1
|
Bryniarski MA, Tuhin MTH, Acker TM, Wakefield DL, Sethaputra PG, Cook KD, Soto M, Ponce M, Primack R, Jagarapu A, LaGory EL, Conner KP. Cellular Neonatal Fc Receptor Recycling Efficiencies can Differentiate Target-Independent Clearance Mechanisms of Monoclonal Antibodies. J Pharm Sci 2024; 113:2879-2894. [PMID: 38906252 DOI: 10.1016/j.xphs.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
In vivo clearance mechanisms of therapeutic monoclonal antibodies (mAbs) encompass both target-mediated and target-independent processes. Two distinct determinants of overall mAb clearance largely separate of target-mediated influences are non-specific cellular endocytosis and subsequent pH-dependent mAb recycling mediated by the neonatal Fc receptor (FcRn), where inter-mAb variability in the efficiency of both processes is observed. Here, we implemented a functional cell-based FcRn recycling assay via Madin-Darby canine kidney type II cells stably co-transfected with human FcRn and its light chain β2-microglobulin. Next, a series of pH-dependent internalization studies using a model antibody demonstrated proper function of the human FcRn complex. We then applied our cellular assays to assess the contribution of both FcRn and non-specific interactions in the cellular turnover for a panel of 8 clinically relevant mAbs exhibiting variable human pharmacokinetic behavior. Our results demonstrate that the interplay of non-specific endocytosis rates, pH-dependent non-specific interactions, and engagement with FcRn all contribute to the overall recycling efficiency of therapeutic monoclonal antibodies. The predictive capacity of our assay approach was highlighted by successful identification of all mAbs within our panel possessing clearance in humans greater than 5 mL/day/kg. These results demonstrate that a combination of cell-based in vitro assays can properly resolve individual mechanisms underlying the overall in vivo recycling efficiency and non-target mediated clearance of therapeutic mAbs.
Collapse
Affiliation(s)
- Mark A Bryniarski
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA.
| | - Md Tariqul Haque Tuhin
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Timothy M Acker
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Devin L Wakefield
- Research Biomics, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Panijaya Gemy Sethaputra
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Kevin D Cook
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Marcus Soto
- Pharmacokinetics & Drug Metabolism, Amgen Research, One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Manuel Ponce
- Pharmacokinetics & Drug Metabolism, Amgen Research, One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Ronya Primack
- Pharmacokinetics & Drug Metabolism, Amgen Research, One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Aditya Jagarapu
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Edward L LaGory
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Kip P Conner
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA.
| |
Collapse
|
2
|
Karakioulaki M, Eyerich K, Patsatsi A. Advancements in Bullous Pemphigoid Treatment: A Comprehensive Pipeline Update. Am J Clin Dermatol 2024; 25:195-212. [PMID: 38157140 PMCID: PMC10866767 DOI: 10.1007/s40257-023-00832-1] [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] [Accepted: 11/19/2023] [Indexed: 01/03/2024]
Abstract
ABASTRACT Bullous pemphigoid (BP) is a common autoimmune bullous disease affecting mainly the elderly, with rising incidence due to increased life expectancy. This disease is characterized by tense bullous lesions on normal or erythematous skin, accompanied by pruritus. BP pathogenesis involves autoantibodies against hemidesmosomal proteins BP180 and BP230, leading to detachment at the dermo-epidermal junction as well as blister formation. BP is associated with coexisting comorbidities and drug exposure, and its management often requires high doses or chronic use of systemic glucocorticoids, posing risks of adverse effects. This review focuses on novel treatment options for BP, exploring therapies targeting different immune pathways. Rituximab, a CD20 monoclonal antibody, depletes B-lymphocytes and has shown efficacy in severe cases. Dupilumab, targeting interleukin (IL)-4 receptor α and thus blocking IL-4 and IL-13, downregulates type 2 helper (Th2) responses and has demonstrated promising results. Targeting eosinophil-related molecules using bertilimumab and AKST4290 has yielded positive results in clinical trials. Omalizumab, an immunoglobulin (Ig) E antibody, can reduce disease severity and allows corticosteroid tapering in a number of cases. Complement inhibitors such as nomacopan and avdoralimab are being investigated. IL-17 and IL-23 inhibitors such as secukinumab and tildrakizumab have shown potential in a limited number of case reports. Neonatal Fc receptor antagonists such as efgartigimod are under investigation. Additionally, topical therapies and Janus kinase inhibitors are being explored as potential treatments for BP. These novel therapies offer promising alternatives for managing BP, with potential to improve outcomes and reduce high cumulative doses of systemic corticosteroids and related toxicities. Further research, including controlled clinical trials, is needed to establish their efficacy, safety, and optimal dosing regimens for BP management.
