1
|
Creppy JR, Delache B, Lemaitre J, Horvat B, Vecellio L, Ducancel F. Administration of airborne pathogens in non-human primates. Inhal Toxicol 2024:1-26. [PMID: 39388247 DOI: 10.1080/08958378.2024.2412685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE Airborne pathogen scan penetrate in human respiratory tract and can cause illness. The use of animal models to predict aerosol deposition and study respiratory disease pathophysiology is therefore important for research and a prerequisite to test and study the mechanism of action of treatment. NHPs are relevant animal species for inhalation studies because of their similarities with humans in terms of anatomical structure, respiratory parameters and immune system. MATERIALS AND METHODS The aim of this review is to provide an overview of the state of the art of pathogen aerosol studies performed in non-human primates (NHPs). Herein, we present and discuss the deposition of aerosolized bacteria and viruses. In this review, we present important advantages of using NHPs as model for inhalation studies. RESULTS We demonstrate that deposition in the respiratory tract is not only a function of aerosol size but also the technique of administration influences the biological activity and site of aerosol deposition. Finally, we observe an influence of a region of pathogen deposition in the respiratory tract on the development of the pathophysiological effect in NHPs. CONCLUSION The wide range of methods used for the delivery of pathogento NHP respiratory airways is associated with varying doses and deposition profiles in the airways.
Collapse
Affiliation(s)
- Justina R Creppy
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Benoit Delache
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Julien Lemaitre
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Branka Horvat
- CIRI, Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, Université de Lyon, Lyon, France
| | - Laurent Vecellio
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Frédéric Ducancel
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
2
|
Frampton S, Smith R, Ferson L, Gibson J, Hollox EJ, Cragg MS, Strefford JC. Fc gamma receptors: Their evolution, genomic architecture, genetic variation, and impact on human disease. Immunol Rev 2024. [PMID: 39345014 DOI: 10.1111/imr.13401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Fc gamma receptors (FcγRs) are a family of receptors that bind IgG antibodies and interface at the junction of humoral and innate immunity. Precise regulation of receptor expression provides the necessary balance to achieve healthy immune homeostasis by establishing an appropriate immune threshold to limit autoimmunity but respond effectively to infection. The underlying genetics of the FCGR gene family are central to achieving this immune threshold by regulating affinity for IgG, signaling efficacy, and receptor expression. The FCGR gene locus was duplicated during evolution, retaining very high homology and resulting in a genomic region that is technically difficult to study. Here, we review the recent evolution of the gene family in mammals, its complexity and variation through copy number variation and single-nucleotide polymorphism, and impact of these on disease incidence, resolution, and therapeutic antibody efficacy. We also discuss the progress and limitations of current approaches to study the region and emphasize how new genomics technologies will likely resolve much of the current confusion in the field. This will lead to definitive conclusions on the impact of genetic variation within the FCGR gene locus on immune function and disease.
Collapse
Affiliation(s)
- Sarah Frampton
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Rosanna Smith
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Lili Ferson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Jane Gibson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Edward J Hollox
- Department of Genetics, Genomics and Cancer Sciences, College of Life Sciences, University of Leicester, Leicester, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Jonathan C Strefford
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
3
|
Gunes ME, Wolbrom DH, Nygaard ED, Manell E, Jordache P, Qudus S, Cadelina A, Weiner J, Nowak G. OMIP-108: 22-color flow cytometry panel for detection and monitoring of chimerism and immune reconstitution in porcine-to-baboon models of operational xenotransplant tolerance studies. Cytometry A 2024. [PMID: 39291632 DOI: 10.1002/cyto.a.24899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/01/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Affiliation(s)
- M Esad Gunes
- Columbia Center of Translational Immunology, Department of Medicine, Columbia University, New York, New York, USA
| | - Daniel H Wolbrom
- Columbia Center of Translational Immunology, Department of Medicine, Columbia University, New York, New York, USA
| | - Emilie Ditlev Nygaard
- Columbia Center of Translational Immunology, Department of Medicine, Columbia University, New York, New York, USA
| | - Elin Manell
- Columbia Center of Translational Immunology, Department of Medicine, Columbia University, New York, New York, USA
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Philip Jordache
- Columbia Center of Translational Immunology, Department of Medicine, Columbia University, New York, New York, USA
| | - Susan Qudus
- Columbia Center of Translational Immunology, Department of Medicine, Columbia University, New York, New York, USA
| | - Alexander Cadelina
- Columbia Center of Translational Immunology, Department of Medicine, Columbia University, New York, New York, USA
| | - Joshua Weiner
- Columbia Center of Translational Immunology, Department of Medicine, Columbia University, New York, New York, USA
- Department of Surgery, Columbia University, New York, New York, USA
| | - Greg Nowak
- Columbia Center of Translational Immunology, Department of Medicine, Columbia University, New York, New York, USA
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
4
|
Edgar JE, Bournazos S. Fc-FcγR interactions during infections: From neutralizing antibodies to antibody-dependent enhancement. Immunol Rev 2024. [PMID: 39268652 DOI: 10.1111/imr.13393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Advances in antibody technologies have resulted in the development of potent antibody-based therapeutics with proven clinical efficacy against infectious diseases. Several monoclonal antibodies (mAbs), mainly against viruses such as SARS-CoV-2, HIV-1, Ebola virus, influenza virus, and hepatitis B virus, are currently undergoing clinical testing or are already in use. Although these mAbs exhibit potent neutralizing activity that effectively blocks host cell infection, their antiviral activity results not only from Fab-mediated virus neutralization, but also from the protective effector functions mediated through the interaction of their Fc domains with Fcγ receptors (FcγRs) on effector leukocytes. Fc-FcγR interactions confer pleiotropic protective activities, including the clearance of opsonized virions and infected cells, as well as the induction of antiviral T-cell responses. However, excessive or inappropriate activation of specific FcγR pathways can lead to disease enhancement and exacerbated pathology, as seen in the context of dengue virus infections. A comprehensive understanding of the diversity of Fc effector functions during infection has guided the development of engineered antiviral antibodies optimized for maximal effector activity, as well as the design of targeted therapeutic approaches to prevent antibody-dependent enhancement of disease.
Collapse
Affiliation(s)
- Julia E Edgar
- The London School of Hygiene and Tropical Medicine, London, UK
| | - Stylianos Bournazos
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
5
|
Nemphos SM, Green HC, Prusak JE, Fell SL, Goff K, Varnado M, Didier K, Guy N, Moström MJ, Tatum C, Massey C, Barnes MB, Rowe LA, Allers C, Blair RV, Embers ME, Maness NJ, Marx PA, Grasperge B, Kaur A, De Paris K, Shaffer JG, Hensley-McBain T, Londono-Renteria B, Manuzak JA. Elevated Inflammation Associated with Markers of Neutrophil Function and Gastrointestinal Disruption in Pilot Study of Plasmodium fragile Co-Infection of ART-Treated SIVmac239+ Rhesus Macaques. Viruses 2024; 16:1036. [PMID: 39066199 PMCID: PMC11281461 DOI: 10.3390/v16071036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Human immunodeficiency virus (HIV) and malaria, caused by infection with Plasmodium spp., are endemic in similar geographical locations. As a result, there is high potential for HIV/Plasmodium co-infection, which increases the pathology of both diseases. However, the immunological mechanisms underlying the exacerbated disease pathology observed in co-infected individuals are poorly understood. Moreover, there is limited data available on the impact of Plasmodium co-infection on antiretroviral (ART)-treated HIV infection. Here, we used the rhesus macaque (RM) model to conduct a pilot study to establish a model of Plasmodium fragile co-infection during ART-treated simian immunodeficiency virus (SIV) infection, and to begin to characterize the immunopathogenic effect of co-infection in the context of ART. We observed that P. fragile co-infection resulted in parasitemia and anemia, as well as persistently detectable viral loads (VLs) and decreased absolute CD4+ T-cell counts despite daily ART treatment. Notably, P. fragile co-infection was associated with increased levels of inflammatory cytokines, including monocyte chemoattractant protein 1 (MCP-1). P. fragile co-infection was also associated with increased levels of neutrophil elastase, a plasma marker of neutrophil extracellular trap (NET) formation, but significant decreases in markers of neutrophil degranulation, potentially indicating a shift in the neutrophil functionality during co-infection. Finally, we characterized the levels of plasma markers of gastrointestinal (GI) barrier permeability and microbial translocation and observed significant correlations between indicators of GI dysfunction, clinical markers of SIV and Plasmodium infection, and neutrophil frequency and function. Taken together, these pilot data verify the utility of using the RM model to examine ART-treated SIV/P. fragile co-infection, and indicate that neutrophil-driven inflammation and GI dysfunction may underlie heightened SIV/P. fragile co-infection pathogenesis.
Collapse
Affiliation(s)
- Sydney M. Nemphos
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Hannah C. Green
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - James E. Prusak
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Sallie L. Fell
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Kelly Goff
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Megan Varnado
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Kaitlin Didier
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Natalie Guy
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Matilda J. Moström
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Coty Tatum
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Chad Massey
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Mary B. Barnes
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Lori A. Rowe
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Carolina Allers
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Robert V. Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Monica E. Embers
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Nicholas J. Maness
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Preston A. Marx
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Tropical Medicine and Infectious Disease, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
| | - Brooke Grasperge
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Amitinder Kaur
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27559, USA
| | - Jeffrey G. Shaffer
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | | | - Berlin Londono-Renteria
- Department of Tropical Medicine and Infectious Disease, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
| | - Jennifer A. Manuzak
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Tropical Medicine and Infectious Disease, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
| |
Collapse
|
6
|
Clark J, Hoxie I, Adelsberg DC, Sapse IA, Andreata-Santos R, Yong JS, Amanat F, Tcheou J, Raskin A, Singh G, González-Domínguez I, Edgar JE, Bournazos S, Sun W, Carreño JM, Simon V, Ellebedy AH, Bajic G, Krammer F. Protective effect and molecular mechanisms of human non-neutralizing cross-reactive spike antibodies elicited by SARS-CoV-2 mRNA vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582613. [PMID: 38464151 PMCID: PMC10925278 DOI: 10.1101/2024.02.28.582613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Neutralizing antibodies correlate with protection against SARS-CoV-2. Recent studies, however, show that binding antibody titers, in the absence of robust neutralizing activity, also correlate with protection from disease progression. Non-neutralizing antibodies cannot directly protect from infection but may recruit effector cells thus contribute to the clearance of infected cells. Also, they often bind conserved epitopes across multiple variants. We characterized 42 human mAbs from COVID-19 vaccinated individuals. Most of these antibodies exhibited no neutralizing activity in vitro but several non-neutralizing antibodies protected against lethal challenge with SARS-CoV-2 in different animal models. A subset of those mAbs showed a clear dependence on Fc-mediated effector functions. We determined the structures of three non-neutralizing antibodies with two targeting the RBD, and one that targeting the SD1 region. Our data confirms the real-world observation in humans that non-neutralizing antibodies to SARS-CoV-2 can be protective.
