this study reveals that IgG-mediated allergic inflammation regulates lipid metabolism.
Collapse
Affiliation(s)
- Nieves Fernández-Gallego
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Raquel Castillo-González
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Ophthalmology and Ear, Nose and Throat (ENT), Universidad Complutense de Madrid, Madrid, Spain
| | - Lucía Moreno-Serna
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Antonio J García-Cívico
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Elisa Sánchez-Martínez
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Celia López-Sanz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Luiza Fontes
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Lígia L Pimentel
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Ana Gradillas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - David Obeso
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - René Neuhaus
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | | | | | - Emilio Nuñez-Borque
- Department of Allergy and Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Yolanda R Carrasco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Cardiology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Blanco
- Department of Allergy, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Domingo Barber
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Luis M Rodríguez-Alcalá
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Alma Villaseñor
- Department of Basic Medical Sciences, Faculty of Medicine, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Rodrigo Jiménez-Saiz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
- Department of Medicine, McMaster Immunology Research Centre (MIRC), Schroeder Allergy and Immunology Research Institute (SAIRI), McMaster University, Hamilton, Ontario, Canada
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| |
Collapse
|
9
|
Vecchione A, Devlin JC, Tasker C, Ramnarayan VR, Haase P, Conde E, Srivastava D, Atwal GS, Bruhns P, Murphy AJ, Sleeman MA, Limnander A, Lim WK, Asrat S, Orengo JM. IgE plasma cells are transcriptionally and functionally distinct from other isotypes. Sci Immunol 2024; 9:eadm8964. [PMID: 39241058 DOI: 10.1126/sciimmunol.adm8964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/18/2024] [Accepted: 08/08/2024] [Indexed: 09/08/2024]
Abstract
Understanding the phenotypic and transcriptional signature of immunoglobulin E (IgE)-producing cells is fundamental to plasma cell (PC) biology and development of therapeutic interventions for allergy. Here, using a mouse model of intranasal house dust mite (HDM) exposure, we showed that short-lived IgE PCs emerge in lung draining lymph nodes (dLNs) during early exposure (<3 weeks) and long-lived IgE PCs accumulate in the bone marrow (BM) with prolonged exposure (>7 weeks). IgE PCs had distinct surface and gene expression profiles in these different tissues compared with other Ig isotypes. IgE BMPCs up-regulated genes associated with prosurvival and BM homing, whereas IgE dLN PCs expressed genes associated with recent class switching and differentiation. IgE PCs also exhibited higher expression of endoplasmic reticulum (ER) stress and protein coding genes and higher antibody secretion rate when compared with IgG1. Overall, this study highlights the unique developmental path and transcriptional signature of short-lived and long-lived IgE PCs.
Collapse
Affiliation(s)
| | | | - Carley Tasker
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Venkat Raman Ramnarayan
- Institut Pasteur, Université Paris Cité, Inserm UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Paul Haase
- Institut Pasteur, Université Paris Cité, Inserm UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Eva Conde
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | - Pierre Bruhns
- Institut Pasteur, Université Paris Cité, Inserm UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | | | | | | | - Wei Keat Lim
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Jamie M Orengo
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| |
Collapse
|
10
|
Erlandson S, Wang J, Jiang H, Osei-Owusu J, Rockman HA, Kruse AC. Engineering and Characterization of a Long-Half-Life Relaxin Receptor RXFP1 Agonist. Mol Pharm 2024; 21:4441-4449. [PMID: 39134056 PMCID: PMC11372834 DOI: 10.1021/acs.molpharmaceut.4c00368] [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: 04/04/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 09/03/2024]
Abstract
Relaxin-2 is a peptide hormone with important roles in human cardiovascular and reproductive biology. Its ability to activate cellular responses such as vasodilation, angiogenesis, and anti-inflammatory and antifibrotic effects has led to significant interest in using relaxin-2 as a therapeutic for heart failure and several fibrotic conditions. However, recombinant relaxin-2 has a very short serum half-life, limiting its clinical applications. Here, we present protein engineering efforts targeting the relaxin-2 hormone in order to increase its serum half-life while maintaining its ability to activate the G protein-coupled receptor RXFP1. To achieve this, we optimized a fusion between relaxin-2 and an antibody Fc fragment, generating a version of the hormone with a circulating half-life of around 3 to 5 days in mice while retaining potent agonist activity at the RXFP1 receptor both in vitro and in vivo.
Collapse
Affiliation(s)
- Sarah
C. Erlandson
- Department
of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jialu Wang
- Department
of Medicine, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Haoran Jiang
- Department
of Medicine, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - James Osei-Owusu
- Department
of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Howard A. Rockman
- Department
of Medicine, Duke University Medical Center, Durham, North Carolina 27710, United States
- Department
of Cell Biology, Duke University Medical
Center, Durham, North Carolina 27710, United States
| | - Andrew C. Kruse
- Department
of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
11
|
Lai J, Shah S, Martinez-Orengo N, Knight R, Alemu E, Turner ML, Wang B, Lyndaker A, Shi J, Basuli F, Hammoud DA. PET imaging of Aspergillus infection using Zirconium-89 labeled anti-β-glucan antibody fragments. Eur J Nucl Med Mol Imaging 2024; 51:3223-3234. [PMID: 38787397 PMCID: PMC11368974 DOI: 10.1007/s00259-024-06760-4] [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: 11/24/2023] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Invasive fungal diseases, such as pulmonary aspergillosis, are common life-threatening infections in immunocompromised patients and effective treatment is often hampered by delays in timely and specific diagnosis. Fungal-specific molecular imaging ligands can provide non-invasive readouts of deep-seated fungal pathologies. In this study, the utility of antibodies and antibody fragments (Fab) targeting β-glucans in the fungal cell wall to detect Aspergillus infections was evaluated both in vitro and in preclinical mouse models. METHODS The binding characteristics of two commercially available β-glucan antibody clones and their respective antigen-binding Fabs were tested using biolayer interferometry (BLI) assays and immunofluorescence staining. In vivo binding of the Zirconium-89 labeled antibodies/Fabs to fungal pathogens was then evaluated using PET/CT imaging in mouse models of fungal infection, bacterial infection and sterile inflammation. RESULTS One of the evaluated antibodies (HA-βG-Ab) and its Fab (HA-βG-Fab) bound to β-glucans with high affinity (KD = 0.056 & 21.5 nM respectively). Binding to the fungal cell wall was validated by immunofluorescence staining and in vitro binding assays. ImmunoPET imaging with intact antibodies however showed slow clearance and high background signal as well as nonspecific accumulation in sites of infection/inflammation. Conversely, specific binding of [89Zr]Zr-DFO-HA-βG-Fab to sites of fungal infection was observed when compared to the isotype control Fab and was significantly higher in fungal infection than in bacterial infection or sterile inflammation. CONCLUSIONS [89Zr]Zr-DFO-HA-βG-Fab can be used to detect fungal infections in vivo. Targeting distinct components of the fungal cell wall is a viable approach to developing fungal-specific PET tracers.
Collapse
Affiliation(s)
- Jianhao Lai
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), 10 Center Drive, Room 1C368, Bethesda, MD, 20892, USA
| | - Swati Shah
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), 10 Center Drive, Room 1C368, Bethesda, MD, 20892, USA
| | - Neysha Martinez-Orengo
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), 10 Center Drive, Room 1C368, Bethesda, MD, 20892, USA
| | - Rekeya Knight
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), 10 Center Drive, Room 1C368, Bethesda, MD, 20892, USA
| | - Eyob Alemu
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), 10 Center Drive, Room 1C368, Bethesda, MD, 20892, USA
| | - Mitchell L Turner
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), 10 Center Drive, Room 1C368, Bethesda, MD, 20892, USA
| | - Benjamin Wang
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), 10 Center Drive, Room 1C368, Bethesda, MD, 20892, USA
| | - Anna Lyndaker
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), 10 Center Drive, Room 1C368, Bethesda, MD, 20892, USA
| | - Jianfeng Shi
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute (NHLBI), NIH, Rockville, MD, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute (NHLBI), NIH, Rockville, MD, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), 10 Center Drive, Room 1C368, Bethesda, MD, 20892, USA.
| |
Collapse
|
12
|
Humphries J, Fletcher NL, Sonderegger SE, Bell CA, Kempe K, Thurecht KJ. Mitigating the Effects of Persistent Antipolymer Immune Reactions in Nanomedicine: Evaluating Materials-Based Approaches Using Molecular Imaging. ACS NANO 2024. [PMID: 39037055 DOI: 10.1021/acsnano.4c07317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Poly(ethylene glycol) (PEG) is a hydrophilic polymer ubiquitously used in both medical and nonmedical goods. Recent debate surrounding the observed stimulation of immune responses against PEG has spurred the development of materials that may be suitable replacements for this common polymeric component. The underlying view is that these alternative materials with comparable physicochemical properties can overcome the unfavorable and unpredictable effects of antibody-mediated clearance by being chemically, and therefore antigenically, distinct from PEG. However, this hypothesis has not been thoroughly tested in any defined manner, and the immune response observed against PEG has not been rigorously investigated within the context of these emerging materials. Consequently, it remains unclear whether immunity-mediated discrimination between polymeric entities even occurs in vivo and, if this is the case, how it may be exploited. In this study, we utilize positron emission tomography-computed tomography molecular imaging in mice immunized to develop specific antibody responses to PEG and an alternative polymer in order to visualize and quantify the influence of antipolymer antibodies on the biodistribution of synthetic polymers in vivo as a function of immunization status. Under the conditions of this experiment, mice could be primed to exhibit both innate and adaptive immunity to all of the polymer systems to which they were exposed. We demonstrate that alternating between chemically disparate polymers is a viable approach to extend their efficacy when antipolymer humoral immune responses arise.
Collapse
Affiliation(s)
- James Humphries
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Stefan E Sonderegger
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Craig A Bell
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
13
|
Gavade A, Nagraj AK, Patel R, Pais R, Dhanure P, Scheele J, Seiz W, Patil J. Understanding the Specific Implications of Amino Acids in the Antibody Development. Protein J 2024; 43:405-424. [PMID: 38724751 DOI: 10.1007/s10930-024-10201-4] [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] [Accepted: 04/21/2024] [Indexed: 06/01/2024]
Abstract
As the demand for immunotherapy to treat and manage cancers, infectious diseases and other disorders grows, a comprehensive understanding of amino acids and their intricate role in antibody engineering has become a prime requirement. Naturally produced antibodies may not have the most suitable amino acids at the complementarity determining regions (CDR) and framework regions, for therapeutic purposes. Therefore, to enhance the binding affinity and therapeutic properties of an antibody, the specific impact of certain amino acids on the antibody's architecture must be thoroughly studied. In antibody engineering, it is crucial to identify the key amino acid residues that significantly contribute to improving antibody properties. Therapeutic antibodies with higher binding affinity and improved functionality can be achieved through modifications or substitutions with highly suitable amino acid residues. Here, we have indicated the frequency of amino acids and their association with the binding free energy in CDRs. The review also analyzes the experimental outcome of two studies that reveal the frequency of amino acids in CDRs and provides their significant correlation between the outcomes. Additionally, it discusses the various bond interactions within the antibody structure and antigen binding. A detailed understanding of these amino acid properties should assist in the analysis of antibody sequences and structures needed for designing and enhancing the overall performance of therapeutic antibodies.
