1
|
Jacobs JW, Booth GS, Raza S, Clark LM, Fasano RM, Gavriilaki E, Abels EA, Binns TC, Duque MA, McQuilten ZK, Mingot-Castellano ME, Savani BN, Sharma D, Tran MH, Tormey CA, Moise KJ, Bloch EM, Adkins BD. Current state and potential applications of neonatal Fc receptor (FcRn) inhibitors in hematologic conditions. Am J Hematol 2024; 99:2351-2366. [PMID: 39324647 DOI: 10.1002/ajh.27487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024]
Abstract
The neonatal fragment crystallizable (Fc) receptor (FcRn) transports IgG across mucosal surfaces and the placenta and protects IgG from degradation. Numerous clinical trials are investigating therapeutic FcRn inhibition for various immune-mediated neuromuscular and rheumatologic conditions; however, FcRn inhibition also represents a potential therapy for IgG-mediated hematologic conditions (e.g., immune thrombocytopenia, autoimmune hemolytic anemia, immune thrombotic thrombocytopenic purpura, acquired hemophilia, red blood cell/platelet alloimmunization). Current evidence derived from both in vitro and in vivo studies suggests that FcRn inhibitors effectively reduce total IgG levels without impacting its production or altering the levels of other immunoglobulin isotypes. Moreover, the risk of serious adverse events, including serious infections, appears to be lower than that seen with other commonly used immunomodulatory/immunosuppressive therapies, albeit in the setting of limited clinical trial data. Ultimately, additional clinical trials that include varied patient populations are required prior to incorporating these agents into standard treatment algorithms for most hematologic conditions. However, based on the pathophysiology of IgG-mediated hematologic disorders and the mechanism of action of FcRn inhibitors, these agents may represent a future novel therapeutic strategy for patients with hematologic conditions caused by IgG antibodies.
Collapse
Affiliation(s)
- Jeremy W Jacobs
- Division of Transfusion Medicine, Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Garrett S Booth
- Division of Transfusion Medicine, Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sheharyar Raza
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Canadian Blood Services, Medical Affairs and Innovation, Toronto, Ontario, Canada
| | - Landon M Clark
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ross M Fasano
- Department of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapy, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Eleni Gavriilaki
- Hematology Department and Bone Marrow Transplant (BMT) Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elizabeth A Abels
- Department of Obstetrics and Gynecology, Bridgeport Hospital/Yale University, Bridgeport, Connecticut, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Thomas C Binns
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Miriam Andrea Duque
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Zoe K McQuilten
- Department of Haematology, Monash Health, Melbourne, Victoria, Australia
| | - María Eva Mingot-Castellano
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Deva Sharma
- Division of Transfusion Medicine, Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Minh-Ha Tran
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, California, USA
| | - Christopher A Tormey
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kenneth J Moise
- Department of Women's Health, Dell Medical School-University of Texas at Austin, Austin, Texas, USA
- Comprehensive Fetal Care Center, Dell Children's Medical Center, Austin, Texas, USA
| | - Evan M Bloch
- Division of Transfusion Medicine, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Brian D Adkins
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
2
|
Serreze DV, Dwyer JR, Racine JJ. Advancing Animal Models of Human Type 1 Diabetes. Cold Spring Harb Perspect Med 2024; 14:a041587. [PMID: 38886067 PMCID: PMC11444302 DOI: 10.1101/cshperspect.a041587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Multiple rodent models have been developed to study the basis of type 1 diabetes (T1D). However, nonobese diabetic (NOD) mice and derivative strains still provide the gold standard for dissecting the basis of the autoimmune responses underlying T1D. Here, we review the developmental origins of NOD mice, and how they and derivative strains have been used over the past several decades to dissect the genetic and immunopathogenic basis of T1D. Also discussed are ways in which the immunopathogenic basis of T1D in NOD mice and humans are similar or differ. Additionally reviewed are efforts to "humanize" NOD mice and derivative strains to provide improved models to study autoimmune responses contributing to T1D in human patients.
Collapse
|
3
|
Qian S, Zhang D, Yang Z, Li R, Zhang X, Gao F, Yu L. The role of immunoglobulin transport receptor, neonatal Fc receptor in mucosal infection and immunity and therapeutic intervention. Int Immunopharmacol 2024; 138:112583. [PMID: 38971109 DOI: 10.1016/j.intimp.2024.112583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/15/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
The neonatal Fc receptor (FcRn) can transport IgG and antigen-antibody complexes participating in mucosal immune responses that protect the host from most pathogens' invasion via the respiratory, digestive, and urogenital tracts. FcRn expression can be triggered upon stimulation with pathogenic invasion on mucosal surfaces, which may significantly modulate the innate immune response of the host. As an immunoglobulin transport receptor, FcRn is implicated in the pathophysiology of immune-related diseases such as infection and autoimmune disorders. In this review, we thoroughly summarize the recent advancement of FcRn in mucosal immunity and its therapeutic strategy. This includes insights into its regulation mechanisms of FcRn expression influenced by pathogens, its emerging role in mucosal immunity and its potential probability as a therapeutic target in infection and autoimmune diseases.
Collapse
Affiliation(s)
- Shaoju Qian
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Danqiong Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Zishan Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Ruixue Li
- Department of Otolaryngology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Xuehan Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Feifei Gao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China.
| |
Collapse
|
4
|
Shaw TM, Huey D, Mousa-Makky M, Compaleo J, Nennig K, Shah AP, Jiang F, Qiu X, Klipsic D, Rowland RRR, Slukvin II, Sullender ME, Baldridge MT, Li H, Warren CJ, Bailey AL. The neonatal Fc receptor (FcRn) is a pan-arterivirus receptor. Nat Commun 2024; 15:6726. [PMID: 39112502 PMCID: PMC11306234 DOI: 10.1038/s41467-024-51142-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Arteriviruses infect a variety of mammalian hosts, but the receptors used by these viruses to enter cells are poorly understood. We identified the neonatal Fc receptor (FcRn) as an important pro-viral host factor via comparative genome-wide CRISPR-knockout screens with multiple arteriviruses. Using a panel of cell lines and divergent arteriviruses, we demonstrate that FcRn is required for the entry step of arterivirus infection and serves as a molecular barrier to arterivirus cross-species infection. We also show that FcRn synergizes with another known arterivirus entry factor, CD163, to mediate arterivirus entry. Overexpression of FcRn and CD163 sensitizes non-permissive cells to infection and enables the culture of fastidious arteriviruses. Treatment of multiple cell lines with a pre-clinical anti-FcRn monoclonal antibody blocked infection and rescued cells from arterivirus-induced death. Altogether, this study identifies FcRn as a novel pan-arterivirus receptor, with implications for arterivirus emergence, cross-species infection, and host-directed pan-arterivirus countermeasure development.
Collapse
Affiliation(s)
- Teressa M Shaw
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Devra Huey
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, OH, USA
| | - Makky Mousa-Makky
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, OH, USA
| | - Jared Compaleo
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, OH, USA
| | - Kylie Nennig
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Aadit P Shah
- Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Fei Jiang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Xueer Qiu
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Devon Klipsic
- Research Animal Resources and Compliance (RARC), University of Wisconsin-Madison, Madison, WI, USA
| | - Raymond R R Rowland
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Igor I Slukvin
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Meagan E Sullender
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Megan T Baldridge
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Haichang Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
- Department of Surgery, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Cody J Warren
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA.
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, OH, USA.
- Center for RNA Biology, The Ohio State University, Columbus, OH, USA.
- Center for Retrovirus Research, The Ohio State University, Columbus, OH, USA.
| | - Adam L Bailey
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
5
|
Aschenbrenner I, Böckler M, Franke F, Liebl K, Catici DAM, Brandl M, Behnke J, Feige MJ. Development of an enabling platform biotechnology for the production of proteins. Biol Chem 2024; 405:471-483. [PMID: 38916991 DOI: 10.1515/hsz-2023-0376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/27/2024] [Indexed: 06/27/2024]
Abstract
Protein-based drugs are a mainstay of modern medicine. In contrast to antibodies, most of these need highly individualized production processes which often limits their development. Here, we develop an immunoglobulin domain tag (i-Tag), which can be fused to any protein of interest. This tag is made of a linear arrangement of antibody light chain constant domains. It enhances expression as well as secretion of the fusion partner and allows for simple purification of several structurally and functionally distinct fusion proteins. Furthermore, it improves the biophysical characteristics of most fusion proteins tested, is inert, and does not compromise the fusion partners' functionality. Taken together, the i-Tag should facilitate the development of biopharmaceuticals and diagnostic proteins otherwise lacking a common structural element.
Collapse
Affiliation(s)
- Isabel Aschenbrenner
- TUM School of Natural Sciences, Department of Bioscience, Center for Functional Protein Assemblies (CPA), 9184 Technical University of Munich , D-85748 Garching, Germany
| | - Maximilian Böckler
- TUM School of Natural Sciences, Department of Bioscience, Center for Functional Protein Assemblies (CPA), 9184 Technical University of Munich , D-85748 Garching, Germany
| | - Fabian Franke
- TUM School of Natural Sciences, Department of Bioscience, Center for Functional Protein Assemblies (CPA), 9184 Technical University of Munich , D-85748 Garching, Germany
| | - Korbinian Liebl
- TUM School of Natural Sciences, Department of Bioscience, Center for Functional Protein Assemblies (CPA), 9184 Technical University of Munich , D-85748 Garching, Germany
| | - Dragana A M Catici
- TUM School of Natural Sciences, Department of Bioscience, Center for Functional Protein Assemblies (CPA), 9184 Technical University of Munich , D-85748 Garching, Germany
| | - Matthias Brandl
- TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center (BNMRZ), 9184 Technical University of Munich , D-85748 Garching, Germany
| | - Julia Behnke
- TUM School of Medicine, Department of Surgery, Klinikum Rechts der Isar München, 9184 Technical University of Munich , D-81675 Munich, Germany
| | - Matthias J Feige
- TUM School of Natural Sciences, Department of Bioscience, Center for Functional Protein Assemblies (CPA), 9184 Technical University of Munich , D-85748 Garching, Germany
| |
Collapse
|
6
|
Macri C, Paxman M, Jenika D, Lin XP, Elahi Z, Gleeson PA, Caminschi I, Lahoud MH, Villadangos JA, Mintern JD. FcRn regulates antigen presentation in dendritic cells downstream of DEC205-targeted vaccines. NPJ Vaccines 2024; 9:76. [PMID: 38594284 PMCID: PMC11003989 DOI: 10.1038/s41541-024-00854-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 02/29/2024] [Indexed: 04/11/2024] Open
Abstract
Dendritic cell (DC)-targeted vaccination is a new mode of antigen delivery that relies on the use of monoclonal antibodies (mAb) to target antigen to specific DC subsets. The neonatal Fc receptor (FcRn) is a non-classical Fc receptor that binds to immunoglobulin G (IgG) in acidified endosomes and controls its intracellular transport and recycling. FcRn is known to participate in the antigen presentation of immune complexes, however its contribution to DC-targeted vaccination has not previously been examined. Here we have investigated the role of FcRn in antigen presentation using antigen conjugated to IgG mAb which target specific DC receptors, including DEC205 and Clec9A expressed by the conventional DC 1 (cDC1) subset. We show that FcRn is expressed at high levels by cDC1, both at steady-state and following activation and plays a significant role in MHC I cross-presentation and MHC II presentation of antigens that are targeted to cDC1 via mAb specific for DEC205. This effect of FcRn is intrinsic to cDC1 and FcRn impacts the efficacy of anti-DEC205-mediated vaccination against B cell lymphoma. In contrast, FcRn does not impact presentation of antigens targeted to Clec9A and does not regulate presentation of cell-associated antigen. These data highlight a new and unique role of FcRn in controlling the immunogenicity of anti-DEC205-based vaccination, with consequences for exploiting this pathway to improve DC-targeted vaccine outcomes.
Collapse
Affiliation(s)
- Christophe Macri
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, The University of Melbourne, Victoria, 3010, Australia
| | - Matthew Paxman
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, The University of Melbourne, Victoria, 3010, Australia
| | - Devi Jenika
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, The University of Melbourne, Victoria, 3010, Australia
| | - Xiao Peng Lin
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, The University of Melbourne, Victoria, 3010, Australia
| | - Zahra Elahi
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Paul A Gleeson
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, The University of Melbourne, Victoria, 3010, Australia
| | - Irina Caminschi
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Mireille H Lahoud
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, The University of Melbourne, Victoria, 3010, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, The University of Melbourne, Victoria, 3010, Australia.
| |
Collapse
|
7
|
Haddad G, Blaine J. Identification of Four Mouse FcRn Splice Variants and FcRn-Specific Vesicles. Cells 2024; 13:594. [PMID: 38607033 PMCID: PMC11012118 DOI: 10.3390/cells13070594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
Research into the neonatal Fc receptor (FcRn) has increased dramatically ever since Simister and Mostov first purified a rat version of the receptor. Over the years, FcRn has been shown to function not only as a receptor that transfers immunity from mother to fetus but also performs an array of different functions that include transport and recycling of immunoglobulins and albumin in the adult. Due to its important cellular roles, several clinical trials have been designed to either inhibit/enhance FcRn function or develop of non-invasive therapeutic delivery system such as fusion of drugs to IgG Fc or albumin to enhance delivery inside the cells. Here, we report the accidental identification of several FcRn alternatively spliced variants in both mouse and human cells. The four new mouse splice variants are capable of binding immunoglobulins' Fc and Fab portions. In addition, we have identified FcRn-specific vesicles in which immunoglobulins and albumin can be stored and that are involved in the endosomal-lysosomal system. The complexity of FcRn functions offers significant potential to design and develop novel and targeted therapeutics.