Collapse
Affiliation(s)
- Meropi Karakioulaki
- Department of Dermatology and Venerology, Medical Center, University Hospital Freiburg, Freiburg, Germany
| | - Kilian Eyerich
- Department of Dermatology and Venerology, Medical Center, University Hospital Freiburg, Freiburg, Germany
| | - Aikaterini Patsatsi
- Second Department of Dermatology, School of Medicine, Papageorgiou Hospital, Aristotle University, Thessaloníki, Greece.
| |
Collapse
|
3
|
Chen HC, Wang CW, Toh WH, Lee HE, Chung WH, Chen CB. Advancing Treatment in Bullous Pemphigoid: A Comprehensive Review of Novel Therapeutic Targets and Approaches. Clin Rev Allergy Immunol 2023; 65:331-353. [PMID: 37897588 DOI: 10.1007/s12016-023-08973-1] [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] [Accepted: 10/15/2023] [Indexed: 10/30/2023]
Abstract
Bullous pemphigoid is one of the most common autoimmune bullous diseases occurring primarily in the elderly. Pathogenic autoantibodies against BP180 and BP230 at the dermal-epidermal junction cause subepidermal blisters, erosions, and intense pruritus, all of which adversely affect the patients' quality of life and may increase their morbidity and mortality. Current systemic treatment options for bullous pemphigoid are limited to corticosteroids and immunosuppressants, which can have substantial side effects on these vulnerable patients that even exceed their therapeutic benefits. Therefore, more precisely, targeting therapies to the pathogenic cells and molecules in bullous pemphigoid is an urgent issue. In this review, we describe the pathophysiology of bullous pemphigoid, focusing on autoantibodies, complements, eosinophils, neutrophils, proteases, and the T helper 2 and 17 axes since they are crucial in promoting proinflammatory environments. We also highlight the emerging therapeutic targets for bullous pemphigoid and their latest discoveries in clinical trials or experimental studies. Further well-designed studies are required to establish the efficacy and safety of these prospective therapeutic options.
Collapse
Affiliation(s)
- Hsuan-Chi Chen
- Department of Medical Education, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chuang-Wei Wang
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Linkou, Taipei, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China
| | - Wu Han Toh
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Hua-En Lee
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Linkou, Taipei, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Hung Chung
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Linkou, Taipei, Taiwan.
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan.
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung, Taiwan.
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China.
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan.
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan.
- Department of Dermatology, Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
- Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan.
| | - Chun-Bing Chen
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Linkou, Taipei, Taiwan.
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan.
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung, Taiwan.
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China.
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China.
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan.
| |
Collapse
|
4
|
Zeng FAP, Murrell DF. Bullous pemphigoid-What do we know about the most recent therapies? Front Med (Lausanne) 2022; 9:1057096. [PMID: 36405625 PMCID: PMC9669062 DOI: 10.3389/fmed.2022.1057096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/18/2022] [Indexed: 08/25/2023] Open
Abstract
Introduction Bullous pemphigoid (BP) is the most common subtype of autoimmune blistering diseases that primarily affects the elderly and is classically defined by the presence of IgG and/or complement C3 against the BP180 and BP230 hemidesmosome proteins. However, most recent studies have introduced the role of specific eosinophil receptors and chemokine mediators in the pathogenesis of BP which are helpful in identifying new targets for future treatments. Areas covered This review will focus on the involvement of eosinophils in BP, including the processes that lead to their recruitment, activation, and regulation. Subsequently, covering new therapeutic options in relation to the role of eosinophils. Eotaxin enhances the recruitment of eosinophils in BP, with CCR3 chemoreceptor that is expressed on eosinophils being identified as a key binding site for eotaxin-1. The pathogenic role of IgE and IL-4 in BP is corroborated by successful treatments with Omalizumab and Dupilumab, respectively. IL-5, IL-17 and IL-23 inhibitors may be effective given their roles in promoting eosinophilia. Expert opinion Further research into inhibitors of eotaxin, IL-4, IL-5, IL-17, IL-23, CCR3, and specific complement factors are warranted as preliminary studies have largely identified success in treating BP with these agents. Learning from novel treatments for other IgG-mediated autoimmune diseases may be beneficial.