Collapse
Affiliation(s)
- Jordan Clark
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Irene Hoxie
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel C. Adelsberg
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Iden A. Sapse
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Andreata-Santos
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Retrovirology Laboratory, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Jeremy S. Yong
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Johnstone Tcheou
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ariel Raskin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Julia E. Edgar
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA
| | - Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Goran Bajic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
7
|
Conley HE, He MM, Easterhoff D, Kirshner HF, Cocklin SL, Meyer J, Hoxie T, Berry M, Bradley T, Tolbert WD, Pazgier M, Tomaras GD, Schmitz JE, Moody MA, Wiehe K, Pollara J. Defining genetic diversity of rhesus macaque Fcγ receptors with long-read RNA sequencing. Front Immunol 2024; 14:1306292. [PMID: 38264644 PMCID: PMC10803544 DOI: 10.3389/fimmu.2023.1306292] [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: 10/03/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
Fcγ receptors (FcγRs) are membrane-bound glycoproteins that bind to the fragment crystallizable (Fc) constant regions of IgG antibodies. Interactions between IgG immune complexes and FcγRs can initiate signal transduction that mediates important components of the immune response including activation of immune cells for clearance of opsonized pathogens or infected host cells. In humans, many studies have identified associations between FcγR gene polymorphisms and risk of infection, or progression of disease, suggesting a gene-level impact on FcγR-dependent immune responses. Rhesus macaques are an important translational model for most human health interventions, yet little is known about the breadth of rhesus macaque FcγR genetic diversity. This lack of knowledge prevents evaluation of the impact of FcγR polymorphisms on outcomes of preclinical studies performed in rhesus macaques. In this study we used long-read RNA sequencing to define the genetic diversity of FcγRs in 206 Indian-origin Rhesus macaques, Macaca mulatta. We describe the frequency of single nucleotide polymorphisms, insertions, deletions, frame-shift mutations, and isoforms. We also index the identified diversity using predicted and known rhesus macaque FcγR and Fc-FcγR structures. Future studies that define the functional significance of this genetic diversity will facilitate a better understanding of the correlation between human and macaque FcγR biology that is needed for effective translation of studies with antibody-mediated outcomes performed in rhesus macaques.
Collapse
Affiliation(s)
- Haleigh E. Conley
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Max M. He
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - David Easterhoff
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Hélène Fradin Kirshner
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Sarah L. Cocklin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Jacob Meyer
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Taylor Hoxie
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Madison Berry
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Todd Bradley
- Genomic Medicine Center, Children’s Mercy Kansas City, Kansas City, MO, United States
| | - William D. Tolbert
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Marzena Pazgier
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Georgia D. Tomaras
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Joern E. Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Michael Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Justin Pollara
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
8
|
Tuyishime M, Spreng RL, Hueber B, Nohara J, Goodman D, Chan C, Barfield R, Beck WE, Jha S, Asdell S, Wiehe K, He MM, Easterhoff D, Conley HE, Hoxie T, Gurley T, Jones C, Adhikary ND, Villinger F, Thomas R, Denny TN, Moody MA, Tomaras GD, Pollara J, Reeves RK, Ferrari G. Multivariate analysis of FcR-mediated NK cell functions identifies unique clustering among humans and rhesus macaques. Front Immunol 2023; 14:1260377. [PMID: 38124734 PMCID: PMC10732150 DOI: 10.3389/fimmu.2023.1260377] [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: 07/17/2023] [Accepted: 10/04/2023] [Indexed: 12/23/2023] Open
Abstract
Rhesus macaques (RMs) are a common pre-clinical model used to test HIV vaccine efficacy and passive immunization strategies. Yet, it remains unclear to what extent the Fc-Fc receptor (FcR) interactions impacting antiviral activities of antibodies in RMs recapitulate those in humans. Here, we evaluated the FcR-related functionality of natural killer cells (NKs) from peripheral blood of uninfected humans and RMs to identify intra- and inter-species variation. NKs were screened for FcγRIIIa (human) and FcγRIII (RM) genotypes (FcγRIII(a)), receptor signaling, and antibody-dependent cellular cytotoxicity (ADCC), the latter mediated by a cocktail of monoclonal IgG1 antibodies with human or RM Fc. FcγRIII(a) genetic polymorphisms alone did not explain differences in NK effector functionality in either species cohort. Using the same parameters, hierarchical clustering separated each species into two clusters. Importantly, in principal components analyses, ADCC magnitude, NK contribution to ADCC, FcγRIII(a) cell-surface expression, and frequency of phosphorylated CD3ζ NK cells all contributed similarly to the first principal component within each species, demonstrating the importance of measuring multiple facets of NK cell function. Although ADCC potency was similar between species, we detected significant differences in frequencies of NK cells and pCD3ζ+ cells, level of cell-surface FcγRIII(a) expression, and NK-mediated ADCC (P<0.001), indicating that a combination of Fc-FcR parameters contribute to overall inter-species functional differences. These data strongly support the importance of multi-parameter analyses of Fc-FcR NK-mediated functions when evaluating efficacy of passive and active immunizations in pre- and clinical trials and identifying correlates of protection. The results also suggest that pre-screening animals for multiple FcR-mediated NK function would ensure even distribution of animals among treatment groups in future preclinical trials.
Collapse
Affiliation(s)
- Marina Tuyishime
- Department of Surgery, Duke University, Durham, NC, United States
| | - Rachel L. Spreng
- Duke Human Vaccine Institute, Durham, NC, United States
- Center for Human Systems Immunology, Durham, NC, United States
| | - Brady Hueber
- Center for Human Systems Immunology, Durham, NC, United States
| | - Junsuke Nohara
- Department of Surgery, Duke University, Durham, NC, United States
| | - Derrick Goodman
- Department of Surgery, Duke University, Durham, NC, United States
- Center for Human Systems Immunology, Durham, NC, United States
| | - Cliburn Chan
- Center for Human Systems Immunology, Durham, NC, United States
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, United States
| | - Richard Barfield
- Center for Human Systems Immunology, Durham, NC, United States
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, United States
| | - Whitney E. Beck
- Department of Surgery, Duke University, Durham, NC, United States
| | - Shalini Jha
- Department of Surgery, Duke University, Durham, NC, United States
| | - Stephanie Asdell
- Department of Surgery, Duke University, Durham, NC, United States
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Durham, NC, United States
- Department of Medicine, Duke University, Durham, NC, United States
| | - Max M. He
- Duke Human Vaccine Institute, Durham, NC, United States
| | | | | | - Taylor Hoxie
- Duke Human Vaccine Institute, Durham, NC, United States
| | | | | | - Nihar Deb Adhikary
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, United States
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, United States
| | - Rasmi Thomas
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Thomas N. Denny
- Duke Human Vaccine Institute, Durham, NC, United States
- Department of Medicine, Duke University, Durham, NC, United States
| | - Michael Anthony Moody
- Duke Human Vaccine Institute, Durham, NC, United States
- Department of Pediatrics, Duke University, Durham, NC, United States
- Department of Integrative Immunobiology, Duke University, Durham, NC, United States
| | - Georgia D. Tomaras
- Department of Surgery, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Durham, NC, United States
- Center for Human Systems Immunology, Durham, NC, United States
- Department of Integrative Immunobiology, Duke University, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| | - Justin Pollara
- Department of Surgery, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Durham, NC, United States
- Center for Human Systems Immunology, Durham, NC, United States
| | - R. Keith Reeves
- Department of Surgery, Duke University, Durham, NC, United States
- Center for Human Systems Immunology, Durham, NC, United States
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Guido Ferrari
- Department of Surgery, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Durham, NC, United States
- Center for Human Systems Immunology, Durham, NC, United States
| |
Collapse
|
9
|
Grimaldi C, Ibraghimov A, Kiessling A, Rattel B, Ji C, Fuller CL, Brennan FR, Regenass-Lechner F, Shenton J, Price KD, Piché MS, Steeves MA, Prell R, Dudal S, Kronenberg S, Freebern W, Blanset D. Current nonclinical approaches for immune assessments of immuno-oncology biotherapeutics. Drug Discov Today 2023; 28:103440. [PMID: 36375739 DOI: 10.1016/j.drudis.2022.103440] [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: 06/23/2022] [Revised: 08/30/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Harnessing the immune system to kill tumors has been revolutionary and, as a result, has had an enormous benefit for patients in extending life and resulting in effective cures in some. However, activation of the immune system can come at the cost of undesirable adverse events such as cytokine release syndrome, immune-related adverse events, on-target/off-tumor toxicity, neurotoxicity and tumor lysis syndrome, which are safety risks that can be challenging to assess non-clinically. This article provides a review of the biology and mechanisms that can result in immune-mediated adverse effects and describes industry approaches using in vitro and in vivo models to aid in the nonclinical safety risk assessments for immune-oncology modalities. Challenges and limitations of knowledge and models are also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Sherri Dudal
- Roche Pharmaceutical Research and Early Development, United States
| | - Sven Kronenberg
- Roche Pharmaceutical Research and Early Development, United States
| | | | - Diann Blanset
- Boehringer Ingelheim Pharmaceuticals, Inc., United States.
| |
Collapse
|
10
|
Adedeji AO, Zhong F, Getz JA, Zhong Z, Halpern W. Neutropenia in Cynomolgus Monkeys With Anti-Drug Antibodies Associated With Administration of Afucosylated Humanized Monoclonal Antibodies. Toxicol Pathol 2022; 50:910-919. [PMID: 36329562 PMCID: PMC9806483 DOI: 10.1177/01926233221131510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Removal of the core fucose from the Fc region of humanized monoclonal antibodies (afucosylated antibodies) enhances their antibody-dependent cell cytotoxicity activities in killing cancer cells. Based on the authors' experience and literature, administrations of afucosylated antibodies have been associated with neutropenia in cynomolgus monkeys. However, in a recent general toxicology study conducted with an afucosylated antibody in cynomolgus monkeys, transient neutropenia was observed and correlated with the emergence of anti-drug antibodies (ADAs) in the affected animals. To further explore the relationship between neutropenia, afucosylated antibodies, and ADAs in cynomolgus monkeys, we performed an investigational retrospective meta-analysis of data from general toxicology studies conducted with Genentech's therapeutic antibodies administered to cynomolgus monkeys between 2005 and 2021. In this analysis, transient neutropenia strongly correlated with ADA-induced inflammation in cynomolgus monkeys administered afucosylated antibodies. This may reflect the simultaneous occurrence of two distinct processes of neutrophil elimination and utilization, thus overwhelming bone marrow reserve capacity leading to transient neutropenia. The integrated analysis of immunogenicity, and anatomic and clinical pathology results from these studies highlights the correlation of transient neutropenia in cynomolgus monkeys with ADA-related inflammation, potentially exacerbated by enhanced effector function of afucosylated antibodies.
Collapse
Affiliation(s)
- Adeyemi O. Adedeji
- Genentech, South San Francisco,
California, USA,Adeyemi O. Adedeji, Safety Assessment,
Genentech (a member of the Roche Group), 1 DNA Way, South San Francisco, CA
94080, USA.
| | - Fiona Zhong
- Genentech, South San Francisco,
California, USA
| | | | - Zoe Zhong
- Genentech, South San Francisco,
California, USA
| | | |
Collapse
|
11
|
Cai H, Kakiuchi-Kiyota S, Hendricks R, Zhong S, Liu L, Adedeji AO, Chan P, Schutten MM, Kamath AV, Ovacik MA. Nonclinical Pharmacokinetics, Pharmacodynamics, and Translational Model of RO7297089, A Novel Anti-BCMA/CD16A Bispecific Tetravalent Antibody for the Treatment of Multiple Myeloma. AAPS J 2022; 24:100. [PMID: 36127472 DOI: 10.1208/s12248-022-00744-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/11/2022] [Indexed: 01/18/2023] Open
Abstract
RO7297089, an anti-B-cell maturation antigen (BCMA)/CD16A bispecific tetravalent antibody, is being developed as a multiple myeloma (MM) therapeutic. This study characterized nonclinical pharmacokinetics (PK), pharmacodynamics (PD), soluble BCMA (sBCMA), and soluble CD16 (sCD16) changes following administration of RO7297089 to support clinical trials. Unbound and total RO7297089 concentrations were measured in cynomolgus monkeys. RO7297089 exhibited a bi-phasic systemic concentration-time profile, similar to a typical human immunoglobulin 1 antibody. Target engagement by RO7297089 led to a robust increase (~100-fold) in total systemic sBCMA levels and relatively mild increase (~2-fold) in total sCD16 levels. To describe the relationship of nonclinical PK/PD data, we developed a target-mediated drug disposition (TMDD) model that includes the systemic target engagement of membrane BCMA (mBCMA), sBCMA, membrane CD16 (mCD16), and sCD16. We then used this model to simulate the PK/PD relationship of RO7297089 in MM patients by translating relevant PK parameters and target levels, based on the literature and newly generated data such as baseline sCD16A levels. Our model suggested that the impact of TMDD on RO7297089 exposure may be more significant in MM patients due to significantly higher expression levels of both mBCMA and sBCMA compared to healthy cynomolgus monkeys. Based on model simulations, we propose more frequent dosing of RO7297089 compared to regular monthly frequency in the clinic at the beginning of treatment to ensure sustained target engagement. This study demonstrates a translational research strategy for collecting relevant nonclinical data, establishing a TMDD model, and using simulations from this model to inform clinical dose regimens.