Collapse
Affiliation(s)
| | - Judith Blaine
- Division of Renal Disease and Hypertension, Department of Medicine, School of Medicine, University of Colorado, Aurora, CO 80045, USA;
| |
Collapse
|
8
|
Okamoto M, Sasaki R, Ikeda K, Doi K, Tatsumi F, Oshima K, Kojima T, Mizushima S, Ikegami K, Yoshimura T, Furukawa K, Kobayashi M, Horio F, Murai A. FcRY is a key molecule controlling maternal blood IgY transfer to yolks during egg development in avian species. Front Immunol 2024; 15:1305587. [PMID: 38487530 PMCID: PMC10938909 DOI: 10.3389/fimmu.2024.1305587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Maternal immunoglobulin transfer plays a key role in conferring passive immunity to neonates. Maternal blood immunoglobulin Y (IgY) in avian species is transported to newly-hatched chicks in two steps: 1) IgY is transported from the maternal circulation to the yolk of maturing oocytes, 2) the IgY deposited in yolk is transported to the circulation of the embryo via the yolk sac membrane. An IgY-Fc receptor, FcRY, is involved in the second step, but the mechanism of the first step is still unclear. We determined whether FcRY was also the basis for maternal blood IgY transfer to the yolk in the first step during egg development. Immunohistochemistry revealed that FcRY was expressed in the capillary endothelial cells in the internal theca layer of the ovarian follicle. Substitution of the amino acid residue in Fc region of IgY substantially changed the transport efficiency of IgY into egg yolks when intravenously-injected into laying quail; the G365A mutant had a high transport efficiency, but the Y363A mutant lacked transport ability. Binding analyses of IgY mutants to FcRY indicated that the mutant with a high transport efficiency (G365A) had a strong binding activity to FcRY; the mutants with a low transport efficiency (G365D, N408A) had a weak binding activity to FcRY. One exception, the Y363A mutant had a remarkably strong binding affinity to FcRY, with a small dissociation rate. The injection of neutralizing FcRY antibodies in laying quail markedly reduced IgY uptake into egg yolks. The neutralization also showed that FcRY was engaged in prolongation of half-life of IgY in the blood; FcRY is therefore a multifunctional receptor that controls avian immunity. The pattern of the transport of the IgY mutants from the maternal blood to the egg yolk was found to be identical to that from the fertilized egg yolk to the newly-hatched chick blood circulation, via the yolk sac membrane. FcRY is therefore a critical IgY receptor that regulates the IgY uptake from the maternal blood circulation into the yolk of avian species, further indicating that the two steps of maternal-newly-hatched IgY transfer are controlled by a single receptor.
Collapse
Affiliation(s)
- Mayuko Okamoto
- Laboratory of Animal Nutrition, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Ryo Sasaki
- Laboratory of Animal Nutrition, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Koki Ikeda
- Laboratory of Animal Nutrition, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Kasumi Doi
- Laboratory of Animal Nutrition, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Fumiya Tatsumi
- Laboratory of Animal Nutrition, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Kenzi Oshima
- Laboratory of Molecular Bioregulation, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takaaki Kojima
- Laboratory of Molecular Biotechnology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Shusei Mizushima
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Keisuke Ikegami
- Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takashi Yoshimura
- Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya, Japan
| | - Kyohei Furukawa
- Laboratory of Animal Nutrition, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Misato Kobayashi
- Laboratory of Animal Nutrition, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Fumihiko Horio
- Laboratory of Animal Nutrition, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Atsushi Murai
- Laboratory of Animal Nutrition, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
9
|
Müller L, Dabbiru VAS, Schönborn L, Greinacher A. Therapeutic strategies in FcγIIA receptor-dependent thrombosis and thromboinflammation as seen in heparin-induced thrombocytopenia (HIT) and vaccine-induced immune thrombocytopenia and thrombosis (VITT). Expert Opin Pharmacother 2024; 25:281-294. [PMID: 38465524 DOI: 10.1080/14656566.2024.2328241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/05/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Fcγ-receptors (FcγR) are membrane receptors expressed on a variety of immune cells, specialized in recognition of the Fc part of immunoglobulin G (IgG) antibodies. FcγRIIA-dependent platelet activation in platelet factor 4 (PF4) antibody-related disorders have gained major attention, when these antibodies were identified as the cause of the adverse vaccination event termed vaccine-induced immune thrombocytopenia and thrombosis (VITT) during the COVID-19 vaccination campaign. With the recognition of anti-PF4 antibodies as cause for severe spontaneous and sometimes recurrent thromboses independent of vaccination, their clinical relevance extended far beyond heparin-induced thrombocytopenia (HIT) and VITT. AREAS COVERED Patients developing these disorders show life-threatening thromboses, and the outcome is highly dependent on effective treatment. This narrative literature review summarizes treatment options for HIT and VITT that are currently available for clinical application and provides the perspective toward new developments. EXPERT OPINION Nearly all these novel approaches are based on in vitro, preclinical observations, or case reports with only limited implementation in clinical practice. The therapeutic potential of these approaches still needs to be proven in larger cohort studies to ensure treatment efficacy and long-term patient safety.
Collapse
Affiliation(s)
- Luisa Müller
- Institut für Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Venkata A S Dabbiru
- Institut für Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Linda Schönborn
- Institut für Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Andreas Greinacher
- Institut für Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
10
|
Proctor JR, Wong H. Time-dependent clearance can confound exposure-response analysis of therapeutic antibodies: A comprehensive review of the current literature. Clin Transl Sci 2024; 17:e13676. [PMID: 37905360 PMCID: PMC10766027 DOI: 10.1111/cts.13676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023] Open
Abstract
Exposure-response (ER) analysis is used to optimize dose and dose regimens during clinical development. Characterization of relationships between drug exposure and efficacy or safety outcomes can be utilized to make dose adjustments that improve patient response. Therapeutic antibodies typically show predictable pharmacokinetics (PK) but can exhibit clearance that decreases over time due to treatment. Moreover, time-dependent changes in clearance are frequently associated with drug response, with larger decreases in clearance and increased exposure seen in patients who respond to treatment. This often confounds traditional ER analysis, as drug response influences exposure rather than the reverse. In this review, we survey published population PK analyses for reported time-dependent drug clearance effects across 158 therapeutic antibodies approved or in regulatory review. We describe the mechanisms by which time-dependent clearance can arise, and evaluate trends in frequency, magnitude, and time scale of changes in clearance with respect to indication, mechanistic interpretation of time-dependence, and PK modeling techniques employed. We discuss the modeling and simulation strategies commonly used to characterize time-dependent clearance, and examples where time-dependent clearance has impeded ER analysis. A case study using population model simulation was explored to interrogate the impact of time-dependent clearance on ER analysis and how it can lead to spurious conclusions. Overall, time-dependent clearance arises frequently among therapeutic antibodies and has spurred erroneous conclusions in ER analysis. Appropriate PK modeling techniques aid in identifying and characterizing temporal shifts in exposure that may impede accurate ER assessment and successful dose optimization.
Collapse
Affiliation(s)
- Jeffrey R. Proctor
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Harvey Wong
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
11
|
Nyesiga B, Levin M, Säll A, Rosén A, Jansson K, Fritzell S, Hägerbrand K, Weilguny D, von Schantz L. RUBY® - a tetravalent (2+2) bispecific antibody format with excellent functionality and IgG-like stability, pharmacology and developability properties. MAbs 2024; 16:2330113. [PMID: 38527972 DOI: 10.1080/19420862.2024.2330113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/08/2024] [Indexed: 03/27/2024] Open
Abstract
Despite the large number of existing bispecific antibody (bsAb) formats, the generation of novel bsAbs is still associated with development and bioprocessing challenges. Here, we present RUBY, a novel bispecific antibody format that allows rapid generation of bsAbs that fulfill key development criteria. The RUBYTM format has a 2 + 2 geometry, where two Fab fragments are linked via their light chains to the C-termini of an IgG, and carries mutations for optimal chain pairing. The unique design enables generation of bsAbs with mAb-like attributes. Our data demonstrate that RUBY bsAbs are compatible with small-scale production systems for screening purposes and can be produced at high yields (>3 g/L) from stable cell lines. The bsAbs produced are shown to, in general, contain low amounts of aggregates and display favorable solubility and stress endurance profiles. Further, compatibility with various IgG isotypes is shown and tailored Fc gamma receptor binding confirmed. Also, retained interaction with FcRn is demonstrated to translate into a pharmacokinetic profile in mice and non-human primates that is comparable to mAb controls. Functionality of conditional active RUBY bsAbs is confirmed in vitro. Anti-tumor effects in vivo have previously been demonstrated, and shown to be superior to a comparable mAb, and here it is further shown that RUBY bsAbs penetrate and localize to tumor tissue in vivo. In all, the RUBY format has attractive mAb-like attributes and offers the possibility to mitigate many of the development challenges linked to other bsAb formats, facilitating both high functionality and developability.
Collapse
Affiliation(s)
- Barnabas Nyesiga
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
- Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Mattias Levin
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Anna Säll
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Anna Rosén
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Kim Jansson
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Sara Fritzell
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | | | | |
Collapse
|
12
|
Mina-Osorio P, Tran MH, Habib AA. Therapeutic Plasma Exchange Versus FcRn Inhibition in Autoimmune Disease. Transfus Med Rev 2024; 38:150767. [PMID: 37867088 DOI: 10.1016/j.tmrv.2023.150767] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 10/24/2023]
Abstract
Therapeutic plasma exchange (TPE or PLEX) is used in a broad range of autoimmune diseases, with the goal of removing autoantibodies from the circulation. A newer approach for the selective removal of immunoglobulin G (IgG) antibodies is the use of therapeutic molecules targeting the neonatal Fc receptor (FcRn). FcRn regulates IgG recycling, and its inhibition results in a marked decrease in circulating autoantibodies of the IgG subtype. The difference between FcRn inhibition and PLEX is often questioned. With anti-FcRn monoclonal antibodies (mAbs) and fragments only recently entering this space, limited data are available regarding long-term efficacy and safety. However, the biology of FcRn is well understood, and mounting evidence regarding the efficacy, safety, and potential differences among compounds in development is available, allowing us to compare against nonselective plasma protein depletion methods such as PLEX. FcRn inhibitors may have distinct advantages and disadvantages over PLEX in certain scenarios. Use of PLEX is preferred over FcRn inhibition where removal of antibodies other than IgG or when concomitant repletion of missing plasma proteins is needed for therapeutic benefit. Also, FcRn targeting has not yet been studied for use in acute flares or crisis states of IgG-mediated diseases. Compared with PLEX, FcRn inhibition is associated with less invasive access requirements, more specific removal of IgG versus other immunoglobulins without a broad impact on circulating proteins, and any impacts on other therapeutic drug levels are restricted to other mAbs. In addition, the degree of IgG reduction is similar with FcRn inhibitors compared with that afforded by PLEX. Here we describe the scientific literature regarding the use of PLEX and FcRn inhibitors in autoimmune diseases and provide an expert discussion around the potential benefits of these options in varying clinical conditions and scenarios.
Collapse
Affiliation(s)
| | - Minh-Ha Tran
- Department of Pathology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ali A Habib
- Department of Neurology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
13
|
Racine JJ, Misherghi A, Dwyer JR, Maser R, Forte E, Bedard O, Sattler S, Pugliese A, Landry L, Elso C, Nakayama M, Mannering S, Rosenthal N, Serreze DV. HLA-DQ8 Supports Development of Insulitis Mediated by Insulin-Reactive Human TCR-Transgenic T Cells in Nonobese Diabetic Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1792-1805. [PMID: 37877672 PMCID: PMC10939972 DOI: 10.4049/jimmunol.2300303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/28/2023] [Indexed: 10/26/2023]
Abstract
In an effort to improve HLA-"humanized" mouse models for type 1 diabetes (T1D) therapy development, we previously generated directly in the NOD strain CRISPR/Cas9-mediated deletions of various combinations of murine MHC genes. These new models improved upon previously available platforms by retaining β2-microglobulin functionality in FcRn and nonclassical MHC class I formation. As proof of concept, we generated H2-Db/H2-Kd double knockout NOD mice expressing human HLA-A*0201 or HLA-B*3906 class I variants that both supported autoreactive diabetogenic CD8+ T cell responses. In this follow-up work, we now describe the creation of 10 new NOD-based mouse models expressing various combinations of HLA genes with and without chimeric transgenic human TCRs reactive to proinsulin/insulin. The new TCR-transgenic models develop differing levels of insulitis mediated by HLA-DQ8-restricted insulin-reactive T cells. Additionally, these transgenic T cells can transfer insulitis to newly developed NSG mice lacking classical murine MHC molecules, but expressing HLA-DQ8. These new models can be used to test potential therapeutics for a possible capacity to reduce islet infiltration or change the phenotype of T cells expressing type 1 diabetes patient-derived β cell autoantigen-specific TCRs.