Collapse
Affiliation(s)
- Akshata Gavade
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Anil Kumar Nagraj
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Riya Patel
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Roylan Pais
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Pratiksha Dhanure
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | | | | | - Jaspal Patil
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India.
| |
Collapse
|
14
|
Morimoto K, Takahashi R, Takahashi G, Miyajima M, Nakajima K. Maternal immunoglobulins are distributed in the offspring's brain to support the maintenance of cortical interneurons in the postnatal period. Inflamm Regen 2024; 44:24. [PMID: 38750507 PMCID: PMC11094934 DOI: 10.1186/s41232-024-00336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
It is known that maternal immunoglobulins (Igs) are transferred to the offspring across the placenta. However, receiving maternal Igs, especially before the blood-brain barrier (BBB) is formed in the offspring's brain, carries the risk of transferring some brain-reactive Igs. It is thus hypothesized that there may be some unknown benefit to the offspring's brain that overweighs this risk. In this study, we show that the Ig detected in the embryonic/perinatal mouse brain is IgG not produced by the pups themselves, but is basically transferred from the mother across the placenta using the neonatal Fc receptor (FcRn) during embryonic stages. The amount of IgG in the brain gradually decreases after birth, and almost disappears within 3 weeks postnatally. IgG is detected on axon bundles, microglia, and some meningeal cells, including border-associated macrophages (BAMs), endothelial cells, and fibroblasts. Using Fcer1g knock-out (KO) mice, we show that BAMs and microglia receive maternal IgG in an Fc receptor γ chain (FcRγ)-dependent manner, but IgG on other meningeal cells and axon bundles is received independently of the FcRγ. These results suggest that maternal IgG may be used in multiple ways by different mechanisms. In maternal IgG-deficient mice, the number of interneurons in the cerebral cortex is not altered around birth but is reduced postnatally, suggesting that receipt of maternal IgG is necessary for the maintenance of cortical interneurons in the postnatal period. These data suggest that maternal IgG has an important function in the developing brain, where neither obvious inflammation nor infection is observed.
Collapse
Affiliation(s)
- Keiko Morimoto
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| | - Rikuo Takahashi
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Goro Takahashi
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Michio Miyajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
15
|
Tuz AA, Ghosh S, Karsch L, Ttoouli D, Sata SP, Ulusoy Ö, Kraus A, Hoerenbaum N, Wolf JN, Lohmann S, Zwirnlein F, Kaygusuz V, Lakovic V, Tummes HL, Beer A, Gallert M, Thiebes S, Qefalia A, Cibir Z, Antler M, Korste S, Haj Yehia E, Michel L, Rassaf T, Kaltwasser B, Abdelrahman H, Mohamud Yusuf A, Wang C, Yin D, Haeusler L, Lueong S, Richter M, Engel DR, Stenzel M, Soehnlein O, Frank B, Solo-Nomenjanahary M, Ho-Tin-Noé B, Siveke JT, Totzeck M, Hoffmann D, Grüneboom A, Hagemann N, Hasenberg A, Desilles JP, Mazighi M, Sickmann A, Chen J, Hermann DM, Gunzer M, Singh V. Stroke and myocardial infarction induce neutrophil extracellular trap release disrupting lymphoid organ structure and immunoglobulin secretion. NATURE CARDIOVASCULAR RESEARCH 2024; 3:525-540. [PMID: 39195931 PMCID: PMC11358010 DOI: 10.1038/s44161-024-00462-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 03/12/2024] [Indexed: 08/29/2024]
Abstract
Post-injury dysfunction of humoral immunity accounts for infections and poor outcomes in cardiovascular diseases. Among immunoglobulins (Ig), IgA, the most abundant mucosal antibody, is produced by plasma B cells in intestinal Peyer's patches (PP) and lamina propria. Here we show that patients with stroke and myocardial ischemia (MI) had strongly reduced IgA blood levels. This was phenocopied in experimental mouse models where decreased plasma and fecal IgA were accompanied by rapid loss of IgA-producing plasma cells in PP and lamina propria. Reduced plasma IgG was detectable in patients and experimental mice 3-10 d after injury. Stroke/MI triggered the release of neutrophil extracellular traps (NETs). Depletion of neutrophils, NET degradation or blockade of NET release inhibited the loss of IgA+ cells and circulating IgA in experimental stroke and MI and in patients with stroke. Our results unveil how tissue-injury-triggered systemic NET release disrupts physiological Ig secretion and how this can be inhibited in patients.
Collapse
Affiliation(s)
- Ali A Tuz
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Susmita Ghosh
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., Dortmund, Germany
| | - Laura Karsch
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Dimitris Ttoouli
- Bioinformatics and Computational Biophysics, Faculty of Biology and Centre for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany
| | - Sai P Sata
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., Dortmund, Germany
| | - Özgür Ulusoy
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Andreas Kraus
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Nils Hoerenbaum
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Jan-Niklas Wolf
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Sabrina Lohmann
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Franziska Zwirnlein
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Viola Kaygusuz
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Vivian Lakovic
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Hannah-Lea Tummes
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Alexander Beer
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Markus Gallert
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Stephanie Thiebes
- Department of Immunodynamics, Institute of Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Altea Qefalia
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Zülal Cibir
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Medina Antler
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Sebastian Korste
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Elias Haj Yehia
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Britta Kaltwasser
- Department of Neurology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Hossam Abdelrahman
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Ayan Mohamud Yusuf
- Department of Neurology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Chen Wang
- Department of Neurology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Dongpei Yin
- Department of Neurology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Lars Haeusler
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Smiths Lueong
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mathis Richter
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZMBE), Universität Münster, Münster, Germany
| | - Daniel R Engel
- Department of Immunodynamics, Institute of Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Martin Stenzel
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., Dortmund, Germany
| | - Oliver Soehnlein
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZMBE), Universität Münster, Münster, Germany
| | - Benedikt Frank
- Department of Neurology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Mialitiana Solo-Nomenjanahary
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, U1144 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Benoît Ho-Tin-Noé
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, U1144 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Jens T Siveke
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Daniel Hoffmann
- Bioinformatics and Computational Biophysics, Faculty of Biology and Centre for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany
| | - Anika Grüneboom
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., Dortmund, Germany
| | - Nina Hagemann
- Department of Neurology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Anja Hasenberg
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Jean-Philippe Desilles
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, U1144 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Interventional Neuroradiology Department and Biological Resources Center, Rothschild Foundation Hospital, Paris, France
| | - Mikael Mazighi
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, U1144 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Interventional Neuroradiology Department and Biological Resources Center, Rothschild Foundation Hospital, Paris, France
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., Dortmund, Germany
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jianxu Chen
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., Dortmund, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany.
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., Dortmund, Germany.
| | - Vikramjeet Singh
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
16
|
Kim YS, Jeong YS, Bae GH, Kang JH, Lee M, Zabel BA, Bae YS. CD200R high neutrophils with dysfunctional autophagy establish systemic immunosuppression by increasing regulatory T cells. Cell Mol Immunol 2024; 21:349-361. [PMID: 38311677 PMCID: PMC10978921 DOI: 10.1038/s41423-024-01136-y] [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: 04/24/2023] [Revised: 12/21/2023] [Accepted: 01/13/2024] [Indexed: 02/06/2024] Open
Abstract
Distinct neutrophil populations arise during certain pathological conditions. The generation of dysfunctional neutrophils during sepsis and their contribution to septicemia-related systemic immune suppression remain unclear. In this study, using an experimental sepsis model that features immunosuppression, we identified a novel population of pathogenic CD200Rhigh neutrophils that are generated during the initial stages of sepsis and contribute to systemic immune suppression by enhancing regulatory T (Treg) cells. Compared to their CD200Rlow counterparts, sepsis-generated CD200Rhigh neutrophils exhibit impaired autophagy and dysfunction, with reduced chemotactic migration, superoxide anion production, and TNF-α production. Increased soluble CD200 blocks autophagy and neutrophil maturation in the bone marrow during experimental sepsis, and recombinant CD200 treatment in vitro can induce neutrophil dysfunction similar to that observed in CD200Rhigh neutrophils. The administration of an α-CD200R antibody effectively reversed neutrophil dysfunction by enhancing autophagy and protecting against a secondary infection challenge, leading to increased survival. Transcriptome analysis revealed that CD200Rhigh neutrophils expressed high levels of Igf1, which elicits the generation of Treg cells, while the administration of an α-CD200R antibody inhibited Treg cell generation in a secondary infection model. Taken together, our findings revealed a novel CD200Rhigh neutrophil population that mediates the pathogenesis of sepsis-induced systemic immunosuppression by generating Treg cells.
Collapse
Affiliation(s)
- Ye Seon Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yu Sun Jeong
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Geon Ho Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Ji Hyeon Kang
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Mingyu Lee
- Department of Health Science and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research, Veterans Affairs Hospital, Palo Alto, CA, 94304, USA
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Department of Health Science and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea.
| |
Collapse
|
17
|
Riedl R, Kühn A, Hupfer Y, Hebecker B, Peltner LK, Jordan PM, Werz O, Lorkowski S, Wiegand C, Wallert M. Characterization of Different Inflammatory Skin Conditions in a Mouse Model of DNCB-Induced Atopic Dermatitis. Inflammation 2024; 47:771-788. [PMID: 38150167 DOI: 10.1007/s10753-023-01943-x] [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: 11/10/2023] [Revised: 11/10/2023] [Accepted: 12/04/2023] [Indexed: 12/28/2023]
Abstract
The mouse model of 2,4-dinitrochlorbenzene (DNCB)-induced human-like atopic dermatitis (hlAD) has been widely used to test novel treatment strategies and compounds. However, the study designs and methods are highly diverse, presenting different hlAD disease patterns that occur after sensitization and repeated challenge with DNCB on dorsal skin. In addition, there is a lack of information about the progression of the disease during the experiment and the achieved pheno- and endotypes, especially at the timepoint when therapeutic treatment is initiated. We here examine hlAD in a DNCB-induced BALB/cJRj model at different timepoints: (i) before starting treatment with dexamethasone, representing a standard drug control (day 12) and (ii) at the end of the experiment (day 22). Both timepoints display typical AD-associated characteristics: skin thickening, spongiosis, hyper- and parakeratosis, altered cytokine and gene expression, increased lipid mediator formation, barrier protein and antimicrobial peptide abnormalities, as well as lymphoid organ hypertrophy. Increased mast cell infiltration into the skin and elevated immunoglobulin E plasma concentrations indicate a type I allergy response. The DNCB-treated skin showed an extrinsic moderate sub-acute hlAD lesion at day 12 and an extrinsic mild sub-acute to chronic pheno- and endotype at day 22 with a dominating Th2 response. A dependency of the filaggrin formation and expression in correlation to the disease severity in the DNCB-treated skin was found. In conclusion, our study reveals a detailed classification of a hlAD at two timepoints with different inflammatory skin conditions and pheno- and endotypes, thereby providing a better understanding of the DNCB-induced hlAD model in BALB/cJRj mice.
Collapse
Affiliation(s)
- Rebecca Riedl
- Department of Dermatology, Dermatological Research Laboratory, Jena University Hospital, 07747, Jena, Germany
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Annika Kühn
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Yvonne Hupfer
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Betty Hebecker
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743, Jena, Germany
| | - Lukas K Peltner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743, Jena, Germany
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University, 07743, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University, 07743, Jena, Germany
| | - Stefan Lorkowski
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743, Jena, Germany
| | - Cornelia Wiegand
- Department of Dermatology, Dermatological Research Laboratory, Jena University Hospital, 07747, Jena, Germany
| | - Maria Wallert
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany.
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743, Jena, Germany.
| |
Collapse
|
18
|
Bartl S, Xie Y, Potluri N, Kesineni R, Hencak K, Cengio LD, Balazs K, Oueslati A, Parth M, Salhat N, Siddu A, Smrzka O, Cicchetti F, Straffler G, Hayden MR, Southwell AL. Reducing huntingtin by immunotherapy delays disease progression in a mouse model of Huntington disease. Neurobiol Dis 2024; 190:106376. [PMID: 38092268 DOI: 10.1016/j.nbd.2023.106376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/22/2023] Open
Abstract
In Huntington disease (HD), the mutant huntingtin (mtHTT) protein is the principal cause of pathological changes that initiate primarily along the cortico-striatal axis. mtHTT is ubiquitously expressed and there is, accordingly, growing recognition that HD is a systemic disorder with functional interplay between the brain and the periphery. We have developed a monoclonal antibody, C6-17, targeting an exposed region of HTT near the aa586 Caspase 6 cleavage site. As recently published, mAB C6-17 can block cell-to-cell propagation of mtHTT in vitro. In order to reduce the burden of the mutant protein in vivo, we queried whether extracellular mtHTT could be therapeutically targeted in YAC128 HD mice. In a series of proof of concept experiments, we found that systemic mAB C6-17 treatment resulted in the distribution of the mAB C6-17 to peripheral and CNS tissues and led to the reduction of HTT protein levels. Compared to CTRL mAB or vehicle treated mice, the mAB C6-17 treated YAC128 animals showed improved body weight and motor behaviors, a delayed progression in motor deficits and reduced striatal EM48 immunoreactivity. These results provide the first proof of concept for the feasibility and therapeutic efficacy of an antibody-based anti-HTT passive immunization approach and suggest this modality as a potential new HD treatment strategy.