Collapse
Affiliation(s)
- Hao Cai
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Satoko Kakiuchi-Kiyota
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Robert Hendricks
- BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Shelly Zhong
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Luna Liu
- BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Adeyemi O Adedeji
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Pamela Chan
- Biochemical Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Melissa M Schutten
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Amrita V Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Meric A Ovacik
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA.
| |
Collapse
|
12
|
Bjornson-Hooper ZB, Fragiadakis GK, Spitzer MH, Chen H, Madhireddy D, Hu K, Lundsten K, McIlwain DR, Nolan GP. A Comprehensive Atlas of Immunological Differences Between Humans, Mice, and Non-Human Primates. Front Immunol 2022; 13:867015. [PMID: 35359965 PMCID: PMC8962947 DOI: 10.3389/fimmu.2022.867015] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/16/2022] [Indexed: 01/01/2023] Open
Abstract
Animal models are an integral part of the drug development and evaluation process. However, they are unsurprisingly imperfect reflections of humans, and the extent and nature of many immunological differences are unknown. With the rise of targeted and biological therapeutics, it is increasingly important that we understand the molecular differences in the immunological behavior of humans and model organisms. However, very few antibodies are raised against non-human primate antigens, and databases of cross-reactivity between species are incomplete. Thus, we screened 332 antibodies in five immune cell populations in blood from humans and four non-human primate species generating a comprehensive cross-reactivity catalog that includes cell type-specificity. We used this catalog to create large mass cytometry universal cross-species phenotyping and signaling panels for humans, along with three of the model organisms most similar to humans: rhesus and cynomolgus macaques and African green monkeys; and one of the mammalian models most widely used in drug development: C57BL/6 mice. As a proof-of-principle, we measured immune cell signaling responses across all five species to an array of 15 stimuli using mass cytometry. We found numerous instances of different cellular phenotypes and immune signaling events occurring within and between species, and detailed three examples (double-positive T cell frequency and signaling; granulocyte response to Bacillus anthracis antigen; and B cell subsets). We also explore the correlation of herpes simian B virus serostatus on the immune profile. Antibody panels and the full dataset generated are available online as a resource to enable future studies comparing immune responses across species during the evaluation of therapeutics.
Collapse
Affiliation(s)
| | - Gabriela K. Fragiadakis
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
- Department of Medicine, Division of Rheumatology, University of California San Francisco, San Francisco, CA, United States
- Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA, United States
- University of California, San Francisco (UCSF) Data Science CoLab and University of California, San Francisco (UCSF) Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Matthew H. Spitzer
- Immunology Program, Stanford University, Stanford, CA, United States
- Departments of Otolaryngology – Head and Neck Surgery and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, United States
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| | - Han Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Deepthi Madhireddy
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Kevin Hu
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Kelly Lundsten
- BioLegend Inc, Advanced Cytometry, San Diego, CA, United States
| | - David R. McIlwain
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Garry P. Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| |
Collapse
|
13
|
Pollara J, Tay MZ, Edwards RW, Goodman D, Crowley AR, Edwards RJ, Easterhoff D, Conley HE, Hoxie T, Gurley T, Jones C, Machiele E, Tuyishime M, Donahue E, Jha S, Spreng RL, Hope TJ, Wiehe K, He MM, Moody MA, Saunders KO, Ackerman ME, Ferrari G, Tomaras GD. Functional Homology for Antibody-Dependent Phagocytosis Across Humans and Rhesus Macaques. Front Immunol 2021; 12:678511. [PMID: 34093580 PMCID: PMC8174565 DOI: 10.3389/fimmu.2021.678511] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
Analyses of human clinical HIV-1 vaccine trials and preclinical vaccine studies performed in rhesus macaque (RM) models have identified associations between non-neutralizing Fc Receptor (FcR)-dependent antibody effector functions and reduced risk of infection. Specifically, antibody-dependent phagocytosis (ADP) has emerged as a common correlate of reduced infection risk in multiple RM studies and the human HVTN505 trial. This recurrent finding suggests that antibody responses with the capability to mediate ADP are most likely a desirable component of vaccine responses aimed at protecting against HIV-1 acquisition. As use of RM models is essential for development of the next generation of candidate HIV-1 vaccines, there is a need to determine how effectively ADP activity observed in RMs translates to activity in humans. In this study we compared ADP activity of human and RM monocytes and polymorphonuclear leukocytes (PMN) to bridge this gap in knowledge. We observed considerable variability in the magnitude of monocyte and PMN ADP activity across individual humans and RM that was not dependent on FcR alleles, and only modestly impacted by cell-surface levels of FcRs. Importantly, we found that for both human and RM phagocytes, ADP activity of antibodies targeting the CD4 binding site was greatest when mediated by human IgG3, followed by RM and human IgG1. These results demonstrate that there is functional homology between antibody and FcRs from these two species for ADP. We also used novel RM IgG1 monoclonal antibodies engineered with elongated hinge regions to show that hinge elongation augments RM ADP activity. The RM IgGs with engineered hinge regions can achieve ADP activity comparable to that observed with human IgG3. These novel modified antibodies will have utility in passive immunization studies aimed at defining the role of IgG3 and ADP in protection from virus challenge or control of disease in RM models. Our results contribute to a better translation of human and macaque antibody and FcR biology, and may help to improve testing accuracy and evaluations of future active and passive prevention strategies.
Collapse
Affiliation(s)
- Justin Pollara
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Matthew Zirui Tay
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - R Whitney Edwards
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Derrick Goodman
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Andrew R Crowley
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Robert J Edwards
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - David Easterhoff
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Haleigh E Conley
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Taylor Hoxie
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Thaddeus Gurley
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Caroline Jones
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Emily Machiele
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Marina Tuyishime
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Elizabeth Donahue
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Shalini Jha
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Rachel L Spreng
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Thomas J Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kevin Wiehe
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Max M He
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - M Anthony Moody
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Kevin O Saunders
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | | | - Guido Ferrari
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Georgia D Tomaras
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
14
|
Glucocorticoid-induced eosinopenia results from CXCR4-dependent bone marrow migration. Blood 2021; 136:2667-2678. [PMID: 32659786 DOI: 10.1182/blood.2020005161] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/26/2020] [Indexed: 12/27/2022] Open
Abstract
Glucocorticoids are considered first-line therapy in a variety of eosinophilic disorders. They lead to a transient, profound decrease in circulating human eosinophils within hours of administration. The phenomenon of glucocorticoid-induced eosinopenia has been the basis for the use of glucocorticoids in eosinophilic disorders, and it has intrigued clinicians for 7 decades, yet its mechanism remains unexplained. To investigate, we first studied the response of circulating eosinophils to in vivo glucocorticoid administration in 3 species and found that the response in rhesus macaques, but not in mice, closely resembled that in humans. We then developed an isolation technique to purify rhesus macaque eosinophils from peripheral blood and performed live tracking of zirconium-89-oxine-labeled eosinophils by serial positron emission tomography/computed tomography imaging, before and after administration of glucocorticoids. Glucocorticoids induced rapid bone marrow homing of eosinophils. The kinetics of glucocorticoid-induced eosinopenia and bone marrow migration were consistent with those of the induction of the glucocorticoid-responsive chemokine receptor CXCR4, and selective blockade of CXCR4 reduced or eliminated the early glucocorticoid-induced reduction in blood eosinophils. Our results indicate that glucocorticoid-induced eosinopenia results from CXCR4-dependent migration of eosinophils to the bone marrow. These findings provide insight into the mechanism of action of glucocorticoids in eosinophilic disorders, with implications for the study of glucocorticoid resistance and the development of more targeted therapies. The human study was registered at ClinicalTrials.gov as #NCT02798523.
Collapse
|
15
|
Eosinophil Purification from Peripheral Blood of Rhesus Monkeys. Methods Mol Biol 2021. [PMID: 33486725 DOI: 10.1007/978-1-0716-1095-4_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Eosinophils are granulocytes involved mainly in allergic inflammation and parasitic responses and constitute 1-5% of the circulating leukocytes in human healthy subjects. New immunotherapies targeting eosinophils have been developed and evaluated recently, and the availability of animal models that could mimic human eosinophil responses is important to consider. Differences in eosinophil biology and pathogenesis between humans and murine models have limited their utility in some settings. Isolation of viable eosinophils from rhesus macaque blood suitable for ex vivo and in vitro experimentation could provide a valuable tool for the study of eosinophil-targeted therapies and for the exploration of eosinophilic associated responses. Here, a new technique for the isolation of human eosinophils from rhesus macaque blood by negative selection from whole blood is described.
Collapse
|
16
|
Inhibition of interleukin-6 signaling attenuates aortitis, left ventricular hypertrophy and arthritis in interleukin-1 receptor antagonist deficient mice. Clin Sci (Lond) 2021; 134:2771-2787. [PMID: 33064141 DOI: 10.1042/cs20201036] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/30/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022]
Abstract
The aim of the present study was to examine whether inhibition of Interleukin (IL)-6 signaling by MR16-1, an IL-6 receptor antibody, attenuates aortitis, cardiac hypertrophy, and arthritis in IL-1 receptor antagonist deficient (IL-1RA KO) mice. Four weeks old mice were intraperitoneally administered with either MR16-1 or non-immune IgG at dosages that were adjusted over time for 5 weeks. These mice were stratified into four groups: MR16-1 treatment groups, KO/MR low group (first 2.0 mg, following 0.5 mg/week, n=14) and KO/MR high group (first 4.0 mg, following 2.0 mg/week, n=19) in IL-1RA KO mice, and IgG treatment groups, KO/IgG group (first 2.0 mg, following 1.0 mg/week, n=22) in IL-1RA KO mice, and wild/IgG group (first 2.0 mg, following 1.0 mg/week, n=17) in wild mice. Aortitis, cardiac hypertrophy and arthropathy were histologically analyzed. Sixty-eight percent of the KO/IgG group developed aortitis (53% developed severe aortitis). In contrast, only 21% of the KO/MR high group developed mild aortitis, without severe aortitis (P<0.01, vs KO/IgG group). Infiltration of inflammatory cells, such as neutrophils, T cells, and macrophages, was frequently observed around aortic sinus of the KO/IgG group. Left ventricle and cardiomyocyte hypertrophy were observed in IL-1RA KO mice. Administration of high dosage of MR16-1 significantly suppressed cardiomyocyte hypertrophy. MR16-1 attenuated the incidence and severity of arthritis in IL-1RA KO mice in a dose-dependent manner. In conclusion, blockade of IL-6 signaling may exert a beneficial effect to attenuate severe aortitis, left ventricle hypertrophy, and arthritis.
Collapse
|
17
|
Grunst MW, Grandea AG, Janaka SK, Hammad I, Grimes P, Karl JA, Wiseman R, O'Connor DH, Evans DT. Functional Interactions of Common Allotypes of Rhesus Macaque FcγR2A and FcγR3A with Human and Macaque IgG Subclasses. THE JOURNAL OF IMMUNOLOGY 2020; 205:3319-3332. [PMID: 33208458 DOI: 10.4049/jimmunol.2000501] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/17/2020] [Indexed: 12/18/2022]
Abstract
The rhesus macaque is an important animal model for AIDS and other infectious diseases. However, the investigation of Fc-mediated Ab responses in macaques is complicated by species-specific differences in FcγRs and IgG subclasses relative to humans. To assess the effects of these differences on FcγR-IgG interactions, reporter cell lines expressing common allotypes of human and rhesus macaque FcγR2A and FcγR3A were established. FcγR-mediated responses to B cells were measured in the presence of serial dilutions of anti-CD20 Abs with Fc domains corresponding to each of the four subclasses of human and rhesus IgG and with Fc variants of IgG1 that enhance binding to FcγR2A or FcγR3A. All of the FcγRs were functional and preferentially recognized either IgG1 or IgG2. Whereas allotypes of rhesus FcγR2A were identified with responses similar to variants of human FcγR2A with higher (H131) and lower (R131) affinity for IgG, all of the rhesus FcγR3A allotypes exhibited responses most similar to the higher affinity V158 variant of human FcγR3A. Unlike responses to human IgGs, there was little variation in FcγR-mediated responses to different subclasses of rhesus IgG. Phylogenetic comparisons suggest that this reflects limited sequence variation of macaque IgGs as a result of their relatively recent diversification from a common IGHG gene since humans and macaques last shared a common ancestor. These findings reveal species-specific differences in FcγR-IgG interactions with important implications for investigating Ab effector functions in macaques.