Collapse
Affiliation(s)
- Faith A. P. Zeng
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Dedee F. Murrell
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Dermatology, St George Hospital, Sydney, NSW, Australia
- The George Institute for Global Health, Sydney, NSW, Australia
| |
Collapse
|
5
|
Heo YH, Kim JK, Lee JS, Lee SH, Shin SH, Choi IY, Kim HH. A novel glucagon analog with an extended half-life, HM15136, normalizes glucose levels in rodent models of congenital hyperinsulinism. Sci Rep 2022; 12:16765. [PMID: 36202918 PMCID: PMC9537296 DOI: 10.1038/s41598-022-21251-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022] Open
Abstract
Congenital hyperinsulinism (CHI) is a rare genetic condition characterized by uncontrolled insulin secretion, resulting in hypoglycemia. Although glucagon has lately been regarded as a therapeutic option for CHI, its use is severely hampered by its poor solubility and stability at physiological pH, as well as its short duration of action. To address these constraints, we developed HM15136, a novel long-acting glucagon analog composed of a glucagon analog conjugated to the Fc fragment of human immunoglobulin G4 via a polyethylene glycol linker. In this study, we established that HM15136 was more soluble than natural glucagon (≥ 150 mg/mL vs 0.03 mg/mL). Next, we confirmed that HM15136 activated glucagon receptor in vitro and induced glycogenolysis and gluconeogenesis in rat primary hepatocytes. Pharmacokinetics (PK)/Pharmacodynamics (PD) analysis of HM15136 shows that HM15136 has a markedly longer half-life (36 h vs. < 5 min) and increased bioavailability (90%) compared to native glucagon in mice. Further, HM15136 could effectively reverse acute hypoglycemia induced by insulin challenge, and multiple doses of HM15136 could sustain increased blood glucose levels in CHI rats. In conclusion, our findings indicate that HM15136 promotes sustained elevation of blood glucose, demonstrating the potential for development as a once-weekly therapy for CHI.
Collapse
Affiliation(s)
- Yong Ho Heo
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., 550 Dongtangiheung-ro, Hwaseong-si, 18469, Gyeonggi-do, Republic of Korea
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Jung Kuk Kim
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., 550 Dongtangiheung-ro, Hwaseong-si, 18469, Gyeonggi-do, Republic of Korea
| | - Jong Suk Lee
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., 550 Dongtangiheung-ro, Hwaseong-si, 18469, Gyeonggi-do, Republic of Korea
| | - Sang-Hyun Lee
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., 550 Dongtangiheung-ro, Hwaseong-si, 18469, Gyeonggi-do, Republic of Korea
| | - Seung-Hyun Shin
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., 550 Dongtangiheung-ro, Hwaseong-si, 18469, Gyeonggi-do, Republic of Korea
| | - In Young Choi
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd., 550 Dongtangiheung-ro, Hwaseong-si, 18469, Gyeonggi-do, Republic of Korea.