Collapse
Affiliation(s)
| | - Yuanyun Xie
- University of Central Florida, Burnett School of Biomedical Sciences, Orlando, FL, United States of America; University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | - Nalini Potluri
- University of Central Florida, Burnett School of Biomedical Sciences, Orlando, FL, United States of America
| | - Ratnesh Kesineni
- University of Central Florida, Burnett School of Biomedical Sciences, Orlando, FL, United States of America
| | - Katlin Hencak
- University of Central Florida, Burnett School of Biomedical Sciences, Orlando, FL, United States of America
| | - Louisa Dal Cengio
- University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | | | - Abid Oueslati
- Centre de recherche du CHU - Université Laval, Québec, Canada
| | | | | | - Alberto Siddu
- Centre de recherche du CHU - Université Laval, Québec, Canada
| | | | | | | | - Michael R Hayden
- University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | - Amber L Southwell
- University of Central Florida, Burnett School of Biomedical Sciences, Orlando, FL, United States of America.
| |
Collapse
|
19
|
Limnander A, Kaur N, Asrat S, Tasker C, Boyapati A, Ben LH, Janczy J, Pedraza P, Abreu P, Chen WC, Godin S, Daniel BJ, Chin H, DeVeaux M, Rodriguez Lorenc K, Sirulnik A, Harari O, Stahl N, Sleeman MA, Murphy AJ, Yancopoulos GD, Orengo JM. A therapeutic strategy to target distinct sources of IgE and durably reverse allergy. Sci Transl Med 2023; 15:eadf9561. [PMID: 38091405 DOI: 10.1126/scitranslmed.adf9561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Immunoglobulin E (IgE) is a key driver of type 1 hypersensitivity reactions and allergic disorders, which are globally increasing in number and severity. Although eliminating pathogenic IgE may be a powerful way to treat allergy, no therapeutic strategy reported to date can fully ablate IgE production. Interleukin-4 receptor α (IL-4Rα) signaling is required for IgE class switching, and IL-4Rα blockade gradually reduces, but does not eliminate, IgE. The persistence of IgE after IL-4Rα blockade may be due to long-lived IgE+ plasma cells that maintain serological memory to allergens and thus may be susceptible to plasma cell-targeted therapeutics. We demonstrate that transient administration of a B cell maturation antigen x CD3 (BCMAxCD3) bispecific antibody markedly depletes IgE, as well as other immunoglobulins, by ablating long-lived plasma cells, although IgE and other immunoglobulins rapidly rebound after treatment. Concomitant IL-4Rα blockade specifically and durably prevents the reemergence of IgE by blocking IgE class switching while allowing the restoration of other immunoglobulins. Moreover, this combination treatment prevented anaphylaxis in mice. Together with additional cynomolgus monkey and human data, our studies demonstrate that allergic memory is primarily maintained by both non-IgE+ memory B cells that require class switching and long-lived IgE+ plasma cells. Our combination approach to durably eliminate pathogenic IgE has potential to benefit allergy in humans while preserving antibody-mediated immunity.
Collapse
Affiliation(s)
| | - Navneet Kaur
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Carley Tasker
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Anita Boyapati
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Li-Hong Ben
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - John Janczy
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Pablo Abreu
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Wen-Chi Chen
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Stephen Godin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Harvey Chin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Olivier Harari
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Neil Stahl
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Jamie M Orengo
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| |
Collapse
|
20
|
Amer EI, Allam SR, Hassan AY, El-Fakharany EM, Agwa MM, Khattab SN, Sheta E, El-Faham MH. Can antibody conjugated nanomicelles alter the prospect of antibody targeted therapy against schistosomiasis mansoni? PLoS Negl Trop Dis 2023; 17:e0011776. [PMID: 38039267 PMCID: PMC10691730 DOI: 10.1371/journal.pntd.0011776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 11/07/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND CLA (conjugated linoleic acid)-mediated activation of the schistosome tegument-associated sphingomyelinase and consequent disruption of the outer membrane might allow host antibodies to access the apical membrane antigens. Here, we investigated a novel approach to enhance specific antibody delivery to concealed surface membrane antigens of Schistosoma mansoni utilising antibody-conjugated-CLA nanomicelle technology. METHODOLOGY/PRINCIPAL FINDINGS We invented and characterised an amphiphilic CLA-loaded whey protein co-polymer (CLA-W) as an IV injectable protein nanocarrier. Rabbit anti-Schistosoma mansoni infection (anti-SmI) and anti-Schistosoma mansoni alkaline phosphatase specific IgG antibodies were purified from rabbit sera and conjugated to the surface of CLA-W co-polymer to form antibody-conjugated-CLA-W nanomicelles (Ab-CLA-W). We investigated the schistosomicidal effects of CLA-W and Ab-CLA-W in a mouse model of Schistosoma mansoni against early and late stages of infection. Results showed that conjugation of nanomicelles with antibodies, namely anti-SmI, significantly enhanced the micelles' schistosomicidal and anti-pathology activities at both the schistosomula and adult worm stages of the infection resulting in 64.6%-89.9% reductions in worm number; 72.5-94% and 66.4-85.2% reductions in hepatic eggs and granulomas, respectively. Treatment induced overall improvement in liver histopathology, reducing granuloma size and fibrosis and significantly affecting egg viability. Indirect immunofluorescence confirmed CLA-W-mediated antigen exposure on the worm surface. Electron microscopy revealed extensive ultrastructural damage in worm tegument induced by anti-SmI-CLA-W. CONCLUSION/SIGNIFICANCE The novel antibody-targeted nano-sized CLA delivery system offers great promise for treatment of Schistosoma mansoni infection and control of its transmission. Our in vivo observations confirm an immune-mediated enhanced effect of the schistosomicidal action of CLA and hints at the prospect of nanotechnology-based immunotherapy, not only for schistosomiasis, but also for other parasitic infections in which chemotherapy has been shown to be immune-dependent. The results propose that the immunodominant reactivity of the anti-SmI serum, Schistosoma mansoni fructose biphosphate aldolase, SmFBPA, merits serious attention as a therapeutic and vaccine candidate.
Collapse
Affiliation(s)
- Eglal I. Amer
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Sonia R. Allam
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Aceel Y. Hassan
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Esmail M. El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria, Egypt
| | - Mona M. Agwa
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Giza, Egypt
| | - Sherine N. Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Marwa H. El-Faham
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
21
|
Wang Y, Chen X, Zhou L, Chinn E, Yue J. The mechanism underlying the inverse proportion between serum IgE concentration and Stat3 transcriptional activity depends on Stat3 mutation type. Int Immunopharmacol 2023; 124:111031. [PMID: 37827058 DOI: 10.1016/j.intimp.2023.111031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 09/24/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023]
Abstract
Abnormalities in the IL-6/STAT3 signaling pathway can sometimes result in extremely high levels of IgE concentration in serum, however, the regulatory role of STAT3 in IgE production is elusive. We used several genetically modified mice with distinctive germline Stat3 transcriptional activity to assess the influence of Stat3 on the biochemical characteristics of IgE and found that the IgE concentration in serum is inversely proportional to Stat3 transcriptional activity. Intriguingly, the serum IgE concentration is directly proportional to IgE-producing B cells in Stat3-GOF mice but inversely proportional in mice carrying Stat3 mutations with reduced transcriptional activity. For reduced Stat3 transcriptional activity induced high levels of IgE in the mice, IL-4/Stat6 signaling is indispensable for IgE production, but it was observed that an increased IgE concentration was accompanied by reduced IL-4/Stat6 signaling and lessened IgE-producing B cells, which implies that an increase in IgE concentration may result from an extended half-life of IgE but not an increasing number of IgE-producing cells.
Collapse
Affiliation(s)
- Yuting Wang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Xiangjie Chen
- Department of Laboratory Medicine, Jiangnan University Medical Center (Wuxi No. 2 People's Hospital), Wuxi, China
| | - Lu Zhou
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Eugene Chinn
- Peninsula Cancer Research Center, Binzhou Medical University, Yantai, China
| | - Jicheng Yue
- Peninsula Cancer Research Center, Binzhou Medical University, Yantai, China.
| |
Collapse
|
22
|
Sun CP, Chiu CW, Wu PY, Tsung SI, Lee IJ, Hu CW, Hsu MF, Kuo TJ, Lan YH, Chen LY, Ng HY, Chung MJ, Liao HN, Tseng SC, Lo CH, Chen YJ, Liao CC, Chang CS, Liang JJ, Draczkowski P, Puri S, Chang YC, Huang JS, Chen CC, Kau JH, Chen YH, Liu WC, Wu HC, Danny Hsu ST, Wang IH, Tao MH. Development of AAV-delivered broadly neutralizing anti-human ACE2 antibodies against SARS-CoV-2 variants. Mol Ther 2023; 31:3322-3336. [PMID: 37689971 PMCID: PMC10638075 DOI: 10.1016/j.ymthe.2023.09.002] [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: 05/04/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023] Open
Abstract
The ongoing evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), resulting in the emergence of new variants that are resistant to existing vaccines and therapeutic antibodies, has raised the need for novel strategies to combat the persistent global COVID-19 epidemic. In this study, a monoclonal anti-human angiotensin-converting enzyme 2 (hACE2) antibody, ch2H2, was isolated and humanized to block the viral receptor-binding domain (RBD) binding to hACE2, the major entry receptor of SARS-CoV-2. This antibody targets the RBD-binding site on the N terminus of hACE2 and has a high binding affinity to outcompete the RBD. In vitro, ch2H2 antibody showed potent inhibitory activity against multiple SARS-CoV-2 variants, including the most antigenically drifted and immune-evading variant Omicron. In vivo, adeno-associated virus (AAV)-mediated delivery enabled a sustained expression of monoclonal antibody (mAb) ch2H2, generating a high concentration of antibodies in mice. A single administration of AAV-delivered mAb ch2H2 significantly reduced viral RNA load and infectious virions and mitigated pulmonary pathological changes in mice challenged with SARS-CoV-2 Omicron BA.5 subvariant. Collectively, the results suggest that AAV-delivered hACE2-blocking antibody provides a promising approach for developing broad-spectrum antivirals against SARS-CoV-2 and potentially other hACE2-dependent pathogens that may emerge in the future.
Collapse
Affiliation(s)
- Cheng-Pu Sun
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Chi-Wen Chiu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Department of Clinical Laboratory Science and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Ping-Yi Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Szu-I Tsung
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Graduate Institute of Microbiology, National Taiwan University, Taipei, Taiwan
| | - I-Jung Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Graduate Institute of Microbiology, National Taiwan University, Taipei, Taiwan
| | - Chih-Wei Hu
- Institute of Preventive Medicine, National Defense Medical College, Taipei, Taiwan
| | - Min-Feng Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Tzu-Jiun Kuo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Hua Lan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Li-Yao Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Yee Ng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Meng-Jhe Chung
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hsin-Ni Liao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Sheng-Che Tseng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chia-Hui Lo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yung-Jiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chun-Che Liao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Chih-Shin Chang
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Jian-Jong Liang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | - Sarita Puri
- Department of Bioscience, University of Milan, Milan, Italy
| | - Yuan-Chih Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Jing-Siou Huang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Cheng-Cheung Chen
- Institute of Preventive Medicine, National Defense Medical College, Taipei, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Jyh-Hwa Kau
- Institute of Preventive Medicine, National Defense Medical College, Taipei, Taiwan
| | - Yen-Hui Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wen-Chun Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Han-Chung Wu
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan; Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Shang-Te Danny Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan; International Institute for Sustainability with Knotted Chiral Meta Matter, Hiroshima University, Higashihiroshima, Japan
| | - I-Hsuan Wang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | - Mi-Hua Tao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan; Department of Clinical Laboratory Science and Medical Biotechnology, National Taiwan University, Taipei, Taiwan; Graduate Institute of Microbiology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
23
|
Liu Y, Chen T, Zhu Y, Furey A, Lowary TL, Chan J, Bournazos S, Ravetch JV, Achkar JM. Features and protective efficacy of human mAbs targeting Mycobacterium tuberculosis arabinomannan. JCI Insight 2023; 8:e167960. [PMID: 37733444 PMCID: PMC10619501 DOI: 10.1172/jci.insight.167960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 09/06/2023] [Indexed: 09/23/2023] Open
Abstract
A better understanding of the epitopes most relevant for antibody-mediated protection against tuberculosis (TB) remains a major knowledge gap. We have shown that human polyclonal IgG against the Mycobacterium tuberculosis (M. tuberculosis) surface glycan arabinomannan (AM) and related lipoarabinomannan (LAM) is protective against TB. To investigate the impact of AM epitope recognition and Fcγ receptor (FcγR) binding on antibody functions against M. tuberculosis, we isolated a high-affinity human monoclonal antibody (mAb; P1AM25) against AM and showed its binding to oligosaccharide (OS) motifs we previously found to be associated with in vitro functions of human polyclonal anti-AM IgG. Human IgG1 P1AM25, but not 2 other high-affinity human IgG1 anti-AM mAbs reactive with different AM OS motifs, enhanced M. tuberculosis phagocytosis by macrophages and reduced intracellular growth in an FcγR-dependent manner. P1AM25 in murine IgG2a, but neither murine IgG1 nor a non-FcγR-binding IgG, given intraperitoneally prior to and after aerosolized M. tuberculosis infection, was protective in C57BL/6 mice. Moreover, we demonstrated the protective efficacy of human IgG1 P1AM25 in passive transfer with M. tuberculosis-infected FcγR-humanized mice. These data enhance our knowledge of the important interplay between both antibody epitope specificity and Fc effector functions in the defense against M. tuberculosis and could inform development of vaccines against TB.