Collapse
Affiliation(s)
- Michael W Grunst
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705; and
| | - Andres G Grandea
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705; and.,Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715
| | - Sanath Kumar Janaka
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705; and
| | - Iman Hammad
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705; and
| | - Parker Grimes
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705; and
| | - Julie A Karl
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715
| | - Roger Wiseman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705; and.,Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715
| | - David T Evans
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705; and .,Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715
| |
Collapse
|
18
|
Nagelkerke SQ, Schmidt DE, de Haas M, Kuijpers TW. Genetic Variation in Low-To-Medium-Affinity Fcγ Receptors: Functional Consequences, Disease Associations, and Opportunities for Personalized Medicine. Front Immunol 2019; 10:2237. [PMID: 31632391 PMCID: PMC6786274 DOI: 10.3389/fimmu.2019.02237] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/04/2019] [Indexed: 12/23/2022] Open
Abstract
Fc-gamma receptors (FcγR) are the cellular receptors for Immunoglobulin G (IgG). Upon binding of complexed IgG, FcγRs can trigger various cellular immune effector functions, thereby linking the adaptive and innate immune systems. In humans, six classic FcγRs are known: one high-affinity receptor (FcγRI) and five low-to-medium-affinity FcγRs (FcγRIIA, -B and -C, FcγRIIIA and -B). In this review we describe the five genes encoding the low-to-medium -affinity FcγRs (FCGR2A, FCGR2B, FCGR2C, FCGR3A, and FCGR3B), including well-characterized functionally relevant single nucleotide polymorphisms (SNPs), haplotypes as well as copy number variants (CNVs), which occur in distinct copy number regions across the locus. The evolution of the locus is also discussed. Importantly, we recommend a consistent nomenclature of genetic variants in the FCGR2/3 locus. Next, we focus on the relevance of genetic variation in the FCGR2/3 locus in auto-immune and auto-inflammatory diseases, highlighting pathophysiological insights that are informed by genetic association studies. Finally, we illustrate how specific FcγR variants relate to variation in treatment responses and prognosis amongst autoimmune diseases, cancer and transplant immunology, suggesting novel opportunities for personalized medicine.
Collapse
Affiliation(s)
- Sietse Q Nagelkerke
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - David E Schmidt
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Masja de Haas
- Sanquin Diagnostic Services, Department of Immunohematology Diagnostics, Amsterdam, Netherlands.,Sanquin Research, Center for Clinical Transfusion Research, Leiden, Netherlands.,Jon J. van Rood Center for Clinical Transfusion Science, Leiden University Medical Center, Leiden, Netherlands.,Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Taco W Kuijpers
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
19
|
Lejeune J, Brachet G, Watier H. Evolutionary Story of the Low/Medium-Affinity IgG Fc Receptor Gene Cluster. Front Immunol 2019; 10:1297. [PMID: 31244843 PMCID: PMC6563257 DOI: 10.3389/fimmu.2019.01297] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/21/2019] [Indexed: 11/28/2022] Open
Abstract
Low/intermediate affinity Fc-gamma receptors (FcγR) are crucial for the recognition of immune complexes and IgG-sensitized microorganisms by phagocytic and cytotoxic effector cells. In all mammalian species studied so far, their genes are clustered in a single locus. However, this locus differs between humans and mice, both in the number of genes and the structure/function of the encoded receptors. We show that murine fcgr3 evolved through several steps into FCGR2A, its ortholog, which is specific to primates. One of these steps was the insertion of a retroviral element bringing a new intracellular exon comprising a non-canonical ITAM motif. We also show that the fcgr3-hspa6-fcgr4-fcgr2b module in mammals that has evolved in a FCGR2A-HSPA6-FCGR4-FCGR2B module in primates, was subsequently duplicated in apes through a Non-Allelic Homologous Recombination (NAHR), giving birth to FCGR2C, a hybrid gene between FCGR2B and FCGR2A. The FCGR4 duplication, which occurred simultaneously, eventually resulted in the emergence of FCGR3B, while FCGR3A remained the true FCGR4 ortholog. FCGR2C and FCGR3B, markers of this NAHR, are present in gorillas and chimpanzees, whereas they are absent in orangutans and more distant primates, such as gibbons and macaques. These data need to be taken into account when testing IgG-based therapies in animal species.
Collapse
Affiliation(s)
| | - Guillaume Brachet
- EA 7501 GICC Université de Tours, Tours, France.,CHRU de Tours, Tours, France
| | - Hervé Watier
- EA 7501 GICC Université de Tours, Tours, France.,CHRU de Tours, Tours, France
| |
Collapse
|
20
|
Crowley AR, Ackerman ME. Mind the Gap: How Interspecies Variability in IgG and Its Receptors May Complicate Comparisons of Human and Non-human Primate Effector Function. Front Immunol 2019; 10:697. [PMID: 31024542 PMCID: PMC6463756 DOI: 10.3389/fimmu.2019.00697] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/13/2019] [Indexed: 01/08/2023] Open
Abstract
The field of HIV research relies heavily on non-human primates, particularly the members of the macaque genus, as models for the evaluation of candidate vaccines and monoclonal antibodies. A growing body of research suggests that successful protection of humans will not solely rely on the neutralization activity of an antibody's antigen binding fragment. Rather, immunological effector functions prompted by the interaction of the immunoglobulin G constant region and its cognate Fc receptors help contribute to favorable outcomes. Inherent differences in the sequences, expression, and activities of human and non-human primate antibody receptors and immunoglobulins have the potential to produce disparate results in the observations made in studies conducted in differing species. Having a more complete understanding of these differences, however, should permit the more fluent translation of observations between model organisms and the clinic. Here we present a guide to such translations that encompasses not only what is presently known regarding the affinity of the receptor-ligand interactions but also the influence of expression patterns and allelic variation, with a focus on insights gained from use of this model in HIV vaccines and passive antibody therapy and treatment.
Collapse
Affiliation(s)
- Andrew R. Crowley
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH, United States
| | - Margaret E. Ackerman
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
21
|
Tolbert WD, Subedi GP, Gohain N, Lewis GK, Patel KR, Barb AW, Pazgier M. From Rhesus macaque to human: structural evolutionary pathways for immunoglobulin G subclasses. MAbs 2019; 11:709-724. [PMID: 30939981 PMCID: PMC6601566 DOI: 10.1080/19420862.2019.1589852] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/15/2019] [Accepted: 02/22/2019] [Indexed: 10/27/2022] Open
Abstract
The Old World monkey, Rhesus macaque (Macaca mulatta, Mm), is frequently used as a primate model organism in the study of human disease and to test new vaccines/antibody treatments despite diverging before chimpanzees and orangutans. Mm and humans share 93% genome identity with substantial differences in the genes of the adaptive immune system that lead to different functional IgG subclass characteristics, Fcγ receptors expressed on innate immune cells, and biological interactions. These differences put limitations on Mm use as a primary animal model in the study of human disease and to test new vaccines/antibody treatments. Here, we comprehensively analyzed molecular properties of the Fc domain of the four IgG subclasses of Rhesus macaque to describe potential mechanisms for their interactions with effector cell Fc receptors. Our studies revealed less diversity in the overall structure among the Mm IgG Fc, with MmIgG1 Fc being the most structurally like human IgG3, although its CH2 loops and N297 glycan mobility are comparable to human IgG1. Furthermore, the Fcs of Mm IgG3 and 4 lack the structural properties typical for their human orthologues that determine IgG3's reduced interaction with the neonatal receptor and IgG4's ability for Fab-arm exchange and its weaker Fcγ receptor interactions. Taken together, our data indicate that MmIgG1-4 are less structurally divergent than the human IgGs, with only MmIgG1 matching the molecular properties of human IgG1 and 3, the most active IgGs in terms of Fcγ receptor binding and Fc-mediated functions. PDB accession numbers for deposited structures are 6D4E, 6D4I, 6D4M, and 6D4N for MmIgG1 Fc, MmIgG2 Fc, MmIgG3 Fc, and MmIgG4 Fc, respectively.
Collapse
Affiliation(s)
- William David Tolbert
- Division of Vaccine Research, Institute of Human Virology of University of Maryland School of Medicine, Baltimore, MD, USA
- Infectious Disease Division, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Ganesh Prasad Subedi
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology of Iowa State University, Ames, IA, USA
| | - Neelakshi Gohain
- Division of Vaccine Research, Institute of Human Virology of University of Maryland School of Medicine, Baltimore, MD, USA
| | - George Kenneth Lewis
- Division of Vaccine Research, Institute of Human Virology of University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kashyap Rajesh Patel
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology of Iowa State University, Ames, IA, USA
| | - Adam Wesley Barb
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology of Iowa State University, Ames, IA, USA
| | - Marzena Pazgier
- Division of Vaccine Research, Institute of Human Virology of University of Maryland School of Medicine, Baltimore, MD, USA
- Infectious Disease Division, Uniformed Services University of the Health Sciences, Bethesda, MD
| |
Collapse
|
22
|
Kolb P, Sijmons S, McArdle MR, Taher H, Womack J, Hughes C, Ventura A, Jarvis MA, Stahl-Hennig C, Hansen S, Picker LJ, Malouli D, Hengel H, Früh K. Identification and Functional Characterization of a Novel Fc Gamma-Binding Glycoprotein in Rhesus Cytomegalovirus. J Virol 2019; 93:e02077-18. [PMID: 30487278 PMCID: PMC6364020 DOI: 10.1128/jvi.02077-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/20/2022] Open
Abstract
Receptors recognizing the Fc part of immunoglobulin G (FcγRs) are key determinants in antibody-mediated immune responses. Members of the Herpesviridae interfere with this immune regulatory network by expressing viral FcγRs (vFcγRs). Human cytomegalovirus (HCMV) encodes four distinct vFcγRs that differ with respect to their IgG subtype specificity and their impact on antibody-mediated immune function in vitro The impact of vFcγRs on HCMV pathogenesis and immunomodulation in vivo is not known. The closest evolutionary animal model of HCMV is rhesus CMV (RhCMV) infection of rhesus macaques. To enable the characterization of vFcγR function in this model, we studied IgG binding by RhCMV. We show that lysates of RhCMV-infected cells contain an IgG-binding protein of 30 kDa encoded by the gene Rh05 that is a predicted type I glycoprotein belonging to the RL11 gene family. Upon deletion of Rh05, IgG-Fc binding by RhCMV strain 68-1 is lost, whereas ectopic expression of Rh05 results in IgG binding to transfected cells consistent with Rh05 being a vFcγR. Using a set of reporter cell lines stably expressing human and rhesus FcγRs, we further demonstrate that Rh05 antagonizes host FcγR activation. Compared to Rh05-intact RhCMV, RhCMVΔRh05 showed an increased activation of host FcγR upon exposure of infected cells to IgG from RhCMV-seropositive animals, suggesting that Rh05 protects infected cells from opsonization and IgG-dependent activation of host FcγRs. However, antagonizing host FcγR activation by Rh05 was not required for the establishment and maintenance of infection of RhCMV, even in a seropositive host, as shown by the induction of T cell responses to heterologous antigens expressed by RhCMV lacking the gene region encoding Rh05. In contrast to viral evasion of natural killer cells or T cell recognition, the evasion of antibody-mediated effects does not seem to be absolutely required for infection or reinfection. The identification of the first vFcγR that efficiently antagonizes host FcγR activation in the RhCMV genome will thus permit more detailed studies of this immunomodulatory mechanism in promoting viral dissemination in the presence of natural or vaccine-induced humoral immunity.IMPORTANCE Rhesus cytomegalovirus (RhCMV) offers a unique model for studying human cytomegalovirus (HCMV) pathogenesis and vaccine development. RhCMV infection of nonhuman primates greatly broadened the understanding of mechanisms by which CMVs evade or reprogram T cell and natural killer cell responses in vivo However, the role of humoral immunity and viral modulation of anti-CMV antibodies has not been studied in this model. There is evidence from in vitro studies that HCMVs can evade humoral immunity. By gene mapping and with the help of a novel cell-based reporter assay system we characterized the first RhCMV encoded IgG-Fcγ binding glycoprotein as a potent antagonist of rhesus FcγR activation. We further demonstrate that, unlike evasion of T cell immunity, this viral Fcγ receptor is not required to overcome anti-CMV immunity to establish secondary infections. These findings enable more detailed studies of the in vivo consequences of CMV evasion from IgG responses in nonhuman primate models.