| | - Ha Hyung Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
6
|
Molitoris BA, Sandoval RM, Yadav SPS, Wagner MC. Albumin Uptake and Processing by the Proximal Tubule: Physiologic, Pathologic and Therapeutic Implications. Physiol Rev 2022; 102:1625-1667. [PMID: 35378997 PMCID: PMC9255719 DOI: 10.1152/physrev.00014.2021] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
For nearly 50 years the proximal tubule (PT) has been known to reabsorb, process, and either catabolize or transcytose albumin from the glomerular filtrate. Innovative techniques and approaches have provided insights into these processes. Several genetic diseases, nonselective PT cell defects, chronic kidney disease (CKD), and acute PT injury lead to significant albuminuria, reaching nephrotic range. Albumin is also known to stimulate PT injury cascades. Thus, the mechanisms of albumin reabsorption, catabolism, and transcytosis are being reexamined with the use of techniques that allow for novel molecular and cellular discoveries. Megalin, a scavenger receptor, cubilin, amnionless, and Dab2 form a nonselective multireceptor complex that mediates albumin binding and uptake and directs proteins for lysosomal degradation after endocytosis. Albumin transcytosis is mediated by a pH-dependent binding affinity to the neonatal Fc receptor (FcRn) in the endosomal compartments. This reclamation pathway rescues albumin from urinary losses and cellular catabolism, extending its serum half-life. Albumin that has been altered by oxidation, glycation, or carbamylation or because of other bound ligands that do not bind to FcRn traffics to the lysosome. This molecular sorting mechanism reclaims physiological albumin and eliminates potentially toxic albumin. The clinical importance of PT albumin metabolism has also increased as albumin is now being used to bind therapeutic agents to extend their half-life and minimize filtration and kidney injury. The purpose of this review is to update and integrate evolving information regarding the reabsorption and processing of albumin by proximal tubule cells including discussion of genetic disorders and therapeutic considerations.
Collapse
Affiliation(s)
- Bruce A. Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Dept.of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Ruben M. Sandoval
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Shiv Pratap S. Yadav
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Mark C. Wagner
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
7
|
Grevys A, Frick R, Mester S, Flem-Karlsen K, Nilsen J, Foss S, Sand KMK, Emrich T, Fischer JAA, Greiff V, Sandlie I, Schlothauer T, Andersen JT. Antibody variable sequences have a pronounced effect on cellular transport and plasma half-life. iScience 2022; 25:103746. [PMID: 35118359 PMCID: PMC8800109 DOI: 10.1016/j.isci.2022.103746] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/11/2021] [Accepted: 01/05/2022] [Indexed: 11/15/2022] Open
Abstract
Monoclonal IgG antibodies are the fastest growing class of biologics, but large differences exist in their plasma half-life in humans. Thus, to design IgG antibodies with favorable pharmacokinetics, it is crucial to identify the determinants of such differences. Here, we demonstrate that the variable region sequences of IgG antibodies greatly affect cellular uptake and subsequent recycling and rescue from intracellular degradation by endothelial cells. When the variable sequences are masked by the cognate antigen, it influences both their transport behavior and binding to the neonatal Fc receptor (FcRn), a key regulator of IgG plasma half-life. Furthermore, we show how charge patch differences in the variable domains modulate both binding and transport properties and that a short plasma half-life, due to unfavorable charge patches, may partly be overcome by Fc-engineering for improved FcRn binding. IgG variable region sequences greatly affect cellular uptake and recycling Variable region charge patches affect FcRn binding and transport The presence of cognate antigen modulates cellular transport and FcRn binding Fc-engineering for improved FcRn binding can overcome unfavorable charge patches
Collapse
Affiliation(s)
- Algirdas Grevys
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
- Corresponding author
| | - Rahel Frick
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Simone Mester
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Karine Flem-Karlsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Jeannette Nilsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Stian Foss
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Kine Marita Knudsen Sand
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Thomas Emrich
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | | | - Victor Greiff
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Inger Sandlie
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Jan Terje Andersen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Corresponding author
| |
Collapse
|
8
|
Schmitt T, Waschke J. Autoantibody-Specific Signalling in Pemphigus. Front Med (Lausanne) 2021; 8:701809. [PMID: 34434944 PMCID: PMC8381052 DOI: 10.3389/fmed.2021.701809] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022] Open
Abstract
Pemphigus is a severe autoimmune disease impairing barrier functions of epidermis and mucosa. Autoantibodies primarily target the desmosomal adhesion molecules desmoglein (Dsg) 1 and Dsg 3 and induce loss of desmosomal adhesion. Strikingly, autoantibody profiles in pemphigus correlate with clinical phenotypes. Mucosal-dominant pemphigus vulgaris (PV) is characterised by autoantibodies (PV-IgG) against Dsg3 whereas epidermal blistering in PV and pemphigus foliaceus (PF) is associated with autoantibodies against Dsg1. Therapy in pemphigus is evolving towards specific suppression of autoantibody formation and autoantibody depletion. Nevertheless, during the acute phase and relapses of the disease additional treatment options to stabilise desmosomes and thereby rescue keratinocyte adhesion would be beneficial. Therefore, the mechanisms by which autoantibodies interfere with adhesion of desmosomes need to be characterised in detail. Besides direct inhibition of Dsg adhesion, autoantibodies engage signalling pathways interfering with different steps of desmosome turn-over. With this respect, recent data indicate that autoantibodies induce separate signalling responses in keratinocytes via specific signalling complexes organised by Dsg1 and Dsg3 which transfer the signal of autoantibody binding into the cell. This hypothesis may also explain the different clinical pemphigus phenotypes.