Collapse
Affiliation(s)
- Yanyan Liu
- Department of Microbiology and Immunology and
| | - Tingting Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yongqi Zhu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Aisha Furey
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Todd L. Lowary
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - John Chan
- Public Health Research Institute at the International Center for Public Health, New Jersey Medical School – Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | | | | | - Jacqueline M. Achkar
- Department of Microbiology and Immunology and
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
24
|
Batsuli G, Ito J, York ES, Cox C, Baldwin W, Gill S, Lollar P, Meeks SL. Factor VIII antibody immune complexes modulate the humoral response to factor VIII in an epitope-dependent manner. Front Immunol 2023; 14:1233356. [PMID: 37720212 PMCID: PMC10501482 DOI: 10.3389/fimmu.2023.1233356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/11/2023] [Indexed: 09/19/2023] Open
Abstract
Introduction Soluble antigens complexed with immunoglobulin G (IgG) antibodies can induce robust adaptive immune responses in vitro and in animal models of disease. Factor VIII immune complexes (FVIII-ICs) have been detected in individuals with hemophilia A and severe von Willebrand disease following FVIII infusions. Yet, it is unclear if and how FVIII-ICs affect antibody development over time. Methods In this study, we analyzed internalization of FVIII complexed with epitope-mapped FVIII-specific IgG monoclonal antibodies (MAbs) by murine bone marrow-derived dendritic cells (BMDCs) in vitro and antibody development in hemophilia A (FVIII-/-) mice injected with FVIII-IC over time. Results FVIII complexed with 2-116 (A1 domain MAb), 2-113 (A3 domain MAb), and I55 (C2 domain MAb) significantly increased FVIII uptake by BMDC but only FVIII/2-116 enhanced antibody titers in FVIII-/- mice compared to FVIII alone. FVIII/4A4 (A2 domain MAb) showed similar FVIII uptake by BMDC to that of isolated FVIII yet significantly increased antibody titers when injected in FVIII-/- mice. Enhanced antibody responses observed with FVIII/2-116 and FVIII/4A4 complexes in vivo were abrogated in the absence of the FVIII carrier protein von Willebrand factor. Conclusion These findings suggest that a subset of FVIII-IC modulates the humoral response to FVIII in an epitope-dependent manner, which may provide insight into the antibody response observed in some patients with hemophilia A.
Collapse
Affiliation(s)
- Glaivy Batsuli
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Jasmine Ito
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Elizabeth S. York
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Courtney Cox
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Wallace Baldwin
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Surinder Gill
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Pete Lollar
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Shannon L. Meeks
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, United States
| |
Collapse
|
25
|
Christensen PKF, Hansen AK, Skov S, Larsen J, Høyer-Hansen MH, Koch J. Effect of Systemic Administration of CD4 + T cells and Local Administration of T-cell Stimulants on T-cell Activity in Psoriatic Skin Xenografts on NOG Mice. Comp Med 2023; 73:285-293. [PMID: 37625901 PMCID: PMC10702288 DOI: 10.30802/aalas-cm-23-000006] [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: 02/08/2023] [Revised: 04/13/2023] [Accepted: 05/18/2023] [Indexed: 08/27/2023]
Abstract
Immunodeficient mice engrafted with psoriatic human skin are widely used for the preclinical evaluation of new drug candidates. However, the T-cell activity, including the IL23/IL17 pathway, declines in the graft over time after engraftment, which likely affects the study data. Here, we investigated whether the T-cell activity could be sustained in xenografted psoriatic skin by local stimulation of T cells or systemic injection of autologous CD4 + T cells. We surgically transplanted human psoriatic skin from 5 untreated patients onto female NOG mice. Six days after surgery, mice received an intraperitoneal injection of autologous human CD4+ T cells, a subcutaneous injection under the grafts of a T-cell stimulation cocktail consisting of recombinant human IL2, human IL23, antihuman CD3, and antihuman CD28, or saline. Mice were euthanized 21 d after surgery and spleens and graft biopsies were collected for analysis. Human T cells were present in the grafts, and 60% of the grafts maintained the psoriatic phenotype. However, neither local T-cell stimulation nor systemic injection of autologous CD4+ T cells affected the protein levels of human IL17A, IL22, IFN γ, and TNF α in the grafts. In conclusion, NOG mice seem to accept psoriatic skin grafts, but the 2 approaches studied here did not affect human T-cell activity in the grafts. Therefore, NOG mice do not appear in this regard to be superior to other immunodeficient mice used for psoriasis xenografts.
Collapse
Affiliation(s)
- Pernille KF Christensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- LEO Pharma, Ballerup, Denmark; and
| | - Axel K Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
26
|
Kang J, Mateu-Borrás M, Monroe HL, Sen-Kilic E, Miller SJ, Dublin SR, Huckaby AB, Yang E, Pyles GM, Nunley MA, Chapman JA, Amin MS, Damron FH, Barbier M. Monoclonal antibodies against lipopolysaccharide protect against Pseudomonas aeruginosa challenge in mice. Front Cell Infect Microbiol 2023; 13:1191806. [PMID: 37424774 PMCID: PMC10326049 DOI: 10.3389/fcimb.2023.1191806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023] Open
Abstract
Pseudomonas aeruginosa is a common cause of hospital-acquired infections, including central line-associated bloodstream infections and ventilator-associated pneumonia. Unfortunately, effective control of these infections can be difficult, in part due to the prevalence of multi-drug resistant strains of P. aeruginosa. There remains a need for novel therapeutic interventions against P. aeruginosa, and the use of monoclonal antibodies (mAb) is a promising alternative strategy to current standard of care treatments such as antibiotics. To develop mAbs against P. aeruginosa, we utilized ammonium metavanadate, which induces cell envelope stress responses and upregulates polysaccharide expression. Mice were immunized with P. aeruginosa grown with ammonium metavanadate and we developed two IgG2b mAbs, WVDC-0357 and WVDC-0496, directed against the O-antigen lipopolysaccharide of P. aeruginosa. Functional assays revealed that WVDC-0357 and WVDC-0496 directly reduced the viability of P. aeruginosa and mediated bacterial agglutination. In a lethal sepsis model of infection, prophylactic treatment of mice with WVDC-0357 and WVDC-0496 at doses as low as 15 mg/kg conferred 100% survival against challenge. In both sepsis and acute pneumonia models of infection, treatment with WVDC-0357 and WVDC-0496 significantly reduced bacterial burden and inflammatory cytokine production post-challenge. Furthermore, histopathological examination of the lungs revealed that WVDC-0357 and WVDC-0496 reduced inflammatory cell infiltration. Overall, our results indicate that mAbs directed against lipopolysaccharide are a promising therapy for the treatment and prevention of P. aeruginosa infections.
Collapse
Affiliation(s)
- Jason Kang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Margalida Mateu-Borrás
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Hunter L. Monroe
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University, Morgantown, WV, United States
| | - Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Sarah Jo Miller
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Spencer R. Dublin
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Annalisa B. Huckaby
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Evita Yang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Gage M. Pyles
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Mason A. Nunley
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Josh A. Chapman
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Md Shahrier Amin
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University, Morgantown, WV, United States
| | - F. Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| |
Collapse
|
27
|
Bouyssi A, Déméautis T, Trecourt A, Delles M, Agostini F, Monneret G, Glehen O, Wallon M, Persat F, Devouassoux G, Bentaher A, Menotti J. Characterization of Lung Inflammatory Response to Aspergillus fumigatus Spores. J Fungi (Basel) 2023; 9:682. [PMID: 37367618 DOI: 10.3390/jof9060682] [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: 04/22/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
The airway exposure to Aspergillus fumigatus spores (AFsp) is associated with an inflammatory response, potentially leading to allergic and/or chronic pulmonary aspergillosis. The aim of our study is to better understand the host response, first in vitro, then in vivo, following the chronic exposure of mice to AFsp. We investigated the inflammatory response to AFsp in cell mono- and co-culture systems with murine macrophages and alveolar epithelial cells. The mice were subjected to two intranasal instillations using 105 AFsp. Their lungs were processed for inflammatory and histopathological analyses. In cell culture, the gene expressions significantly increased for TNF-α, CXCL-1, CXCL-2, IL-1β, IL-1α and GM-CSF in macrophages, with these increases being limited for TNF-α, CXCL-1 and IL-1α in epithelial cells. In co-culture, increases in the TNF-α, CXCL-2 and CXCL-1 gene expressions were observed to be associated with increased protein levels. The in vivo lung histological analyses of mice challenged by AFsp showed cellular infiltrates in the peribronchial and/or alveolar spaces. A Bio-Plex approach on the bronchoalveolar lavage revealed significant increases in the protein secretion of selected mediators of the challenged mice compared to the unchallenged mice. In conclusion, the exposure to AFsp resulted in a marked inflammatory response of macrophages and epithelial cells. These inflammatory findings were confirmed in mouse models associated with lung histologic changes.
Collapse
Affiliation(s)
- Alexandra Bouyssi
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Team Inflammation and Immunity of the Respiratory Epithelium, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
| | - Tanguy Déméautis
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Team Inflammation and Immunity of the Respiratory Epithelium, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
| | - Alexis Trecourt
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Team Inflammation and Immunity of the Respiratory Epithelium, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
- Department of Pathology, South Lyon Hospital, Hospices Civils de Lyon, 69495 Pierre Bénite, France
| | - Marie Delles
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Team Inflammation and Immunity of the Respiratory Epithelium, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
| | - Fany Agostini
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Team Inflammation and Immunity of the Respiratory Epithelium, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
| | - Guillaume Monneret
- Immunology Laboratory, EA7426, Edouard Herriot Hospital, Hospices Civils de Lyon and Claude Bernard University-Lyon 1, 69003 Lyon, France
| | - Olivier Glehen
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Surgical Department, South Lyon Hospital, Hospices Civils de Lyon, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
| | - Martine Wallon
- Department of Medical Mycology and Parasitology, Institute of Infectious Agents, Croix-Rousse Hospital, Hospices Civils de Lyon, 69004 Lyon, France
| | - Florence Persat
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Team Inflammation and Immunity of the Respiratory Epithelium, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
- Department of Medical Mycology and Parasitology, Institute of Infectious Agents, Croix-Rousse Hospital, Hospices Civils de Lyon, 69004 Lyon, France
| | - Gilles Devouassoux
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Team Inflammation and Immunity of the Respiratory Epithelium, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
- Department of Pulmonology, Croix-Rousse Hospital, Hospices Civils de Lyon, 69004 Lyon, France
| | - Abderrazzak Bentaher
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Team Inflammation and Immunity of the Respiratory Epithelium, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
| | - Jean Menotti
- UR3738 Centre pour l'lnnovation en Cancérologie de Lyon, Team Inflammation and Immunity of the Respiratory Epithelium, Claude Bernard University-Lyon 1, 69495 Pierre Bénite, France
- Department of Medical Mycology and Parasitology, Institute of Infectious Agents, Croix-Rousse Hospital, Hospices Civils de Lyon, 69004 Lyon, France
| |
Collapse
|
28
|
Zhang F, Jenkins J, de Carvalho RVH, Nakandakari-Higa S, Chen T, Abernathy ME, Baharani VA, Nyakatura EK, Andrew D, Lebedeva IV, Lorenz IC, Hoffmann HH, Rice CM, Victora GD, Barnes CO, Hatziioannou T, Bieniasz PD. Pan-sarbecovirus prophylaxis with human anti-ACE2 monoclonal antibodies. Nat Microbiol 2023; 8:1051-1063. [PMID: 37188812 PMCID: PMC10234812 DOI: 10.1038/s41564-023-01389-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 04/19/2023] [Indexed: 05/17/2023]
Abstract
Human monoclonal antibodies (mAbs) that target the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein have been isolated from convalescent individuals and developed into therapeutics for SARS-CoV-2 infection. However, therapeutic mAbs for SARS-CoV-2 have been rendered obsolete by the emergence of mAb-resistant virus variants. Here we report the generation of a set of six human mAbs that bind the human angiotensin-converting enzyme-2 (hACE2) receptor, rather than the SARS-CoV-2 spike protein. We show that these antibodies block infection by all hACE2 binding sarbecoviruses tested, including SARS-CoV-2 ancestral, Delta and Omicron variants at concentrations of ~7-100 ng ml-1. These antibodies target an hACE2 epitope that binds to the SARS-CoV-2 spike, but they do not inhibit hACE2 enzymatic activity nor do they induce cell-surface depletion of hACE2. They have favourable pharmacology, protect hACE2 knock-in mice against SARS-CoV-2 infection and should present a high genetic barrier to the acquisition of resistance. These antibodies should be useful prophylactic and treatment agents against any current or future SARS-CoV-2 variants and might be useful to treat infection with any hACE2-binding sarbecoviruses that emerge in the future.