Collapse
Affiliation(s)
| | - Adel Misherghi
- The Jackson Laboratory, Bar Harbor, ME
- College of the Atlantic, Bar Harbor, ME
| | | | | | | | | | - Susanne Sattler
- Imperial College London, London, United Kingdom
- Medical University Graz, Graz, Austria
| | - Alberto Pugliese
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Laurie Landry
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Colleen Elso
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Stuart Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, ME
- Imperial College London, London, United Kingdom
| | | |
Collapse
|
14
|
Bauer-Smith H, Sudol ASL, Beers SA, Crispin M. Serum immunoglobulin and the threshold of Fc receptor-mediated immune activation. Biochim Biophys Acta Gen Subj 2023; 1867:130448. [PMID: 37652365 PMCID: PMC11032748 DOI: 10.1016/j.bbagen.2023.130448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Antibodies can mediate immune recruitment or clearance of immune complexes through the interaction of their Fc domain with cellular Fc receptors. Clustering of antibodies is a key step in generating sufficient avidity for efficacious receptor recognition. However, Fc receptors may be saturated with prevailing, endogenous serum immunoglobulin and this raises the threshold by which cellular receptors can be productively engaged. Here, we review the factors controlling serum IgG levels in both healthy and disease states, and discuss how the presence of endogenous IgG is encoded into the functional activation thresholds for low- and high-affinity Fc receptors. We discuss the circumstances where antibody engineering can help overcome these physiological limitations of therapeutic antibodies. Finally, we discuss how the pharmacological control of Fc receptor saturation by endogenous IgG is emerging as a feasible mechanism for the enhancement of antibody therapeutics.
Collapse
Affiliation(s)
- Hannah Bauer-Smith
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK; Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Abigail S L Sudol
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Stephen A Beers
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK.
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
15
|
Dylewski JF, Haddad G, Blaine J. Exploiting the neonatal Fc receptor to treat kidney disease. Kidney Int 2023; 105:S0085-2538(23)00747-0. [PMID: 39491149 PMCID: PMC11068363 DOI: 10.1016/j.kint.2023.09.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 05/07/2024]
Abstract
The neonatal Fc receptor (FcRn) was initially discovered as the receptor that allowed passive immunity in newborns by transporting maternal IgG through the placenta and enterocytes. Since its initial discovery, FcRn has been found to exist throughout all stages of life and in many different cell types. Beyond passive immunity, FcRn is necessary for intrinsic albumin and IgG recycling and is important for antigen processing and presentation. Given its multiple important roles, FcRn has been utilized in many disease treatments including a new class of agents that were developed to inhibit FcRn for treatment of a variety of autoimmune diseases. Certain cell populations within the kidney also express high levels of this receptor. Specifically, podocytes, proximal tubule epithelial cells, and vascular endothelial cells have been found to utilize FcRn. In this review, we summarize what is known about FcRn and its function within the kidney. We also discuss how FcRn has been used for therapeutic benefit, including how newer FcRn inhibiting agents are being used to treat autoimmune diseases. Lastly, we will discuss what renal diseases may respond to FcRn inhibitors and how further work studying FcRn within the kidney may lead to therapies for kidney diseases.
Collapse
Affiliation(s)
- James F Dylewski
- Division of Nephrology, Denver Health Medical Center, Denver, CO, USA; Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA.
| | - George Haddad
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Judith Blaine
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
16
|
Pyzik M, Kozicky LK, Gandhi AK, Blumberg RS. The therapeutic age of the neonatal Fc receptor. Nat Rev Immunol 2023; 23:415-432. [PMID: 36726033 PMCID: PMC9891766 DOI: 10.1038/s41577-022-00821-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 02/03/2023]
Abstract
IgGs are essential soluble components of the adaptive immune response that evolved to protect the body from infection. Compared with other immunoglobulins, the role of IgGs is distinguished and enhanced by their high circulating levels, long half-life and ability to transfer from mother to offspring, properties that are conferred by interactions with neonatal Fc receptor (FcRn). FcRn binds to the Fc portion of IgGs in a pH-dependent manner and protects them from intracellular degradation. It also allows their transport across polarized cells that separate tissue compartments, such as the endothelium and epithelium. Further, it is becoming apparent that FcRn functions to potentiate cellular immune responses when IgGs, bound to their antigens, form IgG immune complexes. Besides the protective role of IgG, IgG autoantibodies are associated with numerous pathological conditions. As such, FcRn blockade is a novel and effective strategy to reduce circulating levels of pathogenic IgG autoantibodies and curtail IgG-mediated diseases, with several FcRn-blocking strategies on the path to therapeutic use. Here, we describe the current state of knowledge of FcRn-IgG immunobiology, with an emphasis on the functional and pathological aspects, and an overview of FcRn-targeted therapy development.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lisa K Kozicky
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amit K Gandhi
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Digestive Diseases Center, Boston, MA, USA.
| |
Collapse
|
17
|
Zhou Y, Jiang S. Roles of FcRn in Antigen-Presenting Cells during Autoimmunity and a Clinical Evaluation of Efgartigimod as an FcRn Blocker. Pathogens 2023; 12:817. [PMID: 37375507 DOI: 10.3390/pathogens12060817] [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: 05/14/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The immune system is a complex network of multiple cells, tissues, and organs that protects the body against foreign pathogenic invaders. However, the immune system may mistakenly attack healthy cells and tissues due to the cross-reactivity of anti-pathogen immunity, leading to autoimmunity by autoreactive T cells and/or autoantibody-secreting B cells. Autoantibodies can accumulate, resulting in tissue or organ damage. The neonatal crystallizable fragment receptor (FcRn) is an important factor in immune regulation through controlling the trafficking and recycling of immunoglobulin G (IgG) molecules, the most abundant antibody in humoral immunity. In addition to its role in IgG trafficking and recycling, FcRn is also involved in antigen presentation, which is a crucial step in the activation of the adaptive immune response via directing the internalization and trafficking of antigen-bound IgG immune complexes into compartments of degradation and presentation in antigen-presenting cells. Efgartigimod, an FcRn inhibitor, has shown promise in reducing the levels of autoantibodies and alleviating the autoimmune severity of myasthenia gravis, primary immune thrombocytopenia, and pemphigus vulgaris/foliaceus. This article aims to provide an overview of the importance of FcRn in antigen-presenting cells and its potential as a therapeutic target in autoimmune diseases, using efgartigimod as an example.
Collapse
Affiliation(s)
- Yihan Zhou
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Shisong Jiang
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
18
|
Gjølberg TT, Wik JA, Johannessen H, Krüger S, Bassi N, Christopoulos PF, Bern M, Foss S, Petrovski G, Moe MC, Haraldsen G, Fosse JH, Skålhegg BS, Andersen JT, Sundlisæter E. Antibody blockade of Jagged1 attenuates choroidal neovascularization. Nat Commun 2023; 14:3109. [PMID: 37253747 PMCID: PMC10229650 DOI: 10.1038/s41467-023-38563-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/08/2023] [Indexed: 06/01/2023] Open
Abstract
Antibody-based blocking of vascular endothelial growth factor (VEGF) reduces choroidal neovascularization (CNV) and retinal edema, rescuing vision in patients with neovascular age-related macular degeneration (nAMD). However, poor response and resistance to anti-VEGF treatment occurs. We report that targeting the Notch ligand Jagged1 by a monoclonal antibody reduces neovascular lesion size, number of activated phagocytes and inflammatory markers and vascular leakage in an experimental CNV mouse model. Additionally, we demonstrate that Jagged1 is expressed in mouse and human eyes, and that Jagged1 expression is independent of VEGF signaling in human endothelial cells. When anti-Jagged1 was combined with anti-VEGF in mice, the decrease in lesion size exceeded that of either antibody alone. The therapeutic effect was solely dependent on blocking, as engineering antibodies to abolish effector functions did not impair the therapeutic effect. Targeting of Jagged1 alone or in combination with anti-VEGF may thus be an attractive strategy to attenuate CNV-bearing diseases.
Collapse
Affiliation(s)
- Torleif Tollefsrud Gjølberg
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway
- Center of Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, 0450, Oslo, Norway
| | - Jonas Aakre Wik
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Department of Nutrition, Division of Molecular Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
| | - Hanna Johannessen
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Department of Pediatric Surgery, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Stig Krüger
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Nicola Bassi
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | | | - Malin Bern
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway
| | - Stian Foss
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway
| | - Goran Petrovski
- Center of Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, 0450, Oslo, Norway
| | - Morten C Moe
- Center of Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, 0450, Oslo, Norway
| | - Guttorm Haraldsen
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Johanna Hol Fosse
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Bjørn Steen Skålhegg
- Department of Nutrition, Division of Molecular Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway.
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway.
| | - Eirik Sundlisæter
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway.
| |
Collapse
|
19
|
Goldberg BS, Spencer DA, Pandey S, Ordonez T, Barnette P, Yu Y, Gao L, Dufloo J, Bruel T, Schwartz O, Ackerman ME, Hessell AJ. Complement contributes to antibody-mediated protection against repeated SHIV challenge. Proc Natl Acad Sci U S A 2023; 120:e2221247120. [PMID: 37155897 PMCID: PMC10193994 DOI: 10.1073/pnas.2221247120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
The first clinical efficacy trials of a broadly neutralizing antibody (bNAb) resulted in less benefit than expected and suggested that improvements are needed to prevent HIV infection. While considerable effort has focused on optimizing neutralization breadth and potency, it remains unclear whether augmenting the effector functions elicited by broadly neutralizing antibodies (bNAbs) may also improve their clinical potential. Among these effector functions, complement-mediated activities, which can culminate in the lysis of virions or infected cells, have been the least well studied. Here, functionally modified variants of the second-generation bNAb 10-1074 with ablated and enhanced complement activation profiles were used to examine the role of complement-associated effector functions. When administered prophylactically against simian-HIV challenge in rhesus macaques, more bNAb was required to prevent plasma viremia when complement activity was eliminated. Conversely, less bNAb was required to protect animals from plasma viremia when complement activity was enhanced. These results suggest that complement-mediated effector functions contribute to in vivo antiviral activity, and that their engineering may contribute to the further improvements in the efficacy of antibody-mediated prevention strategies.
Collapse
Affiliation(s)
| | - David A. Spencer
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| | - Shilpi Pandey
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| | - Tracy Ordonez
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| | - Philip Barnette
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| | - Yun Yu
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health and Science University, Portland, OR97239
- Biostatistics & Bioinformatics Core, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR97006
| | - Lina Gao
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health and Science University, Portland, OR97239
- Biostatistics & Bioinformatics Core, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR97006
| | - Jérémy Dufloo
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015Paris, France
- Université de Paris, École doctorale BioSPC 562, 75013Paris, France
| | - Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015Paris, France
- Vaccine Research Institute, 94000Créteil, France
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015Paris, France
- Vaccine Research Institute, 94000Créteil, France
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH03755
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH03755
| | - Ann J. Hessell
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| |
Collapse
|
20
|
Esawi E, Mahmoud IS, Abdullah MS, Abuarqoub DA, Ahram MA, Alshaer WM. 1,4-Naphthoquinone Induces FcRn Protein Expression and Albumin Recycling in Human THP-1 Cells. ACS OMEGA 2023; 8:16491-16499. [PMID: 37179634 PMCID: PMC10173444 DOI: 10.1021/acsomega.3c01678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/31/2023] [Indexed: 05/15/2023]
Abstract
The neonatal Fc receptor (FcRn) has been established as a major factor in regulating the metabolism of albumin and IgG in humans by protecting them from intracellular degradation after they are endocytosed into cells. We assume that increasing the levels of endogenous FcRn proteins in cells would be beneficial to enhance the recycling of these molecules. In this study, we identify the compound 1,4-naphthoquinone as an efficient stimulator of FcRn protein expression in human THP-1 monocytic cells with potency at the submicromolar range. Also, the compound increased the subcellular localization of FcRn to the endocytic recycling compartment and enhanced human serum albumin recycling in the PMA-induced THP-1 cells. These results suggest that 1,4-naphthoquinone stimulates FcRn expression and activity in human monocytic cells in vitro and it could open a new avenue for designing cotreatment agents to enhance the efficacy of biological treatments such as albumin-conjugated drugs in vivo.
Collapse
Affiliation(s)
- Ezaldeen
Ismael Esawi
- Department
of Pathology and Laboratory Medicine, King
Hussein Cancer Centre, Amman 11941, Jordan
| | - Ismail Sami Mahmoud
- Department
of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa 13133, Jordan
- . Tel: 00962797545880
| | | | - Duaa Azmi Abuarqoub
- Cell
Therapy Centre, The University of Jordan, Amman 11942, Jordan
- Department
of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical
Sciences, University of Petra, Amman 11180, Jordan
| | - Mamoun Ahmad Ahram
- Department
of Physiology and Biochemistry, The University
of Jordan, Amman 11942, Jordan
| | | |
Collapse
|
21
|
Nasir G, Sinnis P. Transport of antibody into the skin is only partially dependent upon the neonatal Fc-receptor. PLoS One 2023; 18:e0273960. [PMID: 37093800 PMCID: PMC10124839 DOI: 10.1371/journal.pone.0273960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
The dermis is the portal of entry for most vector-transmitted pathogens, making the host's immune response at this site critical in mitigating the magnitude of infection. For malaria, antibody-mediated neutralization of Plasmodium parasites in the dermis was recently demonstrated. However, surprisingly little is known about the mechanisms that govern antibody transport into the skin. Since the neonatal Fc receptor (FcRn) has been shown to transcytose IgG into various tissues, we sought to understand its contribution to IgG transport into the skin and antibody-mediated inhibition of Plasmodium parasites following mosquito bite inoculation. Using confocal imaging, we show that the transport of an anti-Langerin mAb into the skin occurs but is only partially reduced in mice lacking FcRn. To understand the relevance of FcRn in the context of malaria infection, we use the rodent parasite Plasmodium berghei and show that passively-administered anti-malarial antibody in FcRn deficient mice, does not reduce parasite burden to the same extent as previously observed in wildtype mice. Overall, our data suggest that FcRn plays a role in the transport of IgG into the skin but is not the major driver of IgG transport into this tissue. These findings have implications for the rational design of antibody-based therapeutics for malaria as well as other vector-transmitted pathogens.