Collapse
Affiliation(s)
- Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Steven Sijmons
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Matthew R McArdle
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Jennie Womack
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Colette Hughes
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Abigail Ventura
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Michael A Jarvis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Scott Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| |
Collapse
|
23
|
Lucas AT, Robinson R, Schorzman AN, Piscitelli JA, Razo JF, Zamboni WC. Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients. Antibodies (Basel) 2019; 8:E3. [PMID: 31544809 PMCID: PMC6640706 DOI: 10.3390/antib8010003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 12/11/2022] Open
Abstract
The rapid advancement in the development of therapeutic proteins, including monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), has created a novel mechanism to selectively deliver highly potent cytotoxic agents in the treatment of cancer. These agents provide numerous benefits compared to traditional small molecule drugs, though their clinical use still requires optimization. The pharmacology of mAbs/ADCs is complex and because ADCs are comprised of multiple components, individual agent characteristics and patient variables can affect their disposition. To further improve the clinical use and rational development of these agents, it is imperative to comprehend the complex mechanisms employed by antibody-based agents in traversing numerous biological barriers and how agent/patient factors affect tumor delivery, toxicities, efficacy, and ultimately, biodistribution. This review provides an updated summary of factors known to affect the disposition of mAbs/ADCs in development and in clinical use, as well as how these factors should be considered in the selection and design of preclinical studies of ADC agents in development.
Collapse
Affiliation(s)
- Andrew T Lucas
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Ryan Robinson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Joseph A Piscitelli
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - Juan F Razo
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
24
|
Weisgrau KL, Vosler LJ, Pomplun NL, Hayes JM, Simmons HA, Friedrichs KR, Rakasz EG. Neutrophil progenitor populations of rhesus macaques. J Leukoc Biol 2018; 105:113-121. [PMID: 30395351 DOI: 10.1002/jlb.1ta1117-431rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 09/24/2018] [Accepted: 10/13/2018] [Indexed: 01/06/2023] Open
Abstract
Captive-bred rhesus macaques of Indian origin represent one of the most important large animal models for infectious disease, solid organ transplantation, and stem cell research. There is a dearth of information defining hematopoietic development, including neutrophil leukocyte differentiation in this species using multicolor flow cytometry. In the current study, we sought to identify cell surface markers that delineate neutrophil progenitor populations with characteristic immunophenotypes. We defined four different postmitotic populations based on their CD11b and CD87 expression pattern, and further refined their immunophenotypes using CD32, CD64, lactoferrin, and myeloperoxidase as antigenic markers. The four subsets contained myelocyte, metamyelocyte, band, and segmented neutrophil populations. We compared our flow cytometry-based classification with the classical nuclear morphology-based classification. We found overlap of immunological phenotype between populations of different nuclear morphology and identified phenotypically different subsets within populations of similar nuclear morphology. We assessed the responsiveness of these populations to stimulatory signals, such as LPS, fMLP, or PMA, and demonstrated significant differences between human and rhesus macaque neutrophil progenitors. In this study, we provided evidence for species-specific features of granulopoiesis that ultimately manifested in the divergent immunophenotypes of the fully differentiated segmented neutrophils of humans and rhesus macaques. Additionally, we found functional markers that can be used to accurately quantify neutrophil progenitors by flow cytometry. Although these markers do not coincide with the classical nuclear-morphology-based grading, they enable us to perform functional studies monitoring immunophenotypic markers.
Collapse
Affiliation(s)
- Kim L Weisgrau
- Immunology Services Unit, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Logan J Vosler
- Immunology Services Unit, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nicholas L Pomplun
- Immunology Services Unit, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jennifer M Hayes
- Clinical Pathology Unit, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Heather A Simmons
- Clinical Pathology Unit, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kristen R Friedrichs
- Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Eva G Rakasz
- Immunology Services Unit, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
25
|
Engineered hexavalent Fc proteins with enhanced Fc-gamma receptor avidity provide insights into immune-complex interactions. Commun Biol 2018; 1:146. [PMID: 30272022 PMCID: PMC6138732 DOI: 10.1038/s42003-018-0149-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 08/22/2018] [Indexed: 12/14/2022] Open
Abstract
Autoantibody-mediated diseases are currently treated with intravenous immunoglobulin, which is thought to act in part via blockade of Fc gamma receptors, thereby inhibiting autoantibody effector functions and subsequent pathology. We aimed to develop recombinant molecules with enhanced Fc receptor avidity and thus increased potency over intravenous immunoglobulin. Here we describe the molecular engineering of human Fc hexamers and explore their therapeutic and safety profiles. We show Fc hexamers were more potent than IVIG in phagocytosis blockade and disease models. However, in human whole-blood safety assays incubation with IgG1 isotype Fc hexamers resulted in cytokine release, platelet and complement activation, whereas the IgG4 version did not. We used a statistically designed mutagenesis approach to identify the key Fc residues involved in these processes. Cytokine release was found to be dependent on neutrophil FcγRIIIb interactions with L234 and A327 in the Fc. Therefore, Fc hexamers provide unique insights into Fc receptor biology. Tania Rowley et al. present multivalent Fc molecules with enhanced avidity for Fc gamma receptors in order to improve the treatment of autoantibody-mediated human diseases. They found several key amino acids involved in Fc receptor binding interactions.
Collapse
|
26
|
Hensley-McBain T, Berard AR, Manuzak JA, Miller CJ, Zevin AS, Polacino P, Gile J, Agricola B, Cameron M, Hu SL, Estes JD, Reeves RK, Smedley J, Keele BF, Burgener AD, Klatt NR. Intestinal damage precedes mucosal immune dysfunction in SIV infection. Mucosal Immunol 2018; 11:1429-1440. [PMID: 29907866 PMCID: PMC6162106 DOI: 10.1038/s41385-018-0032-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/23/2018] [Accepted: 04/02/2018] [Indexed: 02/04/2023]
Abstract
HIV and pathogenic SIV infection are characterized by mucosal dysfunction including epithelial barrier damage, loss of Th17 cells, neutrophil infiltration, and microbial translocation with accompanying inflammation. However, it is unclear how and when these contributing factors occur relative to one another. In order to determine whether any of these features initiates the cycle of damage, we longitudinally evaluated the kinetics of mucosal and systemic T-cell activation, microbial translocation, and Th17 cell and neutrophil frequencies following intrarectal SIV infection of rhesus macaques. We additionally assessed the colon proteome to elucidate molecular pathways altered early after infection. We demonstrate increased T-cell activation (HLA-DR+) beginning 3-14 days post-SIV challenge, reduced peripheral zonulin 3-14 days post-SIV, and evidence of microbial translocation 14 days post-SIV. The onset of mucosal dysfunction preceded peripheral and mucosal Th17 depletion, which occurred 14-28 days post-SIV, and gut neutrophil accumulation was not observed. Proteins involved in epithelial structure were downregulated 3 days post-SIV followed by an upregulation of immune proteins 14 days post-SIV. These data demonstrate that immune perturbations such as Th17 loss and neutrophil infiltration occur after alterations to epithelial structural protein pathways, suggesting that epithelial damage occurs prior to widespread immune dysfunction.
Collapse
Affiliation(s)
- Tiffany Hensley-McBain
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Alicia R Berard
- National HIV and Retrovirology Labs, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Jennifer A Manuzak
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Charlene J Miller
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Alexander S Zevin
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | | | - Jillian Gile
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Brian Agricola
- Washington National Primate Research Center, Seattle, WA, USA
| | - Mark Cameron
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Shiu-Lok Hu
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Jacob D Estes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - R Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Jeremy Smedley
- Washington National Primate Research Center, Seattle, WA, USA
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Adam D Burgener
- National HIV and Retrovirology Labs, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nichole R Klatt
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA.
- Washington National Primate Research Center, Seattle, WA, USA.
| |
Collapse
|
27
|
Oshima T, Miyashita H, Ishimura Y, Ito Y, Tanaka Y, Hori A, Kokubo T, Kurokawa T. Fc engineering of anti-Nectin-2 antibody improved thrombocytopenic adverse event in monkey. PLoS One 2018; 13:e0196422. [PMID: 29723247 PMCID: PMC5933732 DOI: 10.1371/journal.pone.0196422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 04/12/2018] [Indexed: 11/18/2022] Open
Abstract
Nectin-2 is a transmembrane glycoprotein which is involved in the process of Ca2+-independent cell-cell adhesion. In our previous study, we have demonstrated that Nectin-2 is over-expressed in breast and ovarian cancer tissues by using gene expression analysis and immunohistochemistry. Furthermore, we discovered multiple anti-Nectin-2 fully human monoclonal antibodies which inhibited tumor growth in in vivo subcutaneous xenograft models with antibody-dependent cellular cytotoxicity (ADCC) as the principal mechanism of action. In this report, we assessed the toxicity of Y-443, a fully human IgG1/kappa anti-Nectin-2 monoclonal antibody exhibiting strong in vitro ADCC and in vivo anti-tumor activity in cynomolgus monkeys (Macaca fascicularis (Cynos)). Unexpectedly, upon administration, Y-443 induced strong thrombocytopenia through Nectin-2 expressed on Cyno platelets, presumably followed by phagocytosis in the mononuclear phagocytic system. To mitigate the adverse safety profile, we mutated the Fc region of Y-443 to reduce the Fc binding activity to Fcγ receptor I, which is the primary receptor for phagocytosis on macrophages. Moreover, we further engineered the Fc through defucosylation to maintain ADCC activity. The resultant Fc engineered antibody, termed Y-634, demonstrated diminished thrombocytopenia in Cyno toxicological studies and maintained anti-tumor activity in a mouse xenograft model. These findings suggest that Y-634 may have a therapeutic potential for the treatment of Nectin-2 positive cancers, and moreover, Fc engineering is a potential mitigation strategy to ameliorate safety liabilities in antibody induced thrombocytopenia while maintaining antibody potency.