Collapse
Affiliation(s)
- Fengwen Zhang
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Jesse Jenkins
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | | | | | - Teresia Chen
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Viren A Baharani
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | | | - David Andrew
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | - Irina V Lebedeva
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | - Ivo C Lorenz
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | - H-Heinrich Hoffmann
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Christopher O Barnes
- Department of Biology, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA.
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
29
|
Wang-Bishop L, Kimmel BR, Ngwa VM, Madden MZ, Baljon JJ, Florian DC, Hanna A, Pastora LE, Sheehy TL, Kwiatkowski AJ, Wehbe M, Wen X, Becker KW, Garland KM, Schulman JA, Shae D, Edwards D, Wolf MM, Delapp R, Christov PP, Beckermann KE, Balko JM, Rathmell WK, Rathmell JC, Chen J, Wilson JT. STING-activating nanoparticles normalize the vascular-immune interface to potentiate cancer immunotherapy. Sci Immunol 2023; 8:eadd1153. [PMID: 37146128 PMCID: PMC10226150 DOI: 10.1126/sciimmunol.add1153] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 04/13/2023] [Indexed: 05/07/2023]
Abstract
The tumor-associated vasculature imposes major structural and biochemical barriers to the infiltration of effector T cells and effective tumor control. Correlations between stimulator of interferon genes (STING) pathway activation and spontaneous T cell infiltration in human cancers led us to evaluate the effect of STING-activating nanoparticles (STANs), which are a polymersome-based platform for the delivery of a cyclic dinucleotide STING agonist, on the tumor vasculature and attendant effects on T cell infiltration and antitumor function. In multiple mouse tumor models, intravenous administration of STANs promoted vascular normalization, evidenced by improved vascular integrity, reduced tumor hypoxia, and increased endothelial cell expression of T cell adhesion molecules. STAN-mediated vascular reprogramming enhanced the infiltration, proliferation, and function of antitumor T cells and potentiated the response to immune checkpoint inhibitors and adoptive T cell therapy. We present STANs as a multimodal platform that activates and normalizes the tumor microenvironment to enhance T cell infiltration and function and augments responses to immunotherapy.
Collapse
Affiliation(s)
- Lihong Wang-Bishop
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Blaise R. Kimmel
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Verra M. Ngwa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Matthew Z. Madden
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jessalyn J. Baljon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - David C. Florian
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Ann Hanna
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Lucinda E. Pastora
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Taylor L. Sheehy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Alexander J. Kwiatkowski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Mohamed Wehbe
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Kyle W. Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Kyle M. Garland
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Jacob A. Schulman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Deanna Edwards
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Melissa M. Wolf
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Rossane Delapp
- Department of Civil and Environmental Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Plamen P. Christov
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States
| | - Kathryn E. Beckermann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Justin M. Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - W. Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jin Chen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
30
|
Jossi SE, Arcuri M, Alshayea A, Persaud RR, Marcial-Juárez E, Palmieri E, Di Benedetto R, Pérez-Toledo M, Pillaye J, Channell WM, Schager AE, Lamerton RE, Cook CN, Goodall M, Haneda T, Bäumler AJ, Jackson-Jones LH, Toellner KM, MacLennan CA, Henderson IR, Micoli F, Cunningham AF. Vi polysaccharide and conjugated vaccines afford similar early, IgM or IgG-independent control of infection but boosting with conjugated Vi vaccines sustains the efficacy of immune responses. Front Immunol 2023; 14:1139329. [PMID: 37033932 PMCID: PMC10076549 DOI: 10.3389/fimmu.2023.1139329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Vaccination with Vi capsular polysaccharide (Vi-PS) or protein-Vi typhoid conjugate vaccine (TCV) can protect adults against Salmonella Typhi infections. TCVs offer better protection than Vi-PS in infants and may offer better protection in adults. Potential reasons for why TCV may be superior in adults are not fully understood. Methods and results Here, we immunized wild-type (WT) mice and mice deficient in IgG or IgM with Vi-PS or TCVs (Vi conjugated to tetanus toxoid or CRM197) for up to seven months, with and without subsequent challenge with Vi-expressing Salmonella Typhimurium. Unexpectedly, IgM or IgG alone were similarly able to reduce bacterial burdens in tissues, and this was observed in response to conjugated or unconjugated Vi vaccines and was independent of antibody being of high affinity. Only in the longer-term after immunization (>5 months) were differences observed in tissue bacterial burdens of mice immunized with Vi-PS or TCV. These differences related to the maintenance of antibody responses at higher levels in mice boosted with TCV, with the rate of fall in IgG titres induced to Vi-PS being greater than for TCV. Discussion Therefore, Vi-specific IgM or IgG are independently capable of protecting from infection and any superior protection from vaccination with TCV in adults may relate to responses being able to persist better rather than from differences in the antibody isotypes induced. These findings suggest that enhancing our understanding of how responses to vaccines are maintained may inform on how to maximize protection afforded by conjugate vaccines against encapsulated pathogens such as S. Typhi.
Collapse
Affiliation(s)
- Siân E. Jossi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Melissa Arcuri
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- GSK Vaccines Institute for Global Health SRL, Siena, Italy
| | - Areej Alshayea
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Ruby R. Persaud
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Edith Marcial-Juárez
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health SRL, Siena, Italy
| | | | - Marisol Pérez-Toledo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Jamie Pillaye
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Will M. Channell
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Anna E. Schager
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Rachel E. Lamerton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Charlotte N. Cook
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Margaret Goodall
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Takeshi Haneda
- Laboratory of Microbiology, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, United States
| | - Lucy H. Jackson-Jones
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, United Kingdom
| | - Kai-Michael Toellner
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Calman A. MacLennan
- Bill & Melinda Gates Foundation, London, United Kingdom
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ian R. Henderson
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | | | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
31
|
Hamdan F, Feodoroff M, Russo S, Fusciello M, Feola S, Chiaro J, Antignani G, Greco F, Leusen J, Ylösmäki E, Grönholm M, Cerullo V. Controlled release of enhanced cross-hybrid IgGA Fc PD-L1 inhibitors using oncolytic adenoviruses. Mol Ther Oncolytics 2023; 28:264-276. [PMID: 36911070 PMCID: PMC9995465 DOI: 10.1016/j.omto.2023.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/31/2023] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint inhibitors have clinical success in prolonging the life of many cancer patients. However, only a minority of patients benefit from such therapy, calling for further improvements. Currently, most PD-L1 checkpoint inhibitors in the clinic do not elicit Fc effector mechanisms that would substantially increase their efficacy. To gain potency and circumvent off-target effects, we previously designed an oncolytic adenovirus (Ad-Cab) expressing an Fc fusion peptide against PD-L1 on a cross-hybrid immunoglobulin GA (IgGA) Fc. Ad-Cab elicited antibody effector mechanisms of IgG1 and IgA, which led to higher tumor killing compared with each isotype alone and with clinically approved PD-L1 checkpoint inhibitors. In this study, we further improved the therapy to increase the IgG1 Fc effector mechanisms of the IgGA Fc fusion peptide (Ad-Cab FT) by adding four somatic mutations that increase natural killer (NK) cell activation. Ad-Cab FT was shown to work better at lower concentrations compared with Ad-Cab in vitro and in vivo and to have better tumor- and myeloid-derived suppressor cell killing, likely because of higher NK cell activation. Additionally, the biodistribution of the Fc fusion peptide demonstrated targeted release in the tumor microenvironment with minimal or no leakage to the peripheral blood and organs in mice. These data demonstrate effective and safe use of Ad-Cab FT, bidding for further clinical investigation.
Collapse
Affiliation(s)
- Firas Hamdan
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Michaela Feodoroff
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Salvatore Russo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Manlio Fusciello
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Sara Feola
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jacopo Chiaro
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Gabriella Antignani
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Francesca Greco
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jeanette Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Erkko Ylösmäki
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mikaela Grönholm
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland.,Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.,Department of Molecular Medicine and Medical Biotechnology and CEINGE, Naples University Federico II, Naples, Italy
| |
Collapse
|
32
|
Aranda CJ, Gonzalez-Kozlova E, Saunders SP, Fernandes-Braga W, Ota M, Narayanan S, He JS, Del Duca E, Swaroop B, Gnjatic S, Shattner G, Reibman J, Soter NA, Guttman-Yassky E, Curotto de Lafaille MA. IgG memory B cells expressing IL4R and FCER2 are associated with atopic diseases. Allergy 2023; 78:752-766. [PMID: 36445014 PMCID: PMC9991991 DOI: 10.1111/all.15601] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/25/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Atopic diseases are characterized by IgE antibody responses that are dependent on cognate CD4 T cell help and T cell-produced IL-4 and IL-13. Current models of IgE cell differentiation point to the role of IgG memory B cells as precursors of pathogenic IgE plasma cells. The goal of this work was to identify intrinsic features of memory B cells that are associated with IgE production in atopic diseases. METHODS Peripheral blood B lymphocytes were collected from individuals with physician diagnosed asthma or atopic dermatitis (AD) and from non-atopic individuals. These samples were analyzed by spectral flow cytometry, single cell RNA sequencing (scRNAseq), and in vitro activation assays. RESULTS We identified a novel population of IgG memory B cells characterized by the expression of IL-4/IL-13 regulated genes FCER2/CD23, IL4R, IL13RA1, and IGHE, denoting a history of differentiation during type 2 immune responses. CD23+ IL4R+ IgG+ memory B cells had increased occurrence in individuals with atopic disease. Importantly, the frequency of CD23+ IL4R+ IgG+ memory B cells correlated with levels of circulating IgE. Consistently, in vitro stimulated B cells from atopic individuals generated more IgE+ cells than B cells from non-atopic subjects. CONCLUSIONS These findings suggest that CD23+ IL4R+ IgG+ memory B cells transcribing IGHE are potential precursors of IgE plasma cells and are linked to pathogenic IgE production.