Collapse
Affiliation(s)
- Gibran Nasir
- Johns Hopkins Malaria Institute and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Photini Sinnis
- Johns Hopkins Malaria Institute and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
22
|
Rühle J, Ginzel M, Dietz S, Schwarz J, Lajqi T, Beer-Hammer S, Poets CF, Gille C, Köstlin-Gille N. Depletion of Ly6G-Expressing Neutrophilic Cells Leads to Altered Peripheral T-Cell Homeostasis and Thymic Development in Neonatal Mice. Int J Mol Sci 2023; 24:7763. [PMID: 37175470 PMCID: PMC10178674 DOI: 10.3390/ijms24097763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Newborns and especially preterm infants are much more susceptible to infections than adults. Due to immature adaptive immunity, especially innate immune cells play an important role in a newborn's infection defense. Neonatal neutrophils exhibit profound differences in their functionality compared to neutrophils of adults. In particular, neonates possess a relevant population of suppressive neutrophils, which not only inhibit but also specifically modulate the function of T-cells. In this study, we investigated whether neonatal neutrophils are already involved in T-cell development in the thymus. For this purpose, we used a newly developed model of antibody-mediated immune cell depletion in which we administered a depleting antibody to pregnant and then lactating dams. Using this method, we were able to sufficiently deplete Ly6G-positive neutrophils in offspring. We demonstrated that the depletion of neutrophils in newborn mice resulted in altered peripheral T-cell homeostasis with a decreased CD4+/CD8+ T-cell ratio and decreased expression of CD62L. Neutrophil depletion even affected T-cell development in the thymus, with increased double positive thymocytes and a decreased CD4+/CD8+ single positive thymocyte ratio. Altogether, we demonstrated a previously unknown mechanism mediating neutrophils' immunomodulatory effects in newborns.
Collapse
Affiliation(s)
- Jessica Rühle
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Marco Ginzel
- Department of Pediatric and Adolescent Surgery, Paracelsus Medical University Hospital, A-5020 Salzburg, Austria;
| | - Stefanie Dietz
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Julian Schwarz
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomic and ICePhA, University of Tübingen, D-72074 Tübingen, Germany;
| | - Christian F. Poets
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Natascha Köstlin-Gille
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| |
Collapse
|
23
|
Conner CM, van Fossan D, Read K, Cowley DO, Alvarez O, Xu SXR, Webb DR, Jarnagin K. A precisely humanized FCRN transgenic mouse for preclinical pharmacokinetics studies. Biochem Pharmacol 2023; 210:115470. [PMID: 36870576 DOI: 10.1016/j.bcp.2023.115470] [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: 11/14/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023]
Abstract
Monoclonal antibodies (mAbs) are one of the fastest-growing classes of drugs and have been approved to treat several diseases, including cancers and autoimmune disorders. Preclinical pharmacokinetics studies are performed to determine the therapeutically meaningful dosages and efficacy of candidate drugs. These studies are typically performed in non-human primates; however, using primates is costly and raises ethical considerations. As a result, rodent models that better mimic human-like pharmacokinetics have been generated and remain an area of active investigation. Pharmacokinetic characteristics of a candidate drug, such as half-life, are partly controlled by antibody binding to the human neonatal receptor hFCRN. Due to the abnormally high binding of human antibodies to mouse FCRN, traditional laboratory rodents do not accurately model the pharmacokinetics of human mAbs. In response, humanized rodents expressing hFCRN have been generated. However, these models generally use large inserts randomly integrated into the mouse genome. Here, we report the production and characterization of a CRISPR/Cas9-mediated hFCRN transgenic mouse termed SYNB-hFCRN. Using CRISPR/Cas9-assisted gene targeting, we prepared a strain with a simultaneous knockout of mFcrn and insertion of a hFCRN mini-gene under the control of the endogenous mouse promoter. These mice are healthy and express hFCRN in the appropriate tissues and immune cell subtypes. Pharmacokinetic evaluation of human IgG and adalimumab (Humira®) demonstrate hFCRN-mediated protection. These newly generated SYNB-hFCRN mice provide another valuable animal model for use in preclinical pharmacokinetics studies during early drug development.
Collapse
Affiliation(s)
| | - Don van Fossan
- Synbal, Inc. 1759 Yorktown Rd., San Mateo, CA 94402, USA
| | - Kristen Read
- Synbal, Inc. 1759 Yorktown Rd., San Mateo, CA 94402, USA
| | - Dale O Cowley
- TransViragen, Inc., PO Box 110301, Research Triangle Park, NC 27709, USA
| | - Oscar Alvarez
- Synbal, Inc. 1759 Yorktown Rd., San Mateo, CA 94402, USA
| | | | - David R Webb
- Synbal, Inc. 1759 Yorktown Rd., San Mateo, CA 94402, USA
| | - Kurt Jarnagin
- Synbal, Inc. 1759 Yorktown Rd., San Mateo, CA 94402, USA.
| |
Collapse
|
24
|
Kasse CM, Yu AC, Powell AE, Roth GA, Liong CS, Jons CK, Buahin A, Maikawa CL, Zhou X, Youssef S, Glanville JE, Appel EA. Subcutaneous delivery of an antibody against SARS-CoV-2 from a supramolecular hydrogel depot. Biomater Sci 2023; 11:2065-2079. [PMID: 36723072 PMCID: PMC10012178 DOI: 10.1039/d2bm00819j] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/21/2022] [Indexed: 01/31/2023]
Abstract
Prolonged maintenance of therapeutically-relevant levels of broadly neutralizing antibodies (bnAbs) is necessary to enable passive immunization against infectious disease. Unfortunately, protection only lasts for as long as these bnAbs remain present at a sufficiently high concentration in the body. Poor pharmacokinetics and burdensome administration are two challenges that need to be addressed in order to make pre- and post-exposure prophylaxis with bnAbs feasible and effective. In this work, we develop a supramolecular hydrogel as an injectable, subcutaneous depot to encapsulate and deliver antibody drug cargo. This polymer-nanoparticle (PNP) hydrogel exhibits shear-thinning and self-healing properties that are required for an injectable drug delivery vehicle. In vitro drug release assays and diffusion measurements indicate that the PNP hydrogels prevent burst release and slow the release of encapsulated antibodies. Delivery of bnAbs against SARS-CoV-2 from PNP hydrogels is compared to standard routes of administration in a preclinical mouse model. We develop a multi-compartment model to understand the ability of these subcutaneous depot materials to modulate the pharmacokinetics of released antibodies; the model is extrapolated to explore the requirements needed for novel materials to successfully deliver relevant antibody therapeutics with different pharmacokinetic characteristics.
Collapse
Affiliation(s)
- Catherine M Kasse
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA.
| | - Anthony C Yu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA.
| | - Abigail E Powell
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Gillie A Roth
- Department of Bioengineering, Stanford University, Stanford, CA, 94305 USA
| | - Celine S Liong
- Department of Bioengineering, Stanford University, Stanford, CA, 94305 USA
| | - Carolyn K Jons
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA.
| | - Awua Buahin
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA.
| | - Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA, 94305 USA
| | - Xueting Zhou
- Department of Bioengineering, Stanford University, Stanford, CA, 94305 USA
| | - Sawsan Youssef
- Centivax Inc., 329 Oyster Point Drive, 3rd Floor South San Francisco, CA 94080, USA
| | - Jacob E Glanville
- Centivax Inc., 329 Oyster Point Drive, 3rd Floor South San Francisco, CA 94080, USA
| | - Eric A Appel
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA.
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA, 94305 USA
- Institute for Immunity, Transplantation, & Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pediatrics - Endocrinology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Woods Institute for the Environment, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
25
|
Sethi S, Jordan SC. Novel therapies for treatment of antibody-mediated rejection of the kidney. Curr Opin Organ Transplant 2023; 28:29-35. [PMID: 36579683 DOI: 10.1097/mot.0000000000001037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW We aim to discuss current literature on novel therapies for antibody-mediated rejection (AMR) in kidney transplantation with a focus on chronic AMR. RECENT FINDINGS IL-6/IL-6 receptor blockers appear promising in the treatment of chronic AMR. Blocking this pathway was shown to reduce human leucocyte antigen-antibodies, improve histologic inflammation and increase T-regulatory cells. Based on experience in desensitization, IgG degrading endopeptidase, imlifidase, could be effective in AMR. There have been case reports describing the successful use of plasma cell/natural killer-cell-directed anti-CD38 antibody in the treatment of AMR. Off-target effects have been noted and strategies to mitigate these will be needed when using these agents. Complement inhibitors could be an effective add-on strategy to antibody-depleting therapies but their role in AMR needs to be better defined. Combining proteasome inhibitors and costimulation blockers has shown encouraging results in the prevention of AMR in animal models and is now being investigated in humans. Other novel strategies such as Fc neonatal receptor blockers which inhibit the recycling of pathogenic IgG and bispecific antibodies against B-cell maturation antigen/CD3+ T cells warrant further investigation. SUMMARY There are now a number of emerging therapies with varied targets and mechanism(s) of action that hold promise in the management of AMR and improving allograft survival.
Collapse
Affiliation(s)
- Supreet Sethi
- Division of Nephrology, Department of Medicine, Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, California, USA
| | | |
Collapse
|
26
|
Klich JH, Kasse CM, Mann JL, Huang Y, d’Aquino AI, Grosskopf AK, Baillet J, Fuller GG, Appel EA. Stable High-Concentration Monoclonal Antibody Formulations Enabled by an Amphiphilic Copolymer Excipient. ADVANCED THERAPEUTICS 2023; 6:2200102. [PMID: 36684707 PMCID: PMC9854243 DOI: 10.1002/adtp.202200102] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Monoclonal antibodies are a staple in modern pharmacotherapy. Unfortunately, these biopharmaceuticals are limited by their tendency to aggregate in formulation, resulting in poor stability and often requiring low concentration drug formulations. Moreover, existing excipients designed to stabilize these formulations are often limited by their toxicity and tendency to form particles such as micelles. Here, we demonstrate the ability of a simple "drop-in", amphiphilic copolymer excipient to enhance the stability of high concentration formulations of clinically-relevant monoclonal antibodies without altering their pharmacokinetics or injectability. Through interfacial rheology and surface tension measurements, we demonstrate that the copolymer excipient competitively adsorbs to formulation interfaces. Further, through determination of monomeric composition and retained bioactivity through stressed aging, we show that this excipient confers a significant stability benefit to high concentration antibody formulations. Finally, we demonstrate that the excipient behaves as an inactive ingredient, having no significant impact on the pharmacokinetic profile of a clinically relevant antibody in mice. This amphiphilic copolymer excipient demonstrates promise as a simple formulation additive to create stable, high concentration antibody formulations, thereby enabling improved treatment options such as a route-of-administration switch from low concentration intravenous (IV) to high concentration subcutaneous (SC) delivery while reducing dependence on the cold chain.