Collapse
Affiliation(s)
- Tsutomu Oshima
- Immunobiologics, Takeda California Inc., San Diego, California, United States of America
- * E-mail:
| | - Hideaki Miyashita
- Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, Hikari, Yamaguchi, Japan
| | - Yoshimasa Ishimura
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Yuki Ito
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Yoko Tanaka
- Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, Hikari, Yamaguchi, Japan
| | - Akira Hori
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Toshio Kokubo
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Tomofumi Kurokawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| |
Collapse
|
28
|
Human IgG subclass cross-species reactivity to mouse and cynomolgus monkey Fcγ receptors. Immunol Lett 2018; 197:1-8. [DOI: 10.1016/j.imlet.2018.02.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/23/2018] [Accepted: 02/15/2018] [Indexed: 01/11/2023]
|
29
|
Wines BD, Billings H, Mclean MR, Kent SJ, Hogarth PM. Antibody Functional Assays as Measures of Fc Receptor-Mediated Immunity to HIV - New Technologies and their Impact on the HIV Vaccine Field. Curr HIV Res 2018; 15:202-215. [PMID: 28322167 PMCID: PMC5543561 DOI: 10.2174/1570162x15666170320112247] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/20/2017] [Accepted: 03/09/2017] [Indexed: 12/23/2022]
Abstract
Background: There is now intense interest in the role of HIV-specific antibodies and the engagement of FcγR functions in the control and prevention of HIV infection. The analyses of the RV144 vaccine trial, natural progression cohorts, and macaque models all point to a role for Fc-dependent effector functions, such as cytotoxicity (ADCC) or phagocytosis (ADCP), in the control of HIV. However, reliable assays that can be reproducibly used across different laboratories to measure Fc-dependent functions, such as antibody dependent cellular cytotoxicity (ADCC) are limited. Method: This brief review highlights the importance of Fc properties for immunity to HIV, particular-ly via FcγR diversity and function. We discuss assays used to study FcR mediated functions of HIV-specific Ab, including our recently developed novel cell-free ELISA using homo-dimeric FcγR ecto-domains to detect functionally relevant viral antigen-specific antibodies. Results: The binding of these dimeric FcγR ectodomains, to closely spaced pairs of IgG Fc, mimics the engagement and cross-linking of Fc receptors by IgG opsonized virions or infected cells as the es-sential prerequisite to the induction of Ab-dependent effector functions. The dimeric FcγR ELISA reli-ably correlates with ADCC in patient responses to influenza. The assay is amenable to high throughput and could be standardized across laboratories. Conclusion: We propose the assay has broader implications for the evaluation of the quality of anti-body responses in viral infections and for the rapid evaluation of responses in vaccine development campaigns for HIV and other viral infections.
Collapse
Affiliation(s)
- Bruce D Wines
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia.,Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia.,Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
| | - Hugh Billings
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia
| | - Milla R Mclean
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Parkville, Victoria, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Parkville, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Parkville, Victoria, Australia.,Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Victoria, Australia
| | - P Mark Hogarth
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia.,Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia.,Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
30
|
Lucas AT, Price LSL, Schorzman AN, Storrie M, Piscitelli JA, Razo J, Zamboni WC. Factors Affecting the Pharmacology of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:E10. [PMID: 31544862 PMCID: PMC6698819 DOI: 10.3390/antib7010010] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Major advances in therapeutic proteins, including antibody-drug conjugates (ADCs), have created revolutionary drug delivery systems in cancer over the past decade. While these immunoconjugate agents provide several advantages compared to their small-molecule counterparts, their clinical use is still in its infancy. The considerations in their development and clinical use are complex, and consist of multiple components and variables that can affect the pharmacologic characteristics. It is critical to understand the mechanisms employed by ADCs in navigating biological barriers and how these factors affect their biodistribution, delivery to tumors, efficacy, and toxicity. Thus, future studies are warranted to better understand the complex pharmacology and interaction between ADC carriers and biological systems, such as the mononuclear phagocyte system (MPS) and tumor microenvironment. This review provides an overview of factors that affect the pharmacologic profiles of ADC therapies that are currently in clinical use and development.
Collapse
Affiliation(s)
- Andrew T Lucas
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Lauren S L Price
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Mallory Storrie
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | | | - Juan Razo
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
31
|
Unravelling Immunoglobulin G Fc N-Glycosylation: A Dynamic Marker Potentiating Predictive, Preventive and Personalised Medicine. Int J Mol Sci 2018; 19:ijms19020390. [PMID: 29382131 PMCID: PMC5855612 DOI: 10.3390/ijms19020390] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/10/2018] [Accepted: 01/24/2018] [Indexed: 11/21/2022] Open
Abstract
Multiple factors influence immunoglobulin G glycosylation, which in turn affect the glycoproteins’ function on eliciting an anti-inflammatory or pro-inflammatory response. It is prudent to underscore these processes when considering the use of immunoglobulin G N-glycan moieties as an indication of disease presence, progress, or response to therapeutics. It has been demonstrated that the altered expression of genes that encode enzymes involved in the biosynthesis of immunoglobulin G N-glycans, receptors, or complement factors may significantly modify immunoglobulin G effector response, which is important for regulating the immune system. The immunoglobulin G N-glycome is highly heterogenous; however, it is considered an interphenotype of disease (a link between genetic predisposition and environmental exposure) and so has the potential to be used as a dynamic biomarker from the perspective of predictive, preventive, and personalised medicine. Undoubtedly, a deeper understanding of how the multiple factors interact with each other to alter immunoglobulin G glycosylation is crucial. Herein we review the current literature on immunoglobulin G glycoprotein structure, immunoglobulin G Fc glycosylation, associated receptors, and complement factors, the downstream effector functions, and the factors associated with the heterogeneity of immunoglobulin G glycosylation.
Collapse
|
32
|
Elhmouzi-Younes J, Palgen JL, Tchitchek N, Delandre S, Namet I, Bodinham CL, Pizzoferro K, Lewis DJ, Le Grand R, Cosma A, Beignon AS. In depth comparative phenotyping of blood innate myeloid leukocytes from healthy humans and macaques using mass cytometry. Cytometry A 2017; 91:969-982. [DOI: 10.1002/cyto.a.23107] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/04/2017] [Accepted: 03/15/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Jamila Elhmouzi-Younes
- Immunology of viral infections and autoimmune diseases; CEA - Université Paris Sud 11 - INSERM U1184, 92265 Fontenay-aux-Roses; France
| | - Jean-Louis Palgen
- Immunology of viral infections and autoimmune diseases; CEA - Université Paris Sud 11 - INSERM U1184, 92265 Fontenay-aux-Roses; France
| | - Nicolas Tchitchek
- Immunology of viral infections and autoimmune diseases; CEA - Université Paris Sud 11 - INSERM U1184, 92265 Fontenay-aux-Roses; France
| | - Simon Delandre
- Immunology of viral infections and autoimmune diseases; CEA - Université Paris Sud 11 - INSERM U1184, 92265 Fontenay-aux-Roses; France
| | - Inana Namet
- Immunology of viral infections and autoimmune diseases; CEA - Université Paris Sud 11 - INSERM U1184, 92265 Fontenay-aux-Roses; France
| | | | | | - David J.M. Lewis
- Surrey Clinical Research Centre; University of Surrey; Guildford GU2 7XP UK
| | - Roger Le Grand
- Immunology of viral infections and autoimmune diseases; CEA - Université Paris Sud 11 - INSERM U1184, 92265 Fontenay-aux-Roses; France
| | - Antonio Cosma
- Immunology of viral infections and autoimmune diseases; CEA - Université Paris Sud 11 - INSERM U1184, 92265 Fontenay-aux-Roses; France
| | - Anne-Sophie Beignon
- Immunology of viral infections and autoimmune diseases; CEA - Université Paris Sud 11 - INSERM U1184, 92265 Fontenay-aux-Roses; France
| |
Collapse
|
33
|
Boesch AW, Miles AR, Chan YN, Osei-Owusu NY, Ackerman ME. IgG Fc variant cross-reactivity between human and rhesus macaque FcγRs. MAbs 2017; 9:455-465. [PMID: 28055295 DOI: 10.1080/19420862.2016.1274845] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Non-human primate (NHP) studies are often an essential component of antibody development efforts before human trials. Because the efficacy or toxicity of candidate antibodies may depend on their interactions with Fcγ receptors (FcγR) and their resulting ability to induce FcγR-mediated effector functions such as antibody-dependent cell-meditated cytotoxicity and phagocytosis (ADCP), the evaluation of human IgG variants with modulated affinity toward human FcγR is becoming more prevalent in both infectious disease and oncology studies in NHP. Reliable translation of these results necessitates analysis of the cross-reactivity of these human Fc variants with NHP FcγR. We report evaluation of the binding affinities of a panel of human IgG subclasses, Fc amino acid point mutants and Fc glycosylation variants against the common allotypes of human and rhesus macaque FcγR by applying a high-throughput array-based surface plasmon resonance platform. The resulting data indicate that amino acid variation present in rhesus FcγRs can result in disrupted, matched, or even increased affinity of IgG Fc variants compared with human FcγR orthologs. These observations emphasize the importance of evaluating species cross-reactivity and developing an understanding of the potential limitations or suitability of representative in vitro and in vivo models before human clinical studies when either efficacy or toxicity may be associated with FcγR engagement.
Collapse
Affiliation(s)
- Austin W Boesch
- a Thayer School of Engineering, Dartmouth College , Hanover , NH , USA
| | - Adam R Miles
- b Wasatch Microfluidics , Salt Lake City , UT , USA
| | - Ying N Chan
- a Thayer School of Engineering, Dartmouth College , Hanover , NH , USA
| | - Nana Y Osei-Owusu
- c Department of Microbiology and Immunology , Geisel School of Medicine , Lebanon , NH , USA
| | - Margaret E Ackerman
- a Thayer School of Engineering, Dartmouth College , Hanover , NH , USA.,c Department of Microbiology and Immunology , Geisel School of Medicine , Lebanon , NH , USA
| |
Collapse
|
34
|
Brennan FR, Kiessling A. Translational immunotoxicology of immunomodulatory monoclonal antibodies. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:85-93. [PMID: 27978992 DOI: 10.1016/j.ddtec.2016.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 12/28/2022]
Abstract
While immunomodulatory monoclonal antibodies (mAbs) have a wide therapeutic potential, exaggerated immunopharmacology may drive both acute and delayed immunotoxicity. The existing tools for immunotoxicity assessment do not accurately predict the full range of immunotoxicities observed in humans. New and optimized models, assays, endpoints and biomarkers in animals and humans are required to safeguard patients and allow them access to these often transformational therapies.
Collapse
|
35
|
Chan YN, Boesch AW, Osei-Owusu NY, Emileh A, Crowley AR, Cocklin SL, Finstad SL, Linde CH, Howell RA, Zentner I, Cocklin S, Miles AR, Eckman JW, Alter G, Schmitz JE, Ackerman ME. IgG Binding Characteristics of Rhesus Macaque FcγR. THE JOURNAL OF IMMUNOLOGY 2016; 197:2936-47. [PMID: 27559046 DOI: 10.4049/jimmunol.1502252] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 07/26/2016] [Indexed: 11/19/2022]
Abstract
Indian rhesus macaques (Macaca mulatta) are routinely used in preclinical studies to evaluate therapeutic Abs and candidate vaccines. The efficacy of these interventions in many cases is known to rely heavily on the ability of Abs to interact with a set of Ab FcγR expressed on innate immune cells. Yet, despite their presumed functional importance, M. mulatta Ab receptors are largely uncharacterized, posing a fundamental limit to ensuring accurate interpretation and translation of results from studies in this model. In this article, we describe the binding characteristics of the most prevalent allotypic variants of M. mulatta FcγR for binding to both human and M. mulatta IgG of varying subclasses. The resulting determination of the affinity, specificity, and glycan sensitivity of these receptors promises to be useful in designing and evaluating studies of candidate vaccines and therapeutic Abs in this key animal model and exposes significant evolutionary divergence between humans and macaques.