Collapse
Affiliation(s)
- Carlos J Aranda
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
| | | | - Sean P Saunders
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York University School of Medicine (NYUSM), New York, New York, USA
| | - Weslley Fernandes-Braga
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
| | - Miyo Ota
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
| | - Sriram Narayanan
- Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Jin-Shu He
- Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Ester Del Duca
- Department of Dermatology, ISMMS, New York, New York, USA
| | - Bose Swaroop
- Department of Dermatology, ISMMS, New York, New York, USA
| | - Sacha Gnjatic
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
- Tisch Cancer Institute, ISMMS, New York, New York, USA
| | - Gail Shattner
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York University School of Medicine (NYUSM), New York, New York, USA
| | - Joan Reibman
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York University School of Medicine (NYUSM), New York, New York, USA
| | | | | | - Maria A Curotto de Lafaille
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
| |
Collapse
|
33
|
Pance K, Gramespacher JA, Byrnes JR, Salangsang F, Serrano JAC, Cotton AD, Steri V, Wells JA. Modular cytokine receptor-targeting chimeras for targeted degradation of cell surface and extracellular proteins. Nat Biotechnol 2023; 41:273-281. [PMID: 36138170 PMCID: PMC9931583 DOI: 10.1038/s41587-022-01456-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/01/2022] [Indexed: 12/20/2022]
Abstract
Targeted degradation of cell surface and extracellular proteins via lysosomal delivery is an important means to modulate extracellular biology. However, these approaches have limitations due to lack of modularity, ease of development, restricted tissue targeting and applicability to both cell surface and extracellular proteins. We describe a lysosomal degradation strategy, termed cytokine receptor-targeting chimeras (KineTACs), that addresses these limitations. KineTACs are fully genetically encoded bispecific antibodies consisting of a cytokine arm, which binds its cognate cytokine receptor, and a target-binding arm for the protein of interest. We show that KineTACs containing the cytokine CXCL12 can use the decoy recycling receptor, CXCR7, to target a variety of target proteins to the lysosome for degradation. Additional KineTACs were designed to harness other CXCR7-targeting cytokines, CXCL11 and vMIPII, and the interleukin-2 (IL-2) receptor-targeting cytokine IL-2. Thus, KineTACs represent a general, modular, selective and simple genetically encoded strategy for inducing lysosomal delivery of extracellular and cell surface targets with broad or tissue-specific distribution.
Collapse
Affiliation(s)
- Katarina Pance
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.,EpiBiologics, Inc., San Carlos, CA, USA
| | - Josef A Gramespacher
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.,EpiBiologics, Inc., San Carlos, CA, USA
| | - James R Byrnes
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Fernando Salangsang
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.,Preclinical Therapeutics Core, University of California San Francisco, San Francisco, CA, USA
| | - Juan-Antonio C Serrano
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.,Preclinical Therapeutics Core, University of California San Francisco, San Francisco, CA, USA
| | - Adam D Cotton
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Veronica Steri
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.,Preclinical Therapeutics Core, University of California San Francisco, San Francisco, CA, USA
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA. .,Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
34
|
Advances in antibody-based therapy in oncology. NATURE CANCER 2023; 4:165-180. [PMID: 36806801 DOI: 10.1038/s43018-023-00516-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 01/10/2023] [Indexed: 02/22/2023]
Abstract
Monoclonal antibodies are a growing class of targeted cancer therapeutics, characterized by exquisite specificity, long serum half-life, high affinity and immune effector functions. In this review, we outline key advances in the field with a particular focus on recent and emerging classes of engineered antibody therapeutic candidates, discuss molecular structure and mechanisms of action and provide updates on clinical development and practice.
Collapse
|
35
|
Pathogenesis of Anemia in Canine Babesiosis: Possible Contribution of Pro-Inflammatory Cytokines and Chemokines-A Review. Pathogens 2023; 12:pathogens12020166. [PMID: 36839438 PMCID: PMC9962459 DOI: 10.3390/pathogens12020166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Canine babesiosis is a tick-borne protozoan disease caused by intraerythrocytic parasites of the genus Babesia. The infection may lead to anemia in infected dogs. However, anemia is not directly caused by the pathogen. The parasite's developmental stages only have a marginal role in contributing to a decreased red blood cell (RBC) count. The main cause of anemia in affected dogs is the immune response to the infection. This response includes antibody production, erythrophagocytosis, oxidative damage of RBCs, complement activation, and antibody-dependent cellular cytotoxicity. Moreover, both infected and uninfected erythrocytes are retained in the spleen and sequestered in micro-vessels. All these actions are driven by pro-inflammatory cytokines and chemokines, especially IFN-γ, TNF-α, IL-6, and IL-8. Additionally, imbalance between the actions of pro- and anti-inflammatory cytokines plays a role in patho-mechanisms leading to anemia in canine babesiosis. This article is a review of the studies on the pathogenesis of anemia in canine babesiosis and related diseases, such as bovine or murine babesiosis and human or murine malaria, and the role of pro-inflammatory cytokines and chemokines in the mechanisms leading to anemia in infected dogs.
Collapse
|
36
|
Kirschenbaum D, Dadgar‐Kiani E, Catto F, Voigt FF, Trevisan C, Bichsel O, Shirani H, Nilsson KPR, Frontzek KJ, Paganetti P, Helmchen F, Lee JH, Aguzzi A. Whole-brain microscopy reveals distinct temporal and spatial efficacy of anti-Aβ therapies. EMBO Mol Med 2023; 15:e16789. [PMID: 36382364 PMCID: PMC9832821 DOI: 10.15252/emmm.202216789] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/18/2022] Open
Abstract
Many efforts targeting amyloid-β (Aβ) plaques for the treatment of Alzheimer's Disease thus far have resulted in failures during clinical trials. Regional and temporal heterogeneity of efficacy and dependence on plaque maturity may have contributed to these disappointing outcomes. In this study, we mapped the regional and temporal specificity of various anti-Aβ treatments through high-resolution light-sheet imaging of electrophoretically cleared brains. We assessed the effect on amyloid plaque formation and growth in Thy1-APP/PS1 mice subjected to β-secretase inhibitors, polythiophenes, or anti-Aβ antibodies. Each treatment showed unique spatiotemporal Aβ clearance, with polythiophenes emerging as a potent anti-Aβ compound. Furthermore, aligning with a spatial-transcriptomic atlas revealed transcripts that correlate with the efficacy of each Aβ therapy. As observed in this study, there is a striking dependence of specific treatments on the location and maturity of Aβ plaques. This may also contribute to the clinical trial failures of Aβ-therapies, suggesting that combinatorial regimens may be significantly more effective in clearing amyloid deposition.
Collapse
Affiliation(s)
- Daniel Kirschenbaum
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | | | - Francesca Catto
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Fabian F Voigt
- Laboratory of Neural Circuit Dynamics, Brain Research InstituteUniversity of ZurichZurichSwitzerland
- Neuroscience Center ZurichUniversity of Zurich & ETH ZurichZurichSwitzerland
| | - Chiara Trevisan
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Oliver Bichsel
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Hamid Shirani
- Division of Chemistry, Department of Physics, Chemistry and BiologyLinköping UniversityLinköpingSweden
| | - K Peter R Nilsson
- Division of Chemistry, Department of Physics, Chemistry and BiologyLinköping UniversityLinköpingSweden
| | - Karl J Frontzek
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Paolo Paganetti
- Laboratory for Biomedical NeurosciencesTorricella‐TaverneNeurocenter of Southern Switzerland, Ente Cantonale OspedalieroSwitzerland
- Faculty of Biomedical NeurosciencesUniversità della Svizzera ItalianaLuganoSwitzerland
| | - Fritjof Helmchen
- Laboratory of Neural Circuit Dynamics, Brain Research InstituteUniversity of ZurichZurichSwitzerland
| | - Jin Hyung Lee
- Department of BioengineeringStanford UniversityStanfordCAUSA
- Department of Neurology and Neurological SciencesStanford UniversityStanfordCAUSA
- Department of Electrical EngineeringStanford UniversityStanfordCAUSA
- Department of NeurosurgeryStanford UniversityStanfordCAUSA
| | - Adriano Aguzzi
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| |
Collapse
|
37
|
Strugnell RA. When secretion turns into excretion - the different roles of IgA. Front Immunol 2022; 13:1076312. [PMID: 36618388 PMCID: PMC9812643 DOI: 10.3389/fimmu.2022.1076312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
IgA deficiency is the commonest immunodeficiency affecting up to 1 in 700 individuals. The effects of IgA deficiency are difficult to see in many individuals, are mild in many fewer and severe in fewer still. While monovalent IgA is found in serum, dimeric IgA is secreted through mucosal surfaces where it helps to maintain epithelial homeostasis. Studies with knockout mice have taught us that there are subtle inflammatory consequences of removing secretory IgA (sIgA), and the best explanation for these changes can be related by the loss of the 'excretory' immune system. The excretion of antigens is a logical process in regulating the immune system, given the long half-life of complement fixing antibodies. But the function of IgA as an immune or inflammation regulator may go beyond antigen removal.
Collapse
|
38
|
Alugupalli AS, Cravens MP, Walker JA, Gulandijany D, Dickinson GS, Debes GF, Schifferli DM, Bäumler AJ, Alugupalli KR, Alugupalli KR. The Lack of Natural IgM Increases Susceptibility and Impairs Anti-Vi Polysaccharide IgG Responses in a Mouse Model of Typhoid. Immunohorizons 2022; 6:807-816. [PMID: 36480484 DOI: 10.4049/immunohorizons.2200088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 12/13/2022] Open
Abstract
Circulating IgM present in the body prior to any apparent Ag exposure is referred to as natural IgM. Natural IgM provides protective immunity against a variety of pathogens. Salmonella enterica serovar Typhi (S. Typhi) is the causative agent of typhoid fever in humans. Because mice are not permissive to S. Typhi infection, we employed a murine model of typhoid using S. enterica serovar Typhimurium expressing the Vi polysaccharide (ViPS) of S. Typhi (S. Typhimurium strain RC60) to evaluate the role of natural IgM in pathogenesis. We found that natural mouse IgM binds to S. Typhi and S. Typhimurium. The severity of S. Typhimurium infection in mice is dependent on presence of the natural resistance-associated macrophage protein 1 (Nramp1) allele; therefore, we infected mice deficient in secreted form of IgM (sIgM) on either a Nramp1-resistant (129S) or -susceptible (C57BL/6J) background. We found that the lack of natural IgM results in a significantly increased susceptibility and an exaggerated liver pathology regardless of the route of infection or the Nramp1 allele. Reconstitution of sIgM-/- mice with normal mouse serum or purified polyclonal IgM restored the resistance to that of sIgM+/+ mice. Furthermore, immunization of sIgM-/- mice with heat-killed S. Typhi induced a significantly reduced anti-ViPS IgG and complement-dependent bactericidal activity against S. Typhi in vitro, compared with that of sIgM+/+ mice. These findings indicate that natural IgM is an important factor in reducing the typhoid severity and inducing an optimal anti-ViPS IgG response to vaccination.
Collapse
Affiliation(s)
- Akhil S Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA.,Department of Microbiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Matthew P Cravens
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Justin A Walker
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Dania Gulandijany
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Gregory S Dickinson
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Gudrun F Debes
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Dieter M Schifferli
- Department of Microbiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA; and
| | - Kishore R Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Kishore R Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
39
|
Co-administration of FVIII with IVIG reduces immune response to FVIII in hemophilia A mice. Sci Rep 2022; 12:20074. [PMID: 36418333 PMCID: PMC9684572 DOI: 10.1038/s41598-022-19392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/03/2022] [Indexed: 11/24/2022] Open
Abstract
Hemophilia A is an X-linked recessive congenital bleeding disorder. Exogenous infusion of FVIII is the treatment of choice, and the development of immunoglobulins against FVIII (inhibitors) remains the major challenge in clinical management of the disease. Here, we investigated the effect of co-administration of FVIII with intravenous immunoglobulin (IVIG) on the development of inhibitors in previously untreated hemophilia A mice. A group of hemophilia A mice (C57BL/6FVIII-/-) received weekly injections of recombinant human FVIII (rFVIII) for twelve consecutive weeks while a second group received co-injections of rFVIII + IVIG. An in-house enzyme-linked immunosorbent assay (ELISA) was designed to detect antibodies to rFVIII. Every mouse in the first group developed antibodies to rFVIII. In contrast, mice treated with rFVIII + IVIG showed significantly lower antibody titers. Interestingly, when co-administration of IVIG was discontinued after 12 weeks in some mice (rFVIII continued), these mice experienced an increase in antibody titer. In contrast, mice that continued to receive rFVIII + IVIG retained significantly lower titers. In conclusion, prophylactic rFVIII co-administration with IVIG modulated the immune response to FVIII and resulted in decreased anti-FVIII antibody titer. These findings suggest that co-injection therapy with IVIG could potentially be effective in the management of hemophilia A patients at risk of inhibitor development.