Collapse
Affiliation(s)
| | | | - Joseph L. Mann
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Yaoqi Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Andrea I. d’Aquino
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Abigail K. Grosskopf
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Julie Baillet
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Gerald G. Fuller
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA; Department of Pediatrics – Endocrinology, Stanford University School of Medicine, Stanford, CA 94305, USA; ChEM-H Institute, Stanford University, Stanford, CA 94305, USA; Woods Institute for the Environment, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
27
|
Aweda TA, Cheng SH, Lenhard SC, Sepp A, Skedzielewski T, Hsu CY, Marshall S, Haag H, Kehler J, Jagdale P, Peter A, Schmid MA, Gehman A, Doan M, Mayer AP, Gorycki P, Fanget M, Colas C, Smith B, Maier CC, Alsaid H. In vivo biodistribution and pharmacokinetics of sotrovimab, a SARS-CoV-2 monoclonal antibody, in healthy cynomolgus monkeys. Eur J Nucl Med Mol Imaging 2023; 50:667-678. [PMID: 36305907 PMCID: PMC9614201 DOI: 10.1007/s00259-022-06012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/16/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Sotrovimab (VIR-7831), a human IgG1κ monoclonal antibody (mAb), binds to a conserved epitope on the SARS-CoV-2 spike protein receptor binding domain (RBD). The Fc region of VIR-7831 contains an LS modification to promote neonatal Fc receptor (FcRn)-mediated recycling and extend its serum half-life. Here, we aimed to evaluate the impact of the LS modification on tissue biodistribution, by comparing VIR-7831 to its non-LS-modified equivalent, VIR-7831-WT, in cynomolgus monkeys. METHODS 89Zr-based PET/CT imaging of VIR-7831 and VIR-7831-WT was performed up to 14 days post injection. All major organs were analyzed for absolute concentration as well as tissue:blood ratios, with the focus on the respiratory tract, and a physiologically based pharmacokinetics (PBPK) model was used to evaluate the tissue biodistribution kinetics. Radiomics features were also extracted from the PET images and SUV values. RESULTS SUVmean uptake in the pulmonary bronchi for 89Zr-VIR-7831 was statistically higher than for 89Zr-VIR-7831-WT at days 6 (3.43 ± 0.55 and 2.59 ± 0.38, respectively) and 10 (2.66 ± 0.32 and 2.15 ± 0.18, respectively), while the reverse was observed in the liver at days 6 (5.14 ± 0.80 and 8.63 ± 0.89, respectively), 10 (4.52 ± 0.59 and 7.73 ± 0.66, respectively), and 14 (4.95 ± 0.65 and 7.94 ± 0.54, respectively). Though the calculated terminal half-life was 21.3 ± 3.0 days for VIR-7831 and 16.5 ± 1.1 days for VIR-7831-WT, no consistent differences were observed in the tissue:blood ratios between the antibodies except in the liver. While the lung:blood SUVmean uptake ratio for both mAbs was 0.25 on day 3, the PBPK model predicted the total lung tissue and the interstitial space to serum ratio to be 0.31 and 0.55, respectively. Radiomics analysis showed VIR-7831 had mean-centralized PET SUV distribution in the lung and liver, indicating more uniform uptake than VIR-7831-WT. CONCLUSION The half-life extended VIR-7831 remained in circulation longer than VIR-7831-WT, consistent with enhanced FcRn binding, while the tissue:blood concentration ratios in most tissues for both drugs remained statistically indistinguishable throughout the course of the experiment. In the bronchiolar region, a higher concentration of 89Zr-VIR-7831 was detected. The data also allow unparalleled insight into tissue distribution and elimination kinetics of mAbs that can guide future biologic drug discovery efforts, while the residualizing nature of the 89Zr label sheds light on the sites of antibody catabolism.
Collapse
Affiliation(s)
- Tolulope A Aweda
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Shih-Hsun Cheng
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Stephen C Lenhard
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | | | | | - Chih-Yang Hsu
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Shelly Marshall
- Integrated Biological Platform Sciences, GSK, Collegeville, PA, USA
| | - Heather Haag
- Integrated Biological Platform Sciences, GSK, Collegeville, PA, USA
| | - Jonathan Kehler
- Bioanalysis, Immunogenicity & Biomarkers, GSK, Collegeville, PA, USA
| | | | - Alessia Peter
- mAb Engineering & Bioanalytics, Humabs BioMed SA, Vir Biotechnology, Inc, Bellinzona, Switzerland
| | - Michael A Schmid
- mAb Engineering & Bioanalytics, Humabs BioMed SA, Vir Biotechnology, Inc, Bellinzona, Switzerland
| | - Andrew Gehman
- Non-Clinical and Translational Statistics, GSK, Collegeville, PA, USA
| | - Minh Doan
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Andrew P Mayer
- Bioanalysis, Immunogenicity & Biomarkers, GSK, Collegeville, PA, USA
| | | | - Marie Fanget
- Bioanalytical Department, Vir Biotechnology, Inc, San Francisco, CA, USA
| | | | - Brenda Smith
- Toxicology, Vir Biotechnology, Inc, San Francisco, CA, USA
| | | | - Hasan Alsaid
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA.
| |
Collapse
|
28
|
Fernández-Quintero ML, Ljungars A, Waibl F, Greiff V, Andersen JT, Gjølberg TT, Jenkins TP, Voldborg BG, Grav LM, Kumar S, Georges G, Kettenberger H, Liedl KR, Tessier PM, McCafferty J, Laustsen AH. Assessing developability early in the discovery process for novel biologics. MAbs 2023; 15:2171248. [PMID: 36823021 PMCID: PMC9980699 DOI: 10.1080/19420862.2023.2171248] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/18/2023] [Indexed: 02/25/2023] Open
Abstract
Beyond potency, a good developability profile is a key attribute of a biological drug. Selecting and screening for such attributes early in the drug development process can save resources and avoid costly late-stage failures. Here, we review some of the most important developability properties that can be assessed early on for biologics. These include the influence of the source of the biologic, its biophysical and pharmacokinetic properties, and how well it can be expressed recombinantly. We furthermore present in silico, in vitro, and in vivo methods and techniques that can be exploited at different stages of the discovery process to identify molecules with liabilities and thereby facilitate the selection of the most optimal drug leads. Finally, we reflect on the most relevant developability parameters for injectable versus orally delivered biologics and provide an outlook toward what general trends are expected to rise in the development of biologics.
Collapse
Affiliation(s)
- Monica L. Fernández-Quintero
- Center for Molecular Biosciences Innsbruck (CMBI), Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Franz Waibl
- Center for Molecular Biosciences Innsbruck (CMBI), Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Victor Greiff
- Department of Immunology, University of Oslo, Oslo, Norway
| | - Jan Terje Andersen
- Department of Immunology, University of Oslo, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo, Oslo, Norway
| | | | - Timothy P. Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Bjørn Gunnar Voldborg
- National Biologics Facility, Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lise Marie Grav
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Sandeep Kumar
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Guy Georges
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Hubert Kettenberger
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Klaus R. Liedl
- Center for Molecular Biosciences Innsbruck (CMBI), Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Peter M. Tessier
- Department of Chemical Engineering, Pharmaceutical Sciences and Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - John McCafferty
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Maxion Therapeutics, Babraham Research Campus, Cambridge, UK
| | - Andreas H. Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
29
|
Biophysical differences in IgG1 Fc-based therapeutics relate to their cellular handling, interaction with FcRn and plasma half-life. Commun Biol 2022; 5:832. [PMID: 35982144 PMCID: PMC9388496 DOI: 10.1038/s42003-022-03787-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/01/2022] [Indexed: 01/07/2023] Open
Abstract
Antibody-based therapeutics (ABTs) are used to treat a range of diseases. Most ABTs are either full-length IgG1 antibodies or fusions between for instance antigen (Ag)-binding receptor domains and the IgG1 Fc fragment. Interestingly, their plasma half-life varies considerably, which may relate to how they engage the neonatal Fc receptor (FcRn). As such, there is a need for an in-depth understanding of how different features of ABTs affect FcRn-binding and transport behavior. Here, we report on how FcRn-engagement of the IgG1 Fc fragment compare to clinically relevant IgGs and receptor domain Fc fusions, binding to VEGF or TNF-α. The results reveal FcRn-dependent intracellular accumulation of the Fc, which is in line with shorter plasma half-life than that of full-length IgG1 in human FcRn-expressing mice. Receptor domain fusion to the Fc increases its half-life, but not to the extent of IgG1. This is mirrored by a reduced cellular recycling capacity of the Fc-fusions. In addition, binding of cognate Ag to ABTs show that complexes of similar size undergo cellular transport at different rates, which could be explained by the biophysical properties of each ABT. Thus, the study provides knowledge that should guide tailoring of ABTs regarding optimal cellular sorting and plasma half-life. Analysis of clinically approved antibody-based therapeutics reveals different structural designs, such as full-length IgG1 or Fc-fusions, entail distinct biophysical properties that affect FcRn binding, intracellular transport and plasma half-life.
Collapse
|
30
|
Sand KMK, Gruber MM, Sandlie I, Mathiesen L, Andersen JT, Wadsack C. Contribution of the ex vivo placental perfusion model in understanding transplacental immunoglobulin G transfer. Placenta 2022; 127:77-87. [PMID: 35981406 DOI: 10.1016/j.placenta.2022.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/07/2022] [Accepted: 07/28/2022] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The acquisition of humoral immunity in utero is essential for the fetus. The crucial protein, which is responsible for this part of immunity, is immunoglobulin-G (IgG). Immune functions of IgGs are mediated via the interaction of the crystallizable fragment (Fc) region of IgG with specific Fc γ receptors (FcγRs). However, an atypical FcγR, the neonatal Fc receptor (FcRn), is a key regulator of IgG transfer across the human placenta. During the last four decades ex vivo placental perfusion studies have contributed significantly to the study of mechanisms of IgG transfer across the multicellular placental barrier. METHOD A PubMed search was conducted by using specific keywords: placenta, perfusion and IgG to review manuscripts using human placental perfusion to study the transplacental transfer of IgG. Relevant studies found in reference lists of these manuscripts were also added to the review, and references were included that supported or gave nuance to the discussion of the mechanisms of IgG kinetics in the placenta. RESULTS AND DISCUSSION We found twenty publications on the study of transplacental transfer of IgG using human ex vivo placental perfusion, by research groups with partly different settings. This review summarizes knowledge about placental IgG transfer, with a strong focus on the contributions from ex vivo placental perfusion studies.
Collapse
Affiliation(s)
- Kine Marita Knudsen Sand
- Department of Biosciences, University of Oslo, 0371, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, 0424, Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, 0424, Oslo, Norway
| | - Michael M Gruber
- Department of Obstetrics and Gynaecology, Medical University of Graz, 8036, Graz, Austria
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, 0371, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, 0424, Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, 0424, Oslo, Norway
| | - Line Mathiesen
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark.
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, 0424, Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, 0424, Oslo, Norway
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz, 8036, Graz, Austria; BioTechMed-Graz, Austria
| |
Collapse
|
31
|
Kasse CM, Yu AC, Powell AE, Roth GA, Liong CS, Jons CK, Buahin A, Maikawa CL, Youssef S, Glanville JE, Appel EA. Subcutaneous delivery of an antibody against SARS-CoV-2 from a supramolecular hydrogel depot. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.05.24.493347. [PMID: 35665002 PMCID: PMC9164446 DOI: 10.1101/2022.05.24.493347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Prolonged maintenance of therapeutically-relevant levels of broadly neutralizing antibodies (bnAbs) is necessary to enable passive immunization against infectious disease. Unfortunately, protection only lasts for as long as these bnAbs remain present at a sufficiently high concentration in the body. Poor pharmacokinetics and burdensome administration are two challenges that need to be addressed in order to make pre- and post-exposure prophylaxis with bnAbs feasible and effective. In this work, we develop a supramolecular hydrogel as an injectable, subcutaneous depot to encapsulate and deliver antibody drug cargo. This polymer-nanoparticle (PNP) hydrogel exhibits shear-thinning and self-healing properties that are required for an injectable drug delivery vehicle. In vitro drug release assays and diffusion measurements indicate that the PNP hydrogels prevent burst release and slow the release of encapsulated antibodies. Delivery of bnAbs against SARS-CoV-2 from PNP hydrogels is compared to standard routes of administration in a preclinical mouse model. We develop a multi-compartment model to understand the ability of these subcutaneous depot materials to modulate the pharmacokinetics of released antibodies; the model is extrapolated to explore the requirements needed for novel materials to successfully deliver relevant antibody therapeutics with different pharmacokinetic characteristics.
Collapse
|
32
|
Novel treatment strategies for acetylcholine receptor antibody-positive myasthenia gravis and related disorders. Autoimmun Rev 2022; 21:103104. [PMID: 35452851 DOI: 10.1016/j.autrev.2022.103104] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/18/2022] [Indexed: 11/21/2022]
Abstract
The presence of autoantibodies directed against the muscle nicotinic acetylcholine receptor (AChR) is the most common cause of myasthenia gravis (MG). These antibodies damage the postsynaptic membrane of the neuromuscular junction and cause muscle weakness by depleting AChRs and thus impairing synaptic transmission. As one of the best-characterized antibody-mediated autoimmune diseases, AChR-MG has often served as a reference model for other autoimmune disorders. Classical pharmacological treatments, including broad-spectrum immunosuppressive drugs, are effective in many patients. However, complete remission cannot be achieved in all patients, and 10% of patients do not respond to currently used therapies. This may be attributed to production of autoantibodies by long-lived plasma cells which are resistant to conventional immunosuppressive drugs. Hence, novel therapies specifically targeting plasma cells might be a suitable therapeutic approach for selected patients. Additionally, in order to reduce side effects of broad-spectrum immunosuppression, targeted immunotherapies and symptomatic treatments will be required. This review presents established therapies as well as novel therapeutic approaches for MG and related conditions, with a focus on AChR-MG.
Collapse
|
33
|
Duong SL, Prüss H. Paraneoplastic Autoimmune Neurological Syndromes and the Role of Immune Checkpoint Inhibitors. Neurotherapeutics 2022; 19:848-863. [PMID: 35043373 PMCID: PMC9294109 DOI: 10.1007/s13311-022-01184-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 12/14/2022] Open
Abstract
The introduction of immune checkpoint inhibitors (ICIs) in oncologic therapies has led to a paradigm shift in cancer treatment. ICIs have increased the overall survival in patients with malignant melanoma, small-cell lung cancer, and many other tumor entities. Despite their clinical benefits, these novel cancer immunotherapies can induce neurological immune-related adverse events (irAEs). Such immune-mediated complications can manifest within the spectrum of paraneoplastic neurological syndromes (PNSs). PNSs are rare immune-mediated complications of systemic cancers that can involve every aspect of the nervous system. The emergence of PNSs with ICI treatment opens further pathways to study the complex immunopathological interplay of cancer immunity, cross-reactive neurological autoimmune phenomena, and effects of ICIs on the immune system. ICI-induced PNSs comprise a diverse antibody repertoire and phenotypic spectrum with severe and life-threatening disease progression in some cases. Timely diagnosis and urgent interventions are pivotal for a favorable neurologic and oncologic outcome. This review focuses on the pathogenesis of cancer immunotherapy and the disruption of immune tolerance in PNSs and provides an overview of the most pertinent clinical manifestations and principles of diagnostic and therapeutic managements in light of the expected increase in PNSs due to the widespread use of ICIs in clinical practice. This review further discusses potential and evolving concepts of therapeutic monoclonal antibodies for the treatment of PNSs.