Collapse
Affiliation(s)
- Ying N Chan
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755
| | - Austin W Boesch
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755
| | - Nana Y Osei-Owusu
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH 03755
| | - Ali Emileh
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755
| | - Andrew R Crowley
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH 03755
| | - Sarah L Cocklin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Samantha L Finstad
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Caitlyn H Linde
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Rebecca A Howell
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Isaac Zentner
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Simon Cocklin
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Adam R Miles
- Wasatch Microfluidics, Salt Lake City, UT 84103; and
| | | | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - Joern E Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755; Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH 03755;
| |
Collapse
|
36
|
Grow DA, Simmons DV, Gomez JA, Wanat MJ, McCarrey JR, Paladini CA, Navara CS. Differentiation and Characterization of Dopaminergic Neurons From Baboon Induced Pluripotent Stem Cells. Stem Cells Transl Med 2016; 5:1133-44. [PMID: 27343168 PMCID: PMC4996432 DOI: 10.5966/sctm.2015-0073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 03/23/2016] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED : The progressive death of dopamine producing neurons in the substantia nigra pars compacta is the principal cause of symptoms of Parkinson's disease (PD). Stem cells have potential therapeutic use in replacing these cells and restoring function. To facilitate development of this approach, we sought to establish a preclinical model based on a large nonhuman primate for testing the efficacy and safety of stem cell-based transplantation. To this end, we differentiated baboon fibroblast-derived induced pluripotent stem cells (biPSCs) into dopaminergic neurons with the application of specific morphogens and growth factors. We confirmed that biPSC-derived dopaminergic neurons resemble those found in the human midbrain based on cell type-specific expression of dopamine markers TH and GIRK2. Using the reverse transcriptase quantitative polymerase chain reaction, we also showed that biPSC-derived dopaminergic neurons express PAX6, FOXA2, LMX1A, NURR1, and TH genes characteristic of this cell type in vivo. We used perforated patch-clamp electrophysiology to demonstrate that biPSC-derived dopaminergic neurons fired spontaneous rhythmic action potentials and high-frequency action potentials with spike frequency adaption upon injection of depolarizing current. Finally, we showed that biPSC-derived neurons released catecholamines in response to electrical stimulation. These results demonstrate the utility of the baboon model for testing and optimizing the efficacy and safety of stem cell-based therapeutic approaches for the treatment of PD. SIGNIFICANCE Functional dopamine neurons were produced from baboon induced pluripotent stem cells, and their properties were compared to baboon midbrain cells in vivo. The baboon has advantages as a clinically relevant model in which to optimize the efficacy and safety of stem cell-based therapies for neurodegenerative diseases, such as Parkinson's disease. Baboons possess crucial neuroanatomical and immunological similarities to humans, and baboon pluripotent stem cells can be differentiated into functional neurons that mimic those in the human brain, thus laying the foundation for the utility of the baboon model for evaluating stem cell therapies.
Collapse
Affiliation(s)
- Douglas A Grow
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA; San Antonio Cellular Therapeutics Institute, San Antonio, Texas, USA; University of Texas at San Antonio Neurosciences Institute, San Antonio, Texas, USA
| | - DeNard V Simmons
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA; University of Texas at San Antonio Neurosciences Institute, San Antonio, Texas, USA
| | - Jorge A Gomez
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA; University of Texas at San Antonio Neurosciences Institute, San Antonio, Texas, USA
| | - Matthew J Wanat
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA; University of Texas at San Antonio Neurosciences Institute, San Antonio, Texas, USA
| | - John R McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA; San Antonio Cellular Therapeutics Institute, San Antonio, Texas, USA
| | - Carlos A Paladini
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA; University of Texas at San Antonio Neurosciences Institute, San Antonio, Texas, USA
| | - Christopher S Navara
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA; San Antonio Cellular Therapeutics Institute, San Antonio, Texas, USA;
| |
Collapse
|
37
|
Bolton DL, Pegu A, Wang K, McGinnis K, Nason M, Foulds K, Letukas V, Schmidt SD, Chen X, Todd JP, Lifson JD, Rao S, Michael NL, Robb ML, Mascola JR, Koup RA. Human Immunodeficiency Virus Type 1 Monoclonal Antibodies Suppress Acute Simian-Human Immunodeficiency Virus Viremia and Limit Seeding of Cell-Associated Viral Reservoirs. J Virol 2016; 90:1321-32. [PMID: 26581981 PMCID: PMC4719604 DOI: 10.1128/jvi.02454-15] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/05/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Combination antiretroviral therapy (cART) administered shortly after human immunodeficiency virus type 1 (HIV-1) infection can suppress viremia and limit seeding of the viral reservoir, but lifelong treatment is required for the majority of patients. Highly potent broadly neutralizing HIV-1 monoclonal antibodies (MAbs) can reduce plasma viremia when administered during chronic HIV-1 infection, but the therapeutic potential of these antibodies during acute infection is unknown. We tested the ability of HIV-1 envelope glycoprotein-specific broadly neutralizing MAbs to suppress acute simian-human immunodeficiency virus (SHIV) replication in rhesus macaques. Four groups of macaques were infected with SHIV-SF162P3 and received (i) the CD4-binding-site MAb VRC01; (ii) a combination of a more potent clonal relative of VRC01 (VRC07-523) and a V3 glycan-dependent MAb (PGT121); (iii) daily cART, all on day 10, just prior to expected peak plasma viremia; or (iv) no treatment. Daily cART was initiated 11 days after MAb administration and was continued for 13 weeks in all treated animals. Over a period of 11 days after a single administration, MAb treatment significantly reduced peak viremia, accelerated the decay slope, and reduced total viral replication compared to untreated controls. Proviral DNA in lymph node CD4 T cells was also diminished after treatment with the dual MAb. These data demonstrate the virological effect of potent MAbs and support future clinical trials that investigate HIV-1-neutralizing MAbs as adjunctive therapy with cART during acute HIV-1 infection. IMPORTANCE Treatment of chronic HIV-1 infection with potent broadly neutralizing HIV-1 MAbs has been shown to significantly reduce plasma viremia. However, the antiviral effect of MAb treatment during acute HIV-1 infection is unknown. Here, we demonstrate that MAbs targeting the HIV-1 envelope glycoprotein both suppress acute SHIV plasma viremia and limit CD4 T cell-associated viral DNA. These findings provide support for clinical trials of MAbs as adjunctive therapy with antiretroviral therapy during acute HIV-1 infection.
Collapse
Affiliation(s)
- Diane L Bolton
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA, and Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Keyun Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kathleen McGinnis
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Martha Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kathryn Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Valerie Letukas
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephen D Schmidt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Xuejun Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John Paul Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc./Frederick National Laboratory for Cancer Research, AIDS and Cancer Virus Program, Frederick, Maryland, USA
| | - Srinivas Rao
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Nelson L Michael
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Merlin L Robb
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA, and Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
38
|
Ishii-Watabe A, Tada M, Suzuki T, Kawasaki N. Nonclinical Evaluation of Next-generation Therapeutic Monoclonal Antibodies. YAKUGAKU ZASSHI 2015; 135:857-66. [DOI: 10.1248/yakushi.15-00007-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences
| | - Minoru Tada
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences
| | - Takuo Suzuki
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences
| | - Nana Kawasaki
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences
| |
Collapse
|
39
|
Caldwell RG, Marshall P, Fishel J. Method validation and reference range values for a peripheral blood immunophenotyping assay in non-human primates. J Immunotoxicol 2015; 13:64-76. [PMID: 25600312 DOI: 10.3109/1547691x.2014.1001098] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A peripheral blood immunophenotyping assay was developed and validated for determination of total T-lymphocytes, helper T-lymphocytes, cytotoxic T-lymphocytes, B-lymphocytes, and natural killer cells in cynomolgus monkeys. Validation parameters included assessment of precision, linearity, antibody optimization, stability of peripheral blood samples, and stability of fixed immunophenotyping samples. Total lymphocyte populations were determined using a heterogeneous lymphocyte gating strategy consisting of CD45 fluorescent staining and side-scatter demarcation. Relative lymphocyte subset values were determined using antigen-specific gating strategies. Absolute subset concentrations for each lymphocyte subset were subsequently determined using a dual-platform methodology wherein relative lymphocyte subset values (via flow cytometry analyses) were multiplied by the absolute total lymphocyte (via hematology analyses) values. Reference ranges are presented for cynomolgus monkey, rhesus monkey, and baboon. Additional 1-year longitudinal immunophenotyping values are presented for the cynomolgus monkey. The method validation and reference ranges presented in this research provide a robust analytical methodology for determination of peripheral blood lymphocyte subsets in various non-human primate species.
Collapse
|
40
|
Cauvin AJ, Peters C, Brennan F. Advantages and Limitations of Commonly Used Nonhuman Primate Species in Research and Development of Biopharmaceuticals. THE NONHUMAN PRIMATE IN NONCLINICAL DRUG DEVELOPMENT AND SAFETY ASSESSMENT 2015. [PMCID: PMC7149394 DOI: 10.1016/b978-0-12-417144-2.00019-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nonhuman primates (NHPs) have been used extensively during the past four decades for research and nonclinical development because they are close to humans in terms of genetics, anatomy, physiology, and immunology. They have been widely used in the development of infection models, leading to the generation of vaccines and drugs, as well as in the nonclinical pharmacologic and toxicologic assessment of biopharmaceuticals, especially in the fields of immunotherapy and oncology, despite the constant pressure to move to lower species. In many cases, NHPs are the only species that allows a correct risk assessment for humans. Nevertheless, limitations inherent to each species have to be considered before an investigation. This chapter shines some light on the respective interests and limitations of using cynomolgus monkeys, rhesus monkeys, and marmosets in medical research and nonclinical development, with a specific focus on reproduction and immunology.
Collapse
Affiliation(s)
- Annick J. Cauvin
- UCB Biopharma, New Medicine, Non-Clinical Development, Braine L’Alleud, Belgium
| | - Christopher Peters
- UCB Biopharma, New Medicine, Non-Clinical Development, Braine L’Alleud, Belgium
| | - Frank Brennan
- UCB Pharma, New Medicines, Non-Clinical Development, Slough, UK
| |
Collapse
|
41
|
Abstract
Passive immunotherapy represents a promising therapeutic intervention for a number of neoplastic, chronic inflammatory, and infectious diseases, with several monoclonal antibodies currently under development or already in use in the clinic. While Fab-antigen interactions play a crucial role in the activity of an antibody, it has become clear that Fc-mediated effector functions are involved during antibody-mediated activities in vivo. A complete understanding of the contributions of effector activities mediated by an antibody during its in vivo function is required for the development of antibodies with improved therapeutic efficacies. Animal models that are commonly used for the preclinical evaluation of antibodies include murine and non-human primate species, whose FcγRs present substantial structural, functional, and genetic variation compared with their human counterparts. Therefore, the use of such animal models provides limited information on the role of human IgG Fc-FcγR interactions during the in vivo activities of antibodies intended for human therapeutics. In this chapter, we describe the development and evaluation of an FcγR-humanized mouse model for the study of human FcγR function in vivo. In this model, endogenous mouse FcγR genes have been deleted and human FcγRs are expressed as transgenes that faithfully recapitulate the unique pattern of human FcγR expression. Evaluation of the in vivo activities of a number of cytotoxic or therapeutic antibodies using FcγR-humanized mice provided useful insights into human IgG Fc effector function. This mouse model has become a vital preclinical model for testing therapeutic human antibodies to treat malignancies, autoimmunity, inflammation, and infectious disease.
Collapse
|
42
|
Xiong F, Janko M, Walker M, Makropoulos D, Weinstock D, Kam M, Hrebien L. Analysis of cytokine release assay data using machine learning approaches. Int Immunopharmacol 2014; 22:465-79. [DOI: 10.1016/j.intimp.2014.07.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 07/01/2014] [Accepted: 07/21/2014] [Indexed: 12/18/2022]
|
43
|
Brennan FR, Cauvin A, Tibbitts J, Wolfreys A. Optimized nonclinical safety assessment strategies supporting clinical development of therapeutic monoclonal antibodies targeting inflammatory diseases. Drug Dev Res 2014; 75:115-61. [PMID: 24782266 DOI: 10.1002/ddr.21173] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 02/23/2014] [Indexed: 12/19/2022]
Abstract
An increasing number of immunomodulatory monoclonal antibodies (mAbs) and IgG Fc fusion proteins are either approved or in early-to-late stage clinical trials for the treatment of chronic inflammatory conditions, autoimmune diseases and organ transplant rejection. The exquisite specificity of mAbs, in combination with their multi-functional properties, high potency, long half-life (permitting intermittent dosing and prolonged pharamcological effects), and general lack of off-target toxicity makes them ideal therapeutics. Dosing with mAbs for these severe and debilitating but often non life-threatening diseases is usually prolonged, for several months or years, and not only affects adults, including sensitive populations such as woman of child-bearing potential (WoCBP) and the elderly, but also children. Immunosuppression is usually a therapeutic goal of these mAbs and when administered to patients whose treatment program often involves other immunosuppressive therapies, there is an inherent risk for frank immunosuppression and reduced host defence which when prolonged increases the risk of infection and cancer. In addition when mAbs interact with the immune system they can induce other adverse immune-mediated drug reactions such as infusion reactions, cytokine release syndrome, anaphylaxis, immune-complex-mediated pathology and autoimmunity. An overview of the nonclinical safety assessment and risk mitigation strategies utilized to characterize these immunomodulatory mAbs and Fc fusion proteins to support first-in human (FIH) studies and futher clinical development in inflammatory disease indications is provided. Specific emphasis is placed on the design of studies to qualify animal species for toxicology studies, early studies to investigate safety and define PK/PD relationships, FIH-enabling and chronic toxicology studies, immunotoxicity, developmental, reproductive and juvenile toxicity studies and studies to determine the potential for immunosuppression and reduced host defence against infection and cancer. Nonclinical strategies to facilitate clinical and market entry in the most efficient timeframe are presented.