Collapse
|
40
|
Brishti A, Germundson-Hermanson DL, Smith NA, Kearney AE, Warda Y, Nagamoto-Combs K. Asymptomatic sensitization to a cow's milk protein induces sustained neuroinflammation and behavioral changes with chronic allergen exposure. FRONTIERS IN ALLERGY 2022; 3:870628. [PMID: 36157272 PMCID: PMC9490182 DOI: 10.3389/falgy.2022.870628] [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] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/22/2022] [Indexed: 12/04/2022] Open
Abstract
Mouse models of food allergy have contributed to our understanding of various aspects of the disease, including susceptibilities, symptom spectra, cellular mechanisms, and therapeutic approaches. Previously, we used a mouse model of non-anaphylactic cow's milk allergy (CMA) and investigated sex- and strain-dependent differences in immunological, neurological, and behavioral sequelae. We showed that male C57BL/6J mice sensitized to a bovine whey protein, β-lactoglobulin (BLG; Bos d 5), exhibited anxiety- and depression-like behavior upon acute allergen challenge. Systemic levels of BLG-specific immunoglobulins, cytokines and chemokines were also elevated in the sensitized mice. Furthermore, neuroinflammation and intestinal dysbiosis were evident as the possible causes of the altered behavior. To assess whether frequent allergen exposure influences CMA-associated pathologies over an extended period in this subclinical model, we placed BLG-sensitized mice on a whey protein (WP)-containing or whey-free control (CTL) diet for 3 months. As expected, allergen-specific IgE was significantly elevated in the plasma after completing the 5-week sensitization phase. However, the IgE levels declined in both diet groups after 3 months. In contrast, allergen-specific IgG1 stayed elevated in sensitized mice with the CTL diet, and the WP diet to a lesser extent. Interestingly, BLG-sensitized mice on the WP diet exhibited anxiety-like behavior and a trend toward spatial memory decline compared to the sham or the sensitized mice on the CTL diet. Moreover, increased immunoreactivities for GFAP and Iba1 and elevated levels of CXCL13 and CCL12, the chemokines involved in central leukocyte recruitment and other neurological diseases, were also observed in the brain. We demonstrated that sensitization to the whey protein, particularly with continuous allergen exposure, resulted in persistent neuroinflammation and associated behavioral changes despite lowered allergen-specific immunoglobulin levels. These results suggested that continuous consumption of the offending allergen may lead to adverse consequences in the brain even after desensitization.
Collapse
Affiliation(s)
- Afrina Brishti
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Danielle L. Germundson-Hermanson
- Clinical and Translational Science Graduate Program, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Nicholas A. Smith
- Clinical and Translational Science Graduate Program, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Angela E. Kearney
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Yassmine Warda
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Kumi Nagamoto-Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
41
|
Zhai Y, Dong S, Li H, Zhang Y, Shami P, Chen M. Antibody-mediated depletion of programmed death 1-positive (PD-1 +) cells. J Control Release 2022; 349:425-433. [PMID: 35820540 PMCID: PMC10699550 DOI: 10.1016/j.jconrel.2022.07.010] [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: 03/08/2022] [Revised: 06/17/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
PD-1 immune checkpoint has been intensively investigated in pathogenesis and treatments for cancer and autoimmune diseases. Cells that express PD-1 (PD-1+ cells) draw ever-increasing attention in cancer and autoimmune disease research although the role of PD-1+ cells in the progression and treatments of these diseases remains largely ambiguous. One definite approach to elucidate their roles is to deplete these cells in disease settings and examine how the depletion impacts disease progression and treatments. To execute the depletion, we designed and generated the first depleting antibody (D-αPD-1) that specifically ablates PD-1+ cells. D-αPD-1 has the same variable domains as an anti-mouse PD-1 blocking antibody (RMP1-14). The constant domains of D-αPD-1 were derived from mouse IgG2a heavy and κ-light chain, respectively. D-αPD-1 was verified to bind with mouse PD-1 as well as mouse FcγRIV, an immuno-activating Fc receptor. The cell depletion effect of D-αPD-1 was confirmed in vivo using a PD-1+ cell transferring model. Since transferred PD-1+ cells, EL4 cells, are tumorigenic and EL4 tumors are lethal to host mice, the depleting effect of D-αPD-1 was also manifested by an absolute survival among the antibody-treated mice while groups receiving control treatments had median survival time of merely approximately 30 days. Furthermore, we found that D-αPD-1 leads to elimination of PD-1+ cells through antibody-dependent cell-mediate phagocytosis (ADCP) and complement-dependent cytotoxicity (CDC) mechanisms. These results, altogether, confirmed the specificity and effectiveness of D-αPD-1. The results also highlighted that D-αPD-1 is a robust tool to study PD-1+ cells in cancer and autoimmune diseases and a potential therapeutic for these diseases.
Collapse
Affiliation(s)
- Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Shuyun Dong
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Haojia Li
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Yue Zhang
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Paul Shami
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Mingnan Chen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
42
|
Kinetic of the Antibody Response Following AddaVax-Adjuvanted Immunization with Recombinant Influenza Antigens. Vaccines (Basel) 2022; 10:vaccines10081315. [PMID: 36016202 PMCID: PMC9415944 DOI: 10.3390/vaccines10081315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Notwithstanding the current SARS-CoV-2 pandemic, influenza virus infection still represents a global health concern in terms of hospitalizations and possible pandemic threats. The objective of next-generation influenza vaccines is not only to increase the breadth of response but also to improve the elicitation of an effective and robust immune response, especially in high-risk populations. To achieve this second objective, the administration of adjuvanted influenza vaccines has been considered. In this regard, the monitoring and characterization of the antibody response associated with the administration of adjuvanted vaccines has been evaluated in this study in order to shed light on the kinetic, magnitude and subclass usage of antibody secreting cells (ASCs) as well as of circulating antigen-specific serum antibodies. Specifically, we utilized the DBA/2J mouse model to assess the kinetic, magnitude and IgG subclass usage of the antibody response following an intramuscular (IM) or intraperitoneal (IP) immunization regimen with AddaVax-adjuvanted bivalent H1N1 and H3N2 computationally optimized broadly reactive antigen (COBRA) influenza recombinant hemagglutinins (rHAs). While the serological evaluation revealed a homogeneous kinetic of the antibody response, the detection of the ASCs through a FluoroSpot platform revealed a different magnitude, subclass usage and kinetic of the antigen-specific IgG secreting cells peaking at day 5 and day 9 following the IP and IM immunization, respectively.
Collapse
|
43
|
Pearson JA, Ding H, Hu C, Peng J, Galuppo B, Wong FS, Caprio S, Santoro N, Wen L. IgM-associated gut bacteria in obesity and type 2 diabetes in C57BL/6 mice and humans. Diabetologia 2022; 65:1398-1411. [PMID: 35587276 PMCID: PMC9283171 DOI: 10.1007/s00125-022-05711-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 02/25/2022] [Indexed: 02/03/2023]
Abstract
AIMS/HYPOTHESIS IgM is the primary antibody produced by B cells and we hypothesise that IgM antibodies to gut microbiota may play a role in immunometabolism in obesity and type 2 diabetes. To test our hypothesis, we used B6 mice deficient in activation-induced cytidine deaminase (Aid-/- [also known as Aicda-/-]) which secrete only IgM antibodies, and human faecal samples. METHODS We studied the immunometabolic effects and gut microbial changes in high-fat-diet-induced obesity (HFDIO) in Aid-/- B6 mice compared with wild-type mice. To determine similarities between mice and humans, human stool samples were collected from children and adolescents who were obese with normal glucose tolerance (NGT), obese with glucose intolerance (IGT), or obese and newly diagnosed with type 2 diabetes, for faecal microbiota transplant (FMT) into germ-free (GF) B6 mice and we assessed IgM-bound bacteria and immune responses. RESULTS Compared with wild-type mice, Aid-/- B6 mice developed exacerbated HFDIO due to abundant levels of IgM. FMT from Aid-/- B6 to GF B6 mice promoted greater weight gain in recipient mice compared with FMT using wild-type mouse faecal microbiota. Obese youth with type 2 diabetes had more IgM-bound gut bacteria. Using the stools from the obese youth with type 2 diabetes for FMT to GF B6 mice, we observed that the gut microbiota promoted body weight gain and impaired glucose tolerance in the recipient GF B6 mice. Importantly, some clinical features of these obese young individuals were mirrored in the GF B6 mice following FMT. CONCLUSIONS/INTERPRETATION Our results suggest that IgM-bound gut microbiota may play an important role in the immuno-pathogenesis of obesity and type 2 diabetes, and provide a novel link between IgM in obesity and type 2 diabetes in both mice and humans. DATA AVAILABILITY The 16s rRNA sequencing datasets supporting the current study have been deposited in the NCBI SRA public repository ( https://www.ncbi.nlm.nih.gov/sra ; accession no. SAMN18796639).
Collapse
Affiliation(s)
- James A Pearson
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA.
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK.
| | - Heyuan Ding
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Changyun Hu
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
- Adept Therapeutics, Inc., Beverly, MA, USA
| | - Jian Peng
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
| | - Brittany Galuppo
- Department of Pediatrics, School of Medicine, Yale University, New Haven, CT, USA
| | - F Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Sonia Caprio
- Department of Pediatrics, School of Medicine, Yale University, New Haven, CT, USA
| | - Nicola Santoro
- Department of Pediatrics, School of Medicine, Yale University, New Haven, CT, USA
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
44
|
El-Shershaby HM, Farrag NS, Ebeid NH, Moustafa KA. Radiolabeling and cytotoxicity of monoclonal antibody Isatuximab functionalized silver nanoparticles on the growth of multiple myeloma. Int J Pharm 2022; 624:122019. [PMID: 35842081 DOI: 10.1016/j.ijpharm.2022.122019] [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: 04/19/2022] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/18/2022]
Abstract
The goal of this article was to develop a new therapeutic strategy based on nanotechnology for multiple myeloma (MM) treatment which shows a synergism of different mechanisms. In this concern, 12.9 nm-sized silver nanoparticles (AgNPs) were prepared and functionalized with Isatuximab, anti-MM monoclonal antibody (mAb). Furthermore, the synthesized nanocomposite was radiolabelled with iodine-131 radionuclide and yielded 95.5 ± 1.5%. Then, the synergistic MM-proliferation inhibition efficacy of the radionanocomposite (131I-Isatuximab/AgNPs) was explored in-vitro in comparison to each single agent. The MTT investigation showed that the antiproliferation effect of 131I-Isatuximab/AgNPs increased by more than 1.5 fold if compared with Isatuximab, AgNPs, Isatuximab/AgNPs or 131I-Isatuximab. Additionally, 131I-Isatuximab/AgNPs exhibited an apoptotic effect on MM cells which was more than that of Isatuximab, AgNPs, Isatuximab/AgNPs or 131I-Isatuximab by 2, 1.8, 1.7 and 1.5 folds, respectively. In conclusion, the results expressed 131I-Isatuximab/AgNPs as a potential new anti-MM agent.
Collapse
Affiliation(s)
- Hanan M El-Shershaby
- Labeled Compounds Department, Hot Labs. Center, Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| | - Nourihan S Farrag
- Labeled Compounds Department, Hot Labs. Center, Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt.
| | - Nahed H Ebeid
- Labeled Compounds Department, Hot Labs. Center, Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| | - Kamel A Moustafa
- Labeled Compounds Department, Hot Labs. Center, Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| |
Collapse
|
45
|
Chung CK, Beekmann U, Kralisch D, Bierau K, Chan A, Ossendorp F, Cruz LJ. Bacterial Cellulose as Drug Delivery System for Optimizing Release of Immune Checkpoint Blocking Antibodies. Pharmaceutics 2022; 14:1351. [PMID: 35890247 PMCID: PMC9316226 DOI: 10.3390/pharmaceutics14071351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023] Open
Abstract
Immune checkpoint blocking therapy is a promising cancer treatment modality, though it has limitations such as systemic toxicity, which can often be traced to uncontrolled antibody spread. Controlling antibody release with delivery systems is, therefore, an attractive approach to reduce systemic antibody spread and potentially mitigate the side effects of checkpoint immunotherapy. Here, bacterial cellulose (BC) was produced and investigated as a delivery system for optimizing checkpoint-blocking antibody delivery. BC was produced in 24-well plates, and afterward, the edges were removed to obtain square-shaped BC samples with a surface of ~49 mm2. This customization was necessary to allow smooth in vivo implantation. Scanning electron microscopy revealed the dense cellulose network within BC. Human IgG antibody was included as the model antibody for loading and release studies. IgG antibody solution was injected into the center of BC samples. In vitro, all IgG was released within 24 to 48 h. Cell culture experiments demonstrated that BC neither exerted cytotoxic effects nor induced dendritic cell activation. Antibody binding assays demonstrated that BC does not hamper antibody function. Finally, antibody-loaded BC was implanted in mice, and serum measurements revealed that BC significantly reduced IgG and anti-CTLA-4 spread in mice. BC implantation did not induce side effects in mice. Altogether, BC is a promising and safe delivery system for optimizing the delivery and release of checkpoint-blocking antibodies.