Collapse
Affiliation(s)
- Sophie L Duong
- Department of Neurology and Experimental Neurology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany.
| |
Collapse
|
34
|
Zakrzewicz A, Würth C, Beckert B, Feldhoff S, Vanderheyden K, Foss S, Andersen JT, de Haard H, Verheesen P, Bobkov V, Tikkanen R. Stabilization of Keratinocyte Monolayer Integrity in the Presence of Anti-Desmoglein-3 Antibodies through FcRn Blockade with Efgartigimod: Novel Treatment Paradigm for Pemphigus? Cells 2022; 11:cells11060942. [PMID: 35326398 PMCID: PMC8946243 DOI: 10.3390/cells11060942] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 12/21/2022] Open
Abstract
Pemphigus vulgaris is an autoimmune blistering disease of the epidermis, caused by autoantibodies against desmosomal proteins, mainly desmogleins 1 and 3, which induce an impairment of desmosomal adhesion and blister formation. Recent findings have shown that inhibition of immunoglobulin G binding on the neonatal Fc receptor, FcRn, results in reduced autoantibody recycling and shortens their half-life, providing a valid treatment option for PV. We have here analyzed the role of FcRn in human keratinocytes treated with antibodies isolated from pemphigus vulgaris patient or with recombinant anti-desmoglein-3 antibodies that induce pathogenic changes in desmosomes, such as loss of monolayer integrity, aberrant desmoglein-3 localization and degradation of desmoglein-3. We show that blocking IgG binding on FcRn by efgartigimod, a recombinant Fc fragment undergoing clinical studies for pemphigus, stabilizes the keratinocyte monolayer, whereas the loss of desmoglein-3 is not prevented by efgartigimod. Our data show that FcRn may play a direct role in the pathogenesis of pemphigus at the level of the autoantibody target cells, the epidermal keratinocytes. Our data suggest that in keratinocytes, FcRn may have functions different from its known function in IgG recycling. Therefore, stabilization of keratinocyte adhesion by FcRn blocking entities may provide a novel treatment paradigm for pemphigus.
Collapse
Affiliation(s)
- Anna Zakrzewicz
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany; (A.Z.); (C.W.); (B.B.); (S.F.)
| | - Celina Würth
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany; (A.Z.); (C.W.); (B.B.); (S.F.)
| | - Benedikt Beckert
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany; (A.Z.); (C.W.); (B.B.); (S.F.)
| | - Simon Feldhoff
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany; (A.Z.); (C.W.); (B.B.); (S.F.)
| | - Katrien Vanderheyden
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (K.V.); (H.d.H.); (P.V.); (V.B.)
| | - Stian Foss
- Department of Immunology, University of Oslo and Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway; (S.F.); (J.T.A.)
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372 Oslo, Norway
| | - Jan Terje Andersen
- Department of Immunology, University of Oslo and Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway; (S.F.); (J.T.A.)
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, 0372 Oslo, Norway
| | - Hans de Haard
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (K.V.); (H.d.H.); (P.V.); (V.B.)
| | - Peter Verheesen
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (K.V.); (H.d.H.); (P.V.); (V.B.)
| | - Vladimir Bobkov
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (K.V.); (H.d.H.); (P.V.); (V.B.)
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany; (A.Z.); (C.W.); (B.B.); (S.F.)
- Correspondence:
| |
Collapse
|
35
|
Vázquez-Domínguez I, Garanto A. Considerations for Generating Humanized Mouse Models to Test Efficacy of Antisense Oligonucleotides. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2434:267-279. [PMID: 35213024 DOI: 10.1007/978-1-0716-2010-6_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Over the last decades, animal models have become increasingly important in therapeutic drug development and assessment. The use of these models, mainly mice and rats, allow evaluating drugs in the real-organism environment and context. However, several molecular therapeutic approaches are sequence-dependent, and therefore, the humanization of such models is required to assess the efficacy. The generation of genetically modified humanized mouse models is often an expensive and laborious process that may not always recapitulate the human molecular and/or physiological phenotype. In this chapter, we summarize basic aspects to consider before designing and generating humanized models, especially when they are aimed to test antisense-based therapies.
Collapse
Affiliation(s)
- Irene Vázquez-Domínguez
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Alejandro Garanto
- Department of Pediatrics, Amalia Children's Hospital, Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
36
|
Kelly VW, Sirk SJ. Short FcRn-Binding Peptides Enable Salvage and Transcytosis of scFv Antibody Fragments. ACS Chem Biol 2022; 17:404-413. [PMID: 35050570 DOI: 10.1021/acschembio.1c00862] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Therapeutic antibodies have become one of the most widely used classes of biotherapeutics due to their unique antigen specificity and their ability to be engineered against diverse disease targets. There is significant interest in utilizing truncated antibody fragments as therapeutics, as their small size affords favorable properties such as increased tumor penetration as well as the ability to utilize lower-cost prokaryotic production methods. Their small size and simple architecture, however, also lead to rapid blood clearance, limiting the efficacy of these potentially powerful therapeutics. A common approach to circumvent these limitations is to enable engagement with the half-life extending neonatal Fc receptor (FcRn). This is usually achieved via fusion with a large Fc domain, which negates the benefits of the antibody fragment's small size. In this work, we show that modifying antibody fragments with short FcRn-binding peptide domains that mimic native IgG engagement with FcRn enables binding and FcRn-mediated recycling and transmembrane transcytosis in cell-based assays. Further, we show that rational, single amino acid mutations to the peptide sequence have a significant impact on the receptor-mediated function and investigate the underlying structural basis for this effect using computational modeling. Finally, we report the identification of a short peptide from human serum albumin that enables FcRn-mediated function when grafted onto a single-chain variable fragment (scFv) scaffold, establishing an approach for the rational selection of short-peptide domains from full-length proteins that could enable the transfer of non-native functions to small recombinant proteins without significantly impacting their size or structure.
Collapse
Affiliation(s)
- Vince W. Kelly
- Department of Bioengineering, University of Illinois, Urbana, Illinois 61801, United States
| | - Shannon J. Sirk
- Department of Bioengineering, University of Illinois, Urbana, Illinois 61801, United States
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois, Urbana, Illinois 61801, United States
| |
Collapse
|
37
|
Hori Y, Ohmine K, Katada H, Noguchi Y, Sato K, Nambu T, Adeline LR, Wan GS, Haraya K, Ozeki K, Nanami M, Tachibana T, Sampei Z, Kuramochi T, Nezu J, Hattori K, Igawa T. Elimination of plasma soluble antigen in cynomolgus monkeys by combining pH-dependent antigen binding and novel Fc engineering. MAbs 2022; 14:2068213. [PMID: 35482905 PMCID: PMC9067469 DOI: 10.1080/19420862.2022.2068213] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
A conventional antibody targeting a soluble antigen in circulation typically requires a huge dosage and frequent intravenous administration to neutralize the antigen. This is because antigen degradation is reduced by the formation of antigen–antibody immune complexes, which escape from lysosomal degradation using neonatal Fc receptor (FcRn)-mediated recycling. To address this, we developed an antigen-sweeping antibody that combines pH-dependent antigen binding and Fc engineering to enhance Fc receptor binding. The sweeping antibody actively eliminates the plasma antigens by increasing the cellular uptake of the immune complex and dissociating the antigens in the acidic endosome for degradation. Strong antigen sweeping can reduce the dosage, potentially achieve higher efficacy, and expand the scope of antigen space available for targeting by antibodies. In this study, to further improve the sweeping efficacy, we developed a novel antibody Fc variant by enhancing Fcγ receptor IIb (FcγRIIb) binding and modulating charge characteristics for increased cellular uptake of the immune complex, together with enhancing FcRn binding for efficient salvage of the antigen-free antibodies. Our Fc variant achieved strong antigen sweeping in cynomolgus monkeys with antibody pharmacokinetics comparable to a wild-type human IgG1 antibody. The positive-charge substitutions enhanced uptake of the immune complex by FcγRIIb-expressing cells in vitro, which was completely inhibited by an anti-FcγRIIb antibody. This suggests that the strong in vivo sweeping efficacy improved by the charge engineering is more likely achieved by FcγRIIb-dependent uptake of the immune complex rather than nonspecific uptake. We expect this novel Fc engineering can maximize the antigen sweeping efficacy even in humans and create novel therapeutic antibodies that meet unmet medical needs for patients.
Collapse
Affiliation(s)
- Yuji Hori
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Ken Ohmine
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | | | - Yuki Noguchi
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Kazuki Sato
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | | | | | | | - Kenta Haraya
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Kazuhisa Ozeki
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Masahiko Nanami
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | | | - Zenjiro Sampei
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Taichi Kuramochi
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Junichi Nezu
- Research Division, Chugai Pharmaceutical Co, Ltd, Chuo-ku, Tokyo, Japan
| | - Kunihiro Hattori
- Research Division, Chugai Pharmaceutical Co, Ltd, Kamakura, Japan
| | - Tomoyuki Igawa
- Research Division, Chugai Pharmaceutical Co, Ltd, Chuo-ku, Tokyo, Japan
| |
Collapse
|
38
|
Baumrucker CR, Macrina AL, Bruckmaier RM. Colostrogenesis: Role and Mechanism of the Bovine Fc Receptor of the Neonate (FcRn). J Mammary Gland Biol Neoplasia 2021; 26:419-453. [PMID: 35080749 DOI: 10.1007/s10911-021-09506-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/10/2021] [Indexed: 11/28/2022] Open
Abstract
Colostrogenesis is a separate and unique phase of mammary epithelial cell activity occurring in the weeks before parturition and rather abruptly ending after birth in the bovine. It has been the focus of research to define what controls this process and how it produces high concentrations of specific biologically active components important for the neonate. In this review we consider colostrum composition and focus upon components that appear in first milked colostrum in concentrations exceeding that in blood serum. The Fc Receptor of the Neonate (FcRn) is recognized as the major immunoglobulin G (IgG) and albumin binding protein that accounts for the proteins' long half-lives. We integrate the action of the pinocytotic (fluid phase) uptake of extracellular components and merge them with FcRn in sorting endosomes. We define and explore the means of binding, sorting, and the transcytotic delivery of IgG1 while recycling IgG2 and albumin. We consider the means of releasing the ligands from the receptor within the endosome and describe a new secretion mechanism of cargo release into colostrum without the appearance of FcRn itself in colostrum. We integrate the insulin-like growth factor family, some of which are highly concentrated bioactive components of colostrum, with the mechanisms related to FcRn endosome action. In addition to secretion, we highlight the recent findings of a role of the FcRn in phagocytosis and antigen presentation and relate its significant and abrupt change in cellular location after parturition to a role in the prevention and resistance to mastitis infections.
Collapse
Affiliation(s)
- Craig R Baumrucker
- Department of Animal Science, Penn State University, University Park, PA, 16802, USA.
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland.
| | - Ann L Macrina
- Department of Animal Science, Penn State University, University Park, PA, 16802, USA
| | - Rupert M Bruckmaier
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| |
Collapse
|
39
|
Wolfe GI, Ward ES, de Haard H, Ulrichts P, Mozaffar T, Pasnoor M, Vidarsson G. IgG regulation through FcRn blocking: A novel mechanism for the treatment of myasthenia gravis. J Neurol Sci 2021; 430:118074. [PMID: 34563918 DOI: 10.1016/j.jns.2021.118074] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022]
Abstract
The neonatal Fc receptor (FcRn) is an MHC class I-like molecule that is widely distributed in mammalian organs, tissues, and cells. FcRn is critical to maintaining immunoglobulin G (IgG) and albumin levels through rescuing these molecules from lysosomal degradation. IgG autoantibodies are associated with many autoimmune diseases, including myasthenia gravis (MG), a rare neuromuscular autoimmune disease that causes debilitating and, in its generalized form (gMG), potentially life-threatening muscle weakness. IgG autoantibodies are directly pathogenic in MG and target neuromuscular junction proteins, causing neuromuscular transmission failure. Treatment approaches that reduce autoantibody levels, such as therapeutic plasma exchange and intravenous immunoglobulin, have been shown to be effective for gMG patients but are not indicated as ongoing maintenance therapies and can be associated with burdensome side effects. Agents that block FcRn-mediated recycling of IgG represent a rational and promising approach for the treatment of gMG. Blocking FcRn allows targeted reduction of all IgG subtypes without decreasing concentrations of other Ig isotypes; therefore, FcRn blocking could be a safe and effective treatment strategy for a broad population of gMG patients. Several FcRn-blocking antibodies and one antibody Fc fragment have been developed and are currently in various stages of clinical development. This article describes the mechanism of FcRn blockade as a novel approach for IgG-mediated disease therapy and reviews promising clinical data using such FcRn blockers for the treatment of gMG.