Collapse
Affiliation(s)
- Frank R Brennan
- Preclinical Safety, New Medicines, UCB-Celltech, Slough, SL1 3WE, UK
| | | | | | | |
Collapse
|
44
|
Yeo ASL, Azhar NA, Yeow W, Talbot CC, Khan MA, Shankar EM, Rathakrishnan A, Azizan A, Wang SM, Lee SK, Fong MY, Manikam R, Sekaran SD. Lack of clinical manifestations in asymptomatic dengue infection is attributed to broad down-regulation and selective up-regulation of host defence response genes. PLoS One 2014; 9:e92240. [PMID: 24727912 PMCID: PMC3984081 DOI: 10.1371/journal.pone.0092240] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 02/19/2014] [Indexed: 12/25/2022] Open
Abstract
Objectives Dengue represents one of the most serious life-threatening vector-borne infectious diseases that afflicts approximately 50 million people across the globe annually. Whilst symptomatic infections are frequently reported, asymptomatic dengue remains largely unnoticed. Therefore, we sought to investigate the immune correlates conferring protection to individuals that remain clinically asymptomatic. Methods We determined the levels of neutralizing antibodies (nAbs) and gene expression profiles of host immune factors in individuals with asymptomatic infections, and whose cognate household members showed symptoms consistent to clinical dengue infection. Results We observed broad down-regulation of host defense response (innate, adaptive and matrix metalloprotease) genes in asymptomatic individuals as against symptomatic patients, with selective up-regulation of distinct genes that have been associated with protection. Selected down-regulated genes include: TNF α (TNF), IL8, C1S, factor B (CFB), IL2, IL3, IL4, IL5, IL8, IL9, IL10 and IL13, CD80, CD28, and IL18, MMP8, MMP10, MMP12, MMP15, MMP16, and MMP24. Selected up-regulated genes include: RANTES (CCL5), MIP-1α (CCL3L1/CCL3L3), MIP-1β (CCL4L1), TGFβ (TGFB), and TIMP1. Conclusion Our findings highlight the potential association of certain host genes conferring protection against clinical dengue. These data are valuable to better explore the mysteries behind the hitherto poorly understood immunopathogenesis of subclinical dengue infection.
Collapse
Affiliation(s)
- Adeline S. L. Yeo
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Nur Atiqah Azhar
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
- Perdana University Graduate School of Medicine & Centre for Bioinformatics, MARDI Complex, Jalan MAEPS Perdana, Serdang, Selangor Darul Ehsan, Malaysia
| | - Wanyi Yeow
- Perdana University Graduate School of Medicine & Centre for Bioinformatics, MARDI Complex, Jalan MAEPS Perdana, Serdang, Selangor Darul Ehsan, Malaysia
| | - C. Conover Talbot
- Institute for Basic Biomedical Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Mohammad Asif Khan
- Perdana University Graduate School of Medicine & Centre for Bioinformatics, MARDI Complex, Jalan MAEPS Perdana, Serdang, Selangor Darul Ehsan, Malaysia
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Esaki M. Shankar
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Anusyah Rathakrishnan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Azliyati Azizan
- Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Seok Mui Wang
- Institute for Medical Molecular Biotechnology, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor, Malaysia
| | - Siew Kim Lee
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Mun Yik Fong
- Department of Parasitology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Rishya Manikam
- Department of Trauma and Emergency Medicine, University of Malaya Medical Centre, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Shamala Devi Sekaran
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia
- * E-mail:
| |
Collapse
|
45
|
Bartholomaeus P, Semmler LY, Bukur T, Boisguerin V, Römer PS, Tabares P, Chuvpilo S, Tyrsin DY, Matskevich A, Hengel H, Castle J, Hünig T, Kalinke U. Cell Contact–Dependent Priming and Fc Interaction with CD32+ Immune Cells Contribute to the TGN1412-Triggered Cytokine Response. THE JOURNAL OF IMMUNOLOGY 2014; 192:2091-8. [DOI: 10.4049/jimmunol.1302461] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
46
|
The FcγR of humans and non-human primates and their interaction with IgG: implications for induction of inflammation, resistance to infection and the use of therapeutic monoclonal antibodies. Curr Top Microbiol Immunol 2014; 382:321-52. [PMID: 25116107 DOI: 10.1007/978-3-319-07911-0_15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Considerable effort has focused on the roles of the individual members of the FcγR receptor (FcγR) family in inflammatory diseases and humoral immunity. Recent work has revealed major roles in infection and in particular HIV pathogenesis and immunity. In addition, FcγR functions underpin the action of many of the successful therapeutic monoclonal antibodies. This emphasises the need for a greater understanding of FcγR function in humans and in the NHP which provides a key model for human immunity and preclinical testing of antibodies. We discuss recent key aspects of the human FcγR receptor biology and structure to define differences and similarities in activity between the human and macaque Fc receptors. These differences and similarities nuance the interpretation of infection and vaccine studies in the macaque. Indeed passive IgG antibody protection in lentivirus infection models in the macaque provided early evidence for the role of Fc receptors in anti-HIV immunity that have subsequently gained support from human vaccine trials. None-the-less the diverse functions and cellular contexts of FcγR receptor expression ensure there is much still to understand of the protective and deleterious effects of FcγRs in HIV infection. Careful comparative studies of human and non-human primate FcγRs will facilitate our appreciation of what attributes of HIV specific IgG antibodies, either acquired naturally or via vaccination, are most important for protection.
Collapse
|
47
|
Trist HM, Tan PS, Wines BD, Ramsland PA, Orlowski E, Stubbs J, Gardiner EE, Pietersz GA, Kent SJ, Stratov I, Burton DR, Hogarth PM. Polymorphisms and interspecies differences of the activating and inhibitory FcγRII of Macaca nemestrina influence the binding of human IgG subclasses. THE JOURNAL OF IMMUNOLOGY 2013; 192:792-803. [PMID: 24342805 DOI: 10.4049/jimmunol.1301554] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Little is known of the impact of Fc receptor (FcR) polymorphism in macaques on the binding of human (hu)IgG, and nothing is known of this interaction in the pig-tailed macaque (Macaca nemestrina), which is used in preclinical evaluation of vaccines and therapeutic Abs. We defined the sequence and huIgG binding characteristics of the M. nemestrina activating FcγRIIa (mnFcγRIIa) and inhibitory FcγRIIb (mnFcγRIIb) and predicted their structures using the huIgGFc/huFcγRIIa crystal structure. Large differences were observed in the binding of huIgG by mnFcγRIIa and mnFcγRIIb compared with their human FcR counterparts. MnFcγRIIa has markedly impaired binding of huIgG1 and huIgG2 immune complexes compared with huFcγRIIa (His(131)). In contrast, mnFcγRIIb has enhanced binding of huIgG1 and broader specificity, as, unlike huFcγRIIb, it avidly binds IgG2. Mutagenesis and molecular modeling of mnFcγRIIa showed that Pro(159) and Tyr(160) impair the critical FG loop interaction with huIgG. The enhanced binding of huIgG1 and huIgG2 by mnFcγRIIb was shown to be dependent on His(131) and Met(132). Significantly, both His(131) and Met(132) are conserved across FcγRIIb of rhesus and cynomolgus macaques. We identified functionally significant polymorphism of mnFcγRIIa wherein proline at position 131, also an important polymorphic site in huFcγRIIa, almost abolished binding of huIgG2 and huIgG1 and reduced binding of huIgG3 compared with mnFcγRIIa His(131). These marked interspecies differences in IgG binding between human and macaque FcRs and polymorphisms within species have implications for preclinical evaluation of Abs and vaccines in macaques.
Collapse
Affiliation(s)
- Halina M Trist
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Current challenges and opportunities in nonclinical safety testing of biologics. Drug Discov Today 2013; 18:1138-43. [DOI: 10.1016/j.drudis.2013.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/30/2013] [Accepted: 08/06/2013] [Indexed: 11/18/2022]
|
49
|
Vargas-Inchaustegui DA, Robert-Guroff M. Fc receptor-mediated immune responses: new tools but increased complexity in HIV prevention. Curr HIV Res 2013; 11:407-20. [PMID: 24191937 PMCID: PMC6288814 DOI: 10.2174/1570162x113116660063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/04/2013] [Accepted: 06/06/2013] [Indexed: 12/21/2022]
Abstract
The modest success of the RV144 HIV vaccine trial in Thailand and the ensuing suggestion that a Fc-receptormediated antibody activity might have played a role in the protection observed have intensified investigations on Fcrelated immune responses. HIV neutralizing antibodies have been and continue to be the focal point of research into humoral immune protection. However, recent knowledge that their protective efficacy can be augmented by Fc-FcR interactions has increased the complexity of identifying immune correlates of protection. If anything, continued studies of both humoral and cellular immune mechanisms point to the lack of a single protective anti-HIV immune response. Here we focus on humoral immunity, analyzing the role played by Fc receptor-related responses and discussing how new knowledge of their interactions requires further investigation, but may also spur novel vaccination approaches. We initially address classical Fc-receptor mediated anti-viral mechanisms including antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cell mediated viral inhibition (ADCVI), and antibody-dependent cellular phagocytosis (ADCP), as well as the effector cells that mediate these functions. Next, we summarize key aspects of FcR-Fc interactions that are important for potential control of HIV/SIV such as FcR polymorphisms and post-transcriptional modifications. Finally we discuss less commonly studied non-mechanistic anti-HIV immune functions: antibody avidity and envelopespecific B cell memory. Overall, a spectrum of immune responses, reflecting the immune system's redundancy, will likely be needed to prevent HIV infection and/or disease progression. Aside from elicitation of critical immune mechanisms, a successful vaccine will need to induce mature B cell responses and long-lasting immune memory.
Collapse
Affiliation(s)
- Diego A Vargas-Inchaustegui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, 41 Medlars Drive, Building 41, Room D804, Bethesda, MD 20192-5065, USA.
| | | |
Collapse
|
50
|
Mouse model recapitulating human Fcγ receptor structural and functional diversity. Proc Natl Acad Sci U S A 2012; 109:6181-6. [PMID: 22474370 DOI: 10.1073/pnas.1203954109] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The in vivo biological activities of IgG antibodies result from their bifunctional nature, in which antigen recognition by the Fab is coupled to the effector and immunomodulatory diversity found in the Fc domain. This diversity, resulting from both amino acid and glycan heterogeneity, is translated into cellular responses through Fcγ receptors (FcγRs), a structurally and functionally diverse family of cell surface receptors found throughout the immune system. Although many of the overall features of this system are maintained throughout mammalian evolution, species diversity has precluded direct analysis of human antibodies in animal species, and, thus, detailed investigations into the unique features of the human IgG antibodies and their FcγRs have been limited. We now report the development of a mouse model in which all murine FcγRs have been deleted and human FcγRs, encoded as transgenes, have been inserted into the mouse genome resulting in recapitulation of the unique profile of human FcγR expression. These human FcγRs are shown to function to mediate the immunomodulatory, inflammatory, and cytotoxic activities of human IgG antibodies and Fc engineered variants and provide a platform for the detailed mechanistic analysis of therapeutic and pathogenic IgG antibodies.
Collapse
|