Collapse
Affiliation(s)
- Chih Kit Chung
- Department of Radiology, Division Translational Nanobiomaterials and Imaging, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
- Percuros B.V., Zernikedreef 8, 2333 CL Leiden, The Netherlands;
- JeNaCell GmbH, Göschwitzer Straße 22, 07745 Jena, Germany; (U.B.); (D.K.)
| | - Uwe Beekmann
- JeNaCell GmbH, Göschwitzer Straße 22, 07745 Jena, Germany; (U.B.); (D.K.)
| | - Dana Kralisch
- JeNaCell GmbH, Göschwitzer Straße 22, 07745 Jena, Germany; (U.B.); (D.K.)
| | - Katja Bierau
- Pilotality, Eerbeeklaan 42, 2573 HT Gravenhage, The Netherlands;
| | - Alan Chan
- Percuros B.V., Zernikedreef 8, 2333 CL Leiden, The Netherlands;
| | - Ferry Ossendorp
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Luis J. Cruz
- Department of Radiology, Division Translational Nanobiomaterials and Imaging, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| |
Collapse
|
46
|
Generation of antagonistic monoclonal antibodies against the neoepitope of active mouse interleukin (IL)-18 cleaved by inflammatory caspases. Arch Biochem Biophys 2022; 727:109322. [PMID: 35709966 DOI: 10.1016/j.abb.2022.109322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/23/2022] [Accepted: 06/09/2022] [Indexed: 11/23/2022]
Abstract
Interleukin 18 (IL-18) is a member of the IL-1 family and plays an important role in both the innate and acquired immune systems. It is constitutively expressed as an inactive precursor (24 kDa) in various cell types, and the mature IL-18 (18 kDa) cleaved by inflammatory caspase-1/4 binds to the interleukin-18 receptor, thereby activating downstream signaling pathways. We previously generated anti-human IL-18 antibodies that specifically recognize the human IL-18 neoepitope cleaved by inflammatory caspase-1/4. Because the N-terminal amino acid sequences of the neoepitopes are different between human IL-18 and mouse IL-18, the anti-human IL-18 neoepitope antibodies do not recognize mouse mature IL-18. We have now generated novel anti-mouse IL-18 neoepitope antibodies. We also confirmed CXCL2 secretion from P-815 mouse cells by mouse IL-18 stimulation, and established a simple assay to evaluate the activity of mouse IL-18. Using this evaluation system, we confirmed that the anti-mouse IL-18 neoepitope antibodies could inhibit mouse IL-18. By demonstrating the therapeutic efficacy of the anti-mouse IL-18 neoepitope and function-blocking mAbs established in the present study in mouse models, corresponding to human inflammatory diseases in which IL-18 may be involved, such as inflammatory bowel diseases, we can provide the proof-of-concept that the previously established anti-human IL-18 neoepitope and function-blocking mAbs work in human inflammatory disorders corresponding to mouse models.
Collapse
|
47
|
Yang Z, Rajendran S, Spires V, Poirson B, Gururajan M, Lin Z, Arbanas J, Krystek S, Loy J, Cheng Y, Carl S, Pace S, Wang Y, Mehl J, Xu S, Vasudevan K, Broz M, Lehman-McKeeman LD, Morin P, Graziano RF. Target-Mediated Drug Disposition Affects the Pharmacokinetics of Interleukin-10 Fc-fusion Proteins at Pharmacologically Active Doses. Drug Metab Dispos 2022; 50:898-908. [PMID: 35545256 DOI: 10.1124/dmd.121.000799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Fragment crystallizable (Fc) fusion is commonly used for extending the half-life of biotherapeutics such as cytokines. In this work, we studied the pharmacokinetics of Fc-fused interleukin-10 (IL-10) proteins that exhibited potent anti-tumor activity in mouse syngeneic tumor models. At pharmacologically active doses of {greater than or equal to}0.1 mg/kg, both mouse Fc-mouse IL-10 and human Fc-human IL-10, constructed as the C-terminus of the Fc domain fused with IL-10 via a glycine-serine polypeptide linker, exhibited nonlinear pharmacokinetics after intravenous administration to mice at the doses of 0.05, 0.5, and 5 mg/kg. With a nominal dose ratio of 1:10:100; the ratio of the area under the curve for mouse Fc-mouse IL-10 and human Fc-human IL-10 was 1:181:1830 and 1:75:633, respectively. In contrast, recombinant mouse or human IL-10 proteins exhibited linear pharmacokinetics in mice. Compartmental analysis, using the Michaelis-Menten equation with the in vitro IL-10 receptor alpha binding affinity inputted as the Km, unified the pharmacokinetic data across the dose range. Additionally, non-target-mediated clearance estimated for fusion proteins was ~200-fold slower than that for cytokines, causing the manifestation of target-mediated drug disposition (TMDD) in the fusion protein pharmacokinetics. The experimental data generated with a mouse IL-10 receptor alpha-blocking antibody and a human Fc-human IL-10 mutant with a reduced receptor binding affinity showed significant improvements in pharmacokinetics, supporting TMDD as the cause of nonlinearity. Target expression and its effect on pharmacokinetics must be determined when considering using Fc as a half-life extension strategy, and pharmacokinetic evaluations need to be performed at a range of doses covering pharmacological activity. Significance Statement Target-mediated drug disposition can manifest to affect the pharmacokinetics of a fragment crystallizable (Fc)-fused cytokine when the non-target-mediated clearance of the cytokine is decreased due to neonatal Fc receptor-mediated recycling and molecular weight increases that reduce the renal clearance. The phenomenon was demonstrated with interleukin-10 Fc-fusion proteins in mice at pharmacologically active doses. Future drug designs using Fc as a half-life extension approach for cytokines need to consider target expression and its effect on pharmacokinetics at relevant doses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yun Wang
- Bristol Myers Squibb, United States
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Vattepu R, Sneed SL, Anthony RM. Sialylation as an Important Regulator of Antibody Function. Front Immunol 2022; 13:818736. [PMID: 35464485 PMCID: PMC9021442 DOI: 10.3389/fimmu.2022.818736] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/17/2022] [Indexed: 12/14/2022] Open
Abstract
Antibodies play a critical role in linking the adaptive immune response to the innate immune system. In humans, antibodies are categorized into five classes, IgG, IgM, IgA, IgE, and IgD, based on constant region sequence, structure, and tropism. In serum, IgG is the most abundant antibody, comprising 75% of antibodies in circulation, followed by IgA at 15%, IgM at 10%, and IgD and IgE are the least abundant. All human antibody classes are post-translationally modified by sugars. The resulting glycans take on many divergent structures and can be attached in an N-linked or O-linked manner, and are distinct by antibody class, and by position on each antibody. Many of these glycan structures on antibodies are capped by sialic acid. It is well established that the composition of the N-linked glycans on IgG exert a profound influence on its effector functions. However, recent studies have described the influence of glycans, particularly sialic acid for other antibody classes. Here, we discuss the role of glycosylation, with a focus on terminal sialylation, in the biology and function across all antibody classes. Sialylation has been shown to influence not only IgG, but IgE, IgM, and IgA biology, making it an important and unappreciated regulator of antibody function.
Collapse
Affiliation(s)
- Ravi Vattepu
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sunny Lyn Sneed
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Robert M Anthony
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
49
|
Blunck BN, Angelo LS, Henke D, Avadhanula V, Cusick M, Ferlic-Stark L, Zechiedrich L, Gilbert BE, Piedra PA. Adult Memory T Cell Responses to the Respiratory Syncytial Virus Fusion Protein During a Single RSV Season (2018-2019). Front Immunol 2022; 13:823652. [PMID: 35422803 PMCID: PMC9002099 DOI: 10.3389/fimmu.2022.823652] [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: 11/27/2021] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Respiratory Syncytial Virus (RSV) is ubiquitous and re-infection with both subtypes (RSV/A and RSV/B) is common. The fusion (F) protein of RSV is antigenically conserved, induces neutralizing antibodies, and is a primary target of vaccine development. Insight into the breadth and durability of RSV-specific adaptive immune response, particularly to the F protein, may shed light on susceptibility to re-infection. We prospectively enrolled healthy adult subjects (n = 19) and collected serum and peripheral blood mononuclear cells (PBMCs) during the 2018–2019 RSV season. Previously, we described their RSV-specific antibody responses and identified three distinct antibody kinetic profiles associated with infection status: uninfected (n = 12), acutely infected (n = 4), and recently infected (n = 3). In this study, we measured the longevity of RSV-specific memory T cell responses to the F protein following natural RSV infection. We stimulated PBMCs with overlapping 15-mer peptide libraries spanning the F protein derived from either RSV/A or RSV/B and found that memory T cell responses mimic the antibody responses for all three groups. The uninfected group had stable, robust memory T cell responses and polyfunctionality. The acutely infected group had reduced polyfunctionality of memory T cell response at enrollment compared to the uninfected group, but these returned to comparable levels by end-of-season. The recently infected group, who were unable to maintain high levels of RSV-specific antibody following infection, similarly had decreased memory T cell responses and polyfunctionality during the RSV season. We observed subtype-specific differences in memory T cell responses and polyfunctionality, with RSV/A stimulating stronger memory T cell responses with higher polyfunctionality even though RSV/B was the dominant subtype in circulation. A subset of individuals demonstrated an overall deficiency in the generation of a durable RSV-specific adaptive immune response. Because memory T cell polyfunctionality may be associated with protection against re-infection, this latter group would likely be at greater risk of re-infection. Overall, these results expand our understanding of the longevity of the adaptive immune response to the RSV fusion protein and should be considered in future vaccine development efforts.
Collapse
Affiliation(s)
- Brittani N Blunck
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Laura S Angelo
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - David Henke
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Matthew Cusick
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Laura Ferlic-Stark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Lynn Zechiedrich
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, United States
| | - Brian E Gilbert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
50
|
Lindstad CB, du Pré MF, Stamnaes J, Sollid LM. Injection of prototypic celiac anti-transglutaminase 2 antibodies in mice does not cause enteropathy. PLoS One 2022; 17:e0266543. [PMID: 35385534 PMCID: PMC8985999 DOI: 10.1371/journal.pone.0266543] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/22/2022] [Indexed: 12/19/2022] Open
Abstract
Background Celiac disease is an autoimmune enteropathy driven by dietary intake of gluten proteins. Typical histopathologic features are villous flattening, crypt hyperplasia and infiltration of inflammatory cells in the intestinal epithelium and lamina propria. The disease is hallmarked by the gluten-dependent production of autoantibodies targeting the enzyme transglutaminase 2 (TG2). While these antibodies are specific and sensitive diagnostic markers of the disease, a role in the development of the enteropathy has never been established. Methods We addressed this question by injecting murine antibodies harboring the variable domains of a prototypic celiac anti-TG2 immunoglobulin into TG2-sufficient and TG2-deficient mice evaluating for celiac enteropathy. Results We found no histopathologic abnormalities nor clinical signs of disease related to the injection of anti-TG2 IgG or IgA. Conclusions Our findings do not support a direct role for secreted anti-TG2 antibodies in the development of the celiac enteropathy.
Collapse
Affiliation(s)
- Christian B. Lindstad
- K.G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
- Department of Immunology, University of Oslo, Oslo, Norway
| | - M. Fleur du Pré
- K.G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Jorunn Stamnaes
- K.G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
- Department of Immunology, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Ludvig M. Sollid
- K.G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
- Department of Immunology, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- * E-mail:
| |
Collapse
|