Collapse
Affiliation(s)
- Gil I Wolfe
- Department of Neurology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, SUNY, Buffalo, NY, USA.
| | - E Sally Ward
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| | - Hans de Haard
- argenx, Zwijnaarde, Belgium, University of California, Irvine, CA, USA
| | - Peter Ulrichts
- argenx, Zwijnaarde, Belgium, University of California, Irvine, CA, USA
| | - Tahseen Mozaffar
- Department of Neurology, University of California, Irvine, CA, USA
| | - Mamatha Pasnoor
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gestur Vidarsson
- Sanquin Research, and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
40
|
García-Serra A, Radosevic M, Ríos J, Aguilar E, Maudes E, Landa J, Sabater L, Martinez-Hernandez E, Planagumà J, Dalmau J. Blocking Placental Class G Immunoglobulin Transfer Prevents NMDA Receptor Antibody Effects in Newborn Mice. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1061. [PMID: 34580181 PMCID: PMC8477376 DOI: 10.1212/nxi.0000000000001061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/15/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVES To determine in a mouse model whether neonatal Fc receptor (FcRn) blockade prevents the placental transfer of class G immunoglobulin (IgG) derived from patients with anti-NMDA receptor (NMDAR) encephalitis and their pathogenic effects on the fetuses and offspring. METHODS Pregnant C57BL/6J mice were administered via tail vein FcRn antibody (FcRn-ab) or saline solution 6 hours before administration of patients' or controls' IgG on days 14, 15, and 16 of gestation. Three experimental groups were established: mice receiving controls' IgG, patients' IgG, or patients' IgG along with pretreatment with FcRn-ab. Immunohistochemical staining, confocal microscopy, hippocampal long-term potentiation, and standardized developmental and behavioral tasks were used to assess the efficacy of treatment with FcRn-ab. RESULTS In pregnant mice that received patients' IgG, treatment with FcRn-ab prevented the IgG from reaching the fetal brain, abrogating the decrease of NMDAR clusters and the reduction of cortical plate thickness that were observed in fetuses from untreated pregnant mice. Moreover, among the offspring of mothers that received patients' IgG, those whose mothers were treated with FcRn-ab did not develop the alterations that occurred in offspring of untreated mothers, including impairment in hippocampal plasticity, delay in innate reflexes, and visuospatial memory deficits. DISCUSSION FcRn blockade prevents placental transfer of IgG from patients with anti-NMDAR encephalitis and abrogates the synaptic and neurodevelopmental alterations caused by patients' antibodies. This model has potential therapeutic implications for other antibody-mediated diseases of the CNS during pregnancy.
Collapse
Affiliation(s)
- Anna García-Serra
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Marija Radosevic
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - José Ríos
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Esther Aguilar
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Estibaliz Maudes
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Jon Landa
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Lidia Sabater
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Eugenia Martinez-Hernandez
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Jesús Planagumà
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Josep Dalmau
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain.
| |
Collapse
|
41
|
Macri C, Morgan H, Villadangos JA, Mintern JD. Regulation of dendritic cell function by Fc-γ-receptors and the neonatal Fc receptor. Mol Immunol 2021; 139:193-201. [PMID: 34560415 DOI: 10.1016/j.molimm.2021.07.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 06/28/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023]
Abstract
Dendritic cells (DCs) express receptors to sense pathogens and/or tissue damage and to communicate with other immune cells. Among those receptors, Fc receptors (FcRs) are triggered by the Fc region of antibodies produced during adaptive immunity. In this review, the role of FcγR and neonatal Fc receptor (FcRn) in DC immunity will be discussed. Their expression in DC subsets and impact on antigen uptake and presentation, DC maturation and polarisation of T cell responses will be described. Lastly, we will discuss the importance of FcR-mediated DC function in the context of immunity during viral infection, inflammatory disease, cancer and immunotherapy.
Collapse
Affiliation(s)
- Christophe Macri
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia
| | - Huw Morgan
- ACRF Translational Research Laboratory, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, 3050, Australia; Department of Medicine, University of Melbourne, Parkville, Melbourne, Victoria, 3010, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
42
|
Man F, Koers A, Karagiannis P, Josephs DH, Bax HJ, Gilbert AE, Dodev TS, Mele S, Chiarruttini G, Crescioli S, Chauhan J, Blower JE, Cooper MS, Spicer J, Karagiannis SN, Blower PJ. In vivo trafficking of a tumor-targeting IgE antibody: molecular imaging demonstrates rapid hepatobiliary clearance compared to IgG counterpart. Oncoimmunology 2021; 10:1966970. [PMID: 34513315 PMCID: PMC8425638 DOI: 10.1080/2162402x.2021.1966970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/26/2021] [Accepted: 08/08/2021] [Indexed: 11/30/2022] Open
Abstract
IgE antibodies elicit powerful immune responses, recruiting effector cells to tumors more efficiently and with greater cytotoxicity than IgG antibodies. Consequently, IgE antibodies are a promising alternative to conventional IgG-based therapies in oncology (AllergoOncology). As the pharmacokinetics of IgE antibodies are less well understood, we used molecular imaging in mice to compare the distribution and elimination of IgE and IgG antibodies targeting the human tumor-associated antigen chondroitin sulfate proteoglycan 4 (CSPG4). Anti-CSPG4 IgE and IgG1 antibodies with human Fc domains were radiolabeled with 111In. CSPG4-expressing A375 human melanoma xenografts implanted in NOD-scid IL2rg-/- mice were also engrafted with human immune cells by intravenous administration. 111In-anti-CSPG4 antibodies were administered intravenously. Their distribution was determined by single-photon emission computed tomography (SPECT) and ex vivo gamma-counting over 120 h. SPECT imaging was conducted from 0 to 60 min after antibody administration to precisely measure the early phase of IgE distribution. 111In-labeled anti-CSPG4 IgG and IgE showed serum stability in vitro of >92% after 5 days. In A375 xenograft-bearing mice, anti-CSPG4 IgE showed much faster blood clearance and higher accumulation in the liver compared to anti-CSPG4 IgG. However, tumor-to-blood and tumor-to-muscle ratios were similar between the antibody isotypes and higher compared with a non-tumor-targeting isotype control IgE. IgE excretion was much faster than IgG. In non-tumor-bearing animals, early SPECT imaging revealed a blood clearance half-life of 10 min for IgE. Using image-based quantification, we demonstrated that the blood clearance of IgE is much faster than that of IgG while the two isotypes showed comparable tumor-to-blood ratios.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
- School of Cancer & Pharmaceutical Sciences, Institute of Pharmaceutical Science, King’s College London, London, UK
| | - Alexander Koers
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Panagiotis Karagiannis
- School of Basic & Medical Biosciences, St John’s Institute of Dermatology, King’s College London, London, UK
| | - Debra H. Josephs
- School of Basic & Medical Biosciences, St John’s Institute of Dermatology, King’s College London, London, UK
- School of Cancer & Pharmaceutical Sciences, Guy’s Hospital, King’s College London, London, UK
| | - Heather J. Bax
- School of Basic & Medical Biosciences, St John’s Institute of Dermatology, King’s College London, London, UK
- School of Cancer & Pharmaceutical Sciences, Guy’s Hospital, King’s College London, London, UK
| | - Amy E. Gilbert
- School of Basic & Medical Biosciences, St John’s Institute of Dermatology, King’s College London, London, UK
| | - Tihomir S. Dodev
- School of Cancer & Pharmaceutical Sciences, Guy’s Hospital, King’s College London, London, UK
- School of Basic and Medical Biosciences, Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
- Allergic Mechanisms in Asthma, Asthma UK Centre, King’s College London, London, UK
| | - Silvia Mele
- School of Basic & Medical Biosciences, St John’s Institute of Dermatology, King’s College London, London, UK
| | - Giulia Chiarruttini
- School of Basic & Medical Biosciences, St John’s Institute of Dermatology, King’s College London, London, UK
| | - Silvia Crescioli
- School of Basic & Medical Biosciences, St John’s Institute of Dermatology, King’s College London, London, UK
| | - Jitesh Chauhan
- School of Basic & Medical Biosciences, St John’s Institute of Dermatology, King’s College London, London, UK
- School of Cancer & Pharmaceutical Sciences, Guy’s Hospital, King’s College London, London, UK
| | - Julia E. Blower
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Margaret S. Cooper
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - James Spicer
- School of Cancer & Pharmaceutical Sciences, Guy’s Hospital, King’s College London, London, UK
- Cancer Centre at Guy’s, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Sophia N. Karagiannis
- School of Basic & Medical Biosciences, St John’s Institute of Dermatology, King’s College London, London, UK
- School of Cancer & Pharmaceutical Sciences, Breast Cancer Now Research Unit, King’s College London, Guy’s Hospital, London, UK
| | - Philip J. Blower
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| |
Collapse
|
43
|
Astronomo RD, Lemos MP, Narpala SR, Czartoski J, Fleming LB, Seaton KE, Prabhakaran M, Huang Y, Lu Y, Westerberg K, Zhang L, Gross MK, Hural J, Tieu HV, Baden LR, Hammer S, Frank I, Ochsenbauer C, Grunenberg N, Ledgerwood JE, Mayer K, Tomaras G, McDermott AB, McElrath MJ. Rectal tissue and vaginal tissue from intravenous VRC01 recipients show protection against ex vivo HIV-1 challenge. J Clin Invest 2021; 131:e146975. [PMID: 34166231 DOI: 10.1172/jci146975] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/22/2021] [Indexed: 11/17/2022] Open
Abstract
BackgroundVRC01, a potent, broadly neutralizing monoclonal antibody, inhibits simian-HIV infection in animal models. The HVTN 104 study assessed the safety and pharmacokinetics of VRC01 in humans. We extend the clinical evaluation to determine intravenously infused VRC01 distribution and protective function at mucosal sites of HIV-1 entry.MethodsHealthy, HIV-1-uninfected men (n = 7) and women (n = 5) receiving VRC01 every 2 months provided mucosal and serum samples once, 4-13 days after infusion. Eleven male and 8 female HIV-seronegative volunteers provided untreated control samples. VRC01 levels were measured in serum, secretions, and tissue, and HIV-1 inhibition was determined in tissue explants.ResultsMedian VRC01 levels were quantifiable in serum (96.2 μg/mL or 1.3 pg/ng protein), rectal tissue (0.11 pg/ng protein), rectal secretions (0.13 pg/ng protein), vaginal tissue (0.1 pg/ng protein), and cervical secretions (0.44 pg/ng protein) from all recipients. VRC01/IgG ratios in male serum correlated with those in paired rectal tissue (r = 0.893, P = 0.012) and rectal secretions (r = 0.9643, P = 0.003). Ex vivo HIV-1Bal26 challenge infected 4 of 21 rectal explants from VRC01 recipients versus 20 of 22 from controls (P = 0.005); HIV-1Du422.1 infected 20 of 21 rectal explants from VRC01 recipients and 12 of 12 from controls (P = 0.639). HIV-1Bal26 infected 0 of 14 vaginal explants of VRC01 recipients compared with 23 of 28 control explants (P = 0.003).ConclusionIntravenous VRC01 distributes into the female genital and male rectal mucosa and retains anti-HIV-1 functionality, inhibiting a highly neutralization-sensitive but not a highly resistant HIV-1 strain in mucosal tissue. These findings lend insight into VRC01 mucosal infiltration and provide perspective on in vivo protective efficacy.FundingNational Institute of Allergy and Infectious Diseases and Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Rena D Astronomo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Maria P Lemos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Sandeep R Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Julie Czartoski
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Lamar Ballweber Fleming
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kelly E Seaton
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Madhu Prabhakaran
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Katharine Westerberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Lily Zhang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Mary K Gross
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - John Hural
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Lindsey R Baden
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Scott Hammer
- Columbia University Medical Center, New York, New York, USA
| | - Ian Frank
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Nicole Grunenberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | - Georgia Tomaras
- Department of Surgery, Duke University, Durham, North Carolina, USA.,Department of Immunology and Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
44
|
Valente D, Mauriac C, Schmidt T, Focken I, Beninga J, Mackness B, Qiu H, Vicat P, Kandira A, Radošević K, Rao S, Darbyshire J, Kabiri M. Pharmacokinetics of novel Fc-engineered monoclonal and multispecific antibodies in cynomolgus monkeys and humanized FcRn transgenic mouse models. MAbs 2021; 12:1829337. [PMID: 33079615 PMCID: PMC7587234 DOI: 10.1080/19420862.2020.1829337] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Monoclonal antibodies (mAbs) are among the fastest growing and most effective therapies for myriad diseases. Multispecific antibodies are an emerging class of novel therapeutics that can target more than one tumor- or immune-associated modulators per molecule. The combination of different binding affinities and target classes, such as soluble or membrane-bound antigens, within multispecific antibodies confers unique pharmacokinetic (PK) properties. Numerous factors affect an antibody’s PK, with affinity to the neonatal Fc receptor (FcRn) a key determinant of half-life. Recent work has demonstrated the potential for humanized FcRn transgenic mice to predict the PK of mAbs in humans. However, such work has not been extended to multispecific antibodies. We engineered mAbs and multispecific antibodies with various Fc modifications to enhance antibody performance. PK analyses in humanized FcRn transgenic mouse (homozygous Tg32 and Tg276) and non-human primate (NHP) models showed that FcRn-binding mutations improved the plasma half-lives of the engineered mAbs and multispecific antibodies, while glycan engineering to eliminate effector function did not affect the PK compared with wild-type controls. Furthermore, results suggest that the homozygous Tg32 mouse model can replace NHP models to differentiate PK of variants during lead optimization, not only for wild-type mAbs but also for Fc-engineered mAbs and multispecific antibodies. This Tg32-mouse model would enable prediction of half-life and linear clearance of mAbs and multispecific antibodies in NHPs to guide the design of further pharmacology/safety studies in this species. The allometric exponent for clearance scaling from Tg32 mice to NHPs was estimated to be 0.91 for all antibodies.
Collapse
Affiliation(s)
| | | | | | - Ingo Focken
- Biologics Research, Sanofi , Frankfurt, Germany
| | | | - Brian Mackness
- Translational In vivo Models (TIM), Sanofi, Frankfurt, Germany
| | - Huawei Qiu
- Biologics, Xilio Therapeutics, Inc. Cambridge, US
| | | | | | | | - Srini Rao
- Translational In vivo Models (TIM), Sanofi, Cambridge, US
| | | | - Mostafa Kabiri
- Translational In vivo Models (TIM), Sanofi, Frankfurt, Germany
| |
Collapse
|
45
|
Liu C, Kim YS, Lowe JHN, Chung S. A cell-based FcRn-dependent recycling assay for predictive pharmacokinetic assessment of therapeutic antibodies. Bioanalysis 2021; 13:1135-1144. [PMID: 34289743 DOI: 10.4155/bio-2021-0099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aim: Evaluation of suitable pharmacokinetic properties is critical for successful development of IgG-based biotherapeutics. The prolonged half-lives of IgGs depend on the intracellular trafficking function of neonatal Fc receptor, which rescues internalized IgGs from lysosomal degradation and recycles them back to circulation. Results: Here, we developed a novel cell-based assay to quantify recycling of monoclonal antibodies in a transwell culture system that uses a cell line that stably expresses human neonatal Fc receptor. We tested seven therapeutic antibodies and showed that the recycling output of the assay strongly correlated with the clearance in humans. Conclusion: This recycling assay has potential application as a pharmacokinetic prescreening tool to facilitate development and selection of IgG-based candidate therapeutic monoclonal antibodies.
Collapse
Affiliation(s)
- Chang Liu
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yeon Su Kim
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - John Hok-Nin Lowe
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Shan Chung
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
46
|
Kouhi A, Pachipulusu V, Kapenstein T, Hu P, Epstein AL, Khawli LA. Brain Disposition of Antibody-Based Therapeutics: Dogma, Approaches and Perspectives. Int J Mol Sci 2021; 22:ijms22126442. [PMID: 34208575 PMCID: PMC8235515 DOI: 10.3390/ijms22126442] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
Due to their high specificity, monoclonal antibodies have been widely investigated for their application in drug delivery to the central nervous system (CNS) for the treatment of neurological diseases such as stroke, Alzheimer’s, and Parkinson’s disease. Research in the past few decades has revealed that one of the biggest challenges in the development of antibodies for drug delivery to the CNS is the presence of blood–brain barrier (BBB), which acts to restrict drug delivery and contributes to the limited uptake (0.1–0.2% of injected dose) of circulating antibodies into the brain. This article reviews the various methods currently used for antibody delivery to the CNS at the preclinical stage of development and the underlying mechanisms of BBB penetration. It also describes efforts to improve or modulate the physicochemical and biochemical properties of antibodies (e.g., charge, Fc receptor binding affinity, and target affinity), to adapt their pharmacokinetics (PK), and to influence their distribution and disposition into the brain. Finally, a distinction is made between approaches that seek to modify BBB permeability and those that use a physiological approach or antibody engineering to increase uptake in the CNS. Although there are currently inherent difficulties in developing safe and efficacious antibodies that will cross the BBB, the future prospects of brain-targeted delivery of antibody-based agents are believed to be excellent.
Collapse
|
47
|
Lombardi S, Aaen KH, Nilsen J, Ferrarese M, Gjølberg TT, Bernardi F, Pinotti M, Andersen JT, Branchini A. Fusion of engineered albumin with factor IX Padua extends half-life and improves coagulant activity. Br J Haematol 2021; 194:453-462. [PMID: 34109608 PMCID: PMC8362221 DOI: 10.1111/bjh.17559] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 12/29/2022]
Abstract
The short half‐life of coagulation factor IX (FIX) for haemophilia B (HB) therapy has been prolonged through fusion with human serum albumin (HSA), which drives the neonatal Fc receptor (FcRn)‐mediated recycling of the chimera. However, patients would greatly benefit from further FIX‐HSA half‐life extension. In the present study, we designed a FIX‐HSA variant through the engineering of both fusion partners. First, we developed a novel cleavable linker combining the two FIX activation sites, which resulted in improved HSA release. Second, insertion of the FIX R338L (Padua) substitution conferred hyperactive features (sevenfold higher specific activity) as for FIX Padua alone. Furthermore, we exploited an engineered HSA (QMP), which conferred enhanced human (h)FcRn binding [dissociation constant (KD) 0·5 nM] over wild‐type FIX‐HSA (KD 164·4 nM). In hFcRn transgenic mice, Padua‐QMP displayed a significantly prolonged half‐life (2·7 days, P < 0·0001) versus FIX‐HSA (1 day). Overall, we developed a novel FIX‐HSA protein with improved activity and extended half‐life. These combined properties may result in a prolonged functional profile above the therapeutic threshold, and thus in a potentially widened therapeutic window able to improve HB therapy. This rational engineering of both partners may pave the way for new fusion strategies for the design of engineered biotherapeutics.
Collapse
Affiliation(s)
- Silvia Lombardi
- Department of Life Sciences and Biotechnology and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Kristin H Aaen
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jeannette Nilsen
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Mattia Ferrarese
- Department of Life Sciences and Biotechnology and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Torleif T Gjølberg
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Ophthalmology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Mirko Pinotti
- Department of Life Sciences and Biotechnology and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Jan T Andersen
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Alessio Branchini
- Department of Life Sciences and Biotechnology and LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
48
|
Fung E, Kang L, Sapashnik D, Benard S, Sievers A, Liu Y, Yan G, Zhou J, Rodriguez L, Ma W, Stochaj WR, LaVallie E, Wroblewska L, Kelleher K, Tam A, Bezy O, Breen D, Chabot JR, He T, Lin L, Wu Z, Mosyak L. Fc-GDF15 glyco-engineering and receptor binding affinity optimization for body weight regulation. Sci Rep 2021; 11:8921. [PMID: 33903632 PMCID: PMC8076310 DOI: 10.1038/s41598-021-87959-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Abstract
GDF15 is a distant TGF-β family member that induces anorexia and weight loss. Due to its function, GDF15 has attracted attention as a potential therapeutic for the treatment of obesity and its associated metabolic diseases. However, the pharmacokinetic and physicochemical properties of GDF15 present several challenges for its development as a therapeutic, including a short half-life, high aggregation propensity, and protease susceptibility in serum. Here, we report the design, characterization and optimization of GDF15 in an Fc-fusion protein format with improved therapeutic properties. Using a structure-based engineering approach, we combined knob-into-hole Fc technology and N-linked glycosylation site mutagenesis for half-life extension, improved solubility and protease resistance. In addition, we identified a set of mutations at the receptor binding site of GDF15 that show increased GFRAL binding affinity and led to significant half-life extension. We also identified a single point mutation that increases p-ERK signaling activity and results in improved weight loss efficacy in vivo. Taken together, our findings allowed us to develop GDF15 in a new therapeutic format that demonstrates better efficacy and potential for improved manufacturability.
Collapse
Affiliation(s)
- Ella Fung
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Liya Kang
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Diana Sapashnik
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Susan Benard
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Annette Sievers
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Yan Liu
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Guoying Yan
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Jing Zhou
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Linette Rodriguez
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Weijun Ma
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA.,Sanofi Research and Development, Sanofi North America, Framingham, MA, USA
| | - Wayne R Stochaj
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Edward LaVallie
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | | | - Kerry Kelleher
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Amy Tam
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Olivier Bezy
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA.,Cellarity, Cambridge, MA, USA
| | - Danna Breen
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Jeffrey R Chabot
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Tao He
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA.,JOINN Biologics US Inc, Richmond, CA, USA
| | - Laura Lin
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA
| | - Zhidan Wu
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Lidia Mosyak
- BioMedicine Design, Pfizer Inc., 610 N Main Street, Cambridge, MA, USA.
| |
Collapse
|
49
|
Ke C, Ma Y, Pan D, Wan Z, Feng T, Yu D, Liu X, Wang H, Du M, Huang L, Zhang Y, Du L, Wang X, Li K, Yu D, Zhang M, Huang J, Qu J, Ren L, Hu Y, Cao G, Hu X, Wu S, Han H, Zhao Y. FcRn is not the receptor mediating the transfer of serum IgG to colostrum in pigs. Immunology 2021; 163:448-459. [PMID: 33738807 DOI: 10.1111/imm.13328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 02/02/2023] Open
Abstract
In contrast to humans or rabbits, in which maternal IgG is transmitted to offspring prenatally via the placenta or the yolk sac, large domestic animals such as pigs, cows and sheep transmit IgG exclusively through colostrum feeding after delivery. The extremely high IgG content in colostrum is absorbed by newborns via the small intestine. Although it is widely accepted that the neonatal Fc receptor, FcRn, is the receptor mediating IgG transfer across both the placenta and small intestine, it remains unclear whether FcRn also mediates serum IgG transfer across the mammary barrier to colostrum/milk, especially in large domestic animals. In this study, using a FcRn knockout pig model generated with a CRISPR-Cas9-based approach, we clearly demonstrate that FcRn is not responsible for the IgG transfer from serum to colostrum in pigs, although like in other mammals, it is involved in IgG homeostasis and mediates IgG absorption in the small intestine of newborns.
Collapse
Affiliation(s)
- Cuncun Ke
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Yonghe Ma
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Dengke Pan
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Zihui Wan
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Tao Feng
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Dawei Yu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Xiaojuan Liu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Haitao Wang
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Minjie Du
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Linhua Huang
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yifu Zhang
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lijuan Du
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Xifeng Wang
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Kongpan Li
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Di Yu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Ming Zhang
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Jinwei Huang
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Junwei Qu
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Liming Ren
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Yanzhong Hu
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Gengsheng Cao
- Henan Engineering Laboratory for Mammary Bioreactor, School of Life Science, Henan University, Kaifeng, China
| | - Xiaoxiang Hu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Sen Wu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Haitang Han
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Yaofeng Zhao
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| |
Collapse
|
50
|
Strasser L, Farrell A, Ho JTC, Scheffler K, Cook K, Pankert P, Mowlds P, Viner R, Karger BL, Bones J. Proteomic Profiling of IgG1 Producing CHO Cells Using LC/LC-SPS-MS 3: The Effects of Bioprocessing Conditions on Productivity and Product Quality. Front Bioeng Biotechnol 2021; 9:569045. [PMID: 33898396 PMCID: PMC8062983 DOI: 10.3389/fbioe.2021.569045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
The biopharmaceutical market is dominated by monoclonal antibodies, the majority of which are produced in Chinese hamster ovary (CHO) cell lines. Intense cell engineering, in combination with optimization of various process parameters results in increasing product titers. To enable further improvements in manufacturing processes, detailed information about how certain parameters affect cellular mechanisms in the production cells, and thereby also the expressed drug substance, is required. Therefore, in this study the effects of commonly applied changes in bioprocessing parameters on an anti-IL8 IgG1 producing CHO DP-12 cell line were investigated on the level of host cell proteome expression combined with product quality assessment of the expressed IgG1 monoclonal antibody. Applying shifts in temperature, pH and dissolved oxygen concentration, respectively, resulted in altered productivity and product quality. Furthermore, analysis of the cells using two-dimensional liquid chromatography-mass spectrometry employing tandem mass tag based isotopic quantitation and synchronous precursor selection-MS3 detection revealed substantial changes in the protein expression profiles of CHO cells. Pathway analysis indicated that applied bioprocessing conditions resulted in differential activation of oxidative phosphorylation. Additionally, activation of ERK5 and TNFR1 signaling suggested an affected cell cycle. Moreover, in-depth product characterization by means of charge variant analysis, peptide mapping, as well as structural and functional analysis, revealed posttranslational and structural changes in the expressed drug substance. Taken together, the present study allows the conclusion that, in anti-IL8 IgG1 producing CHO DP-12 cells, an improved energy metabolism achieved by lowering the cell culture pH is favorable when aiming towards high antibody production rates while maintaining product quality.
Collapse
Affiliation(s)
- Lisa Strasser
- Characterization and Comparability Laboratory, National Institute for Bioprocessing Research and Training, Dublin, Ireland
| | - Amy Farrell
- Characterization and Comparability Laboratory, National Institute for Bioprocessing Research and Training, Dublin, Ireland
| | - Jenny T C Ho
- Thermo Fisher Scientific, Hemel Hempstead, United Kingdom
| | | | - Ken Cook
- Thermo Fisher Scientific, Hemel Hempstead, United Kingdom
| | | | - Peter Mowlds
- Thermo Fisher Scientific, Hemel Hempstead, United Kingdom
| | - Rosa Viner
- Thermo Fisher Scientific, San Jose, CA, United States
| | - Barry L Karger
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, United States
| | - Jonathan Bones
- School of Chemical and Bioprocess Engineering, University College Dublin, Dublin, Ireland
| |
Collapse
|