Collapse
Affiliation(s)
- Thomas Schmitt
- Ludwig-Maximilian-Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I - Vegetative Anatomie, Munich, Germany
| | - Jens Waschke
- Ludwig-Maximilian-Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I - Vegetative Anatomie, Munich, Germany
| |
Collapse
|
9
|
Abstract
Purpose Growing evidence suggests different systemic exposure of anti-vascular endothelial growth factor (anti-VEGF) agents with repeated intravitreal application. Since the penetration of anti-VEGF agents through vascular barrier was reported, the interaction of anti-VEGF with nonresident platelets has become a topic of interest. The purpose of this study was to evaluate, with the help of visualization techniques, whether platelets take up the anti-VEGF agents ranibizumab, aflibercept, and bevacizumab. Methods The uptake of anti-VEGF agents with or without VEGF treatment was investigated using immunofluorescence and immunogold staining in human platelets. The role of actin filaments and clathrin-coated vesicles in the transport of ranibizumab, aflibercept, and bevacizumab was evaluated by two pharmacologic inhibitors: staurosporine (protein kinase C inhibitor) and cytochalasin D. Results All three anti-VEGF agents were taken up by platelets and colocalized with VEGF. Ranibizumab and aflibercept were mainly detected in alpha-granules; however, bevacizumab was equally localized in alpha-granules and in platelet vesicles. Both staurosporine and cytochalasin D completely inhibited the uptake of aflibercept into platelets. Both pharmacological inhibitors also decreased the transport of ranibizumab and bevacizumab into platelets. Bevacizumab was significantly more frequently colocalized within clathrin-coated vesicles than ranibizumab and aflibercept. Conclusion All three anti-VEGF agents are taken up by platelets and internalized in alpha-granules, which may result in a higher local exposure of anti-VEGF after the activation of platelets, potentially contributing to arterial thromboembolic events. Clathrin-coated vesicles seem to be more prominent in the transport of bevacizumab than ranibizumab and aflibercept. Nevertheless, whether the different localization and transport of bevacizumab are truly related to specific differences of receptor-mediated endocytosis has to be revealed by further research.
Collapse
|
10
|
Sheth V, Wang L, Bhattacharya R, Mukherjee P, Wilhelm S. Strategies for Delivering Nanoparticles across Tumor Blood Vessels. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007363. [PMID: 37197212 PMCID: PMC10187772 DOI: 10.1002/adfm.202007363] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Indexed: 05/19/2023]
Abstract
Nanoparticle transport across tumor blood vessels is a key step in nanoparticle delivery to solid tumors. However, the specific pathways and mechanisms of this nanoparticle delivery process are not fully understood. Here, the biological and physical characteristics of the tumor vasculature and the tumor microenvironment are explored and how these features affect nanoparticle transport across tumor blood vessels is discussed. The biological and physical methods to deliver nanoparticles into tumors are reviewed and paracellular and transcellular nanoparticle transport pathways are explored. Understanding the underlying pathways and mechanisms of nanoparticle tumor delivery will inform the engineering of safer and more effective nanomedicines for clinical translation.
Collapse
Affiliation(s)
- Vinit Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, 173 Felgar St, Norman, OK 73019, USA
| | - Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, 173 Felgar St, Norman, OK 73019, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, 800 NE 10th St, Oklahoma City, OK 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, 800 NE 10th St, Oklahoma City, OK 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, 173 Felgar St, Norman, OK 73019, USA
| |
Collapse
|
11
|
Influence of FcRn binding properties on the gastrointestinal absorption and exposure profile of Fc molecules. Bioorg Med Chem 2021; 32:115942. [PMID: 33461147 DOI: 10.1016/j.bmc.2020.115942] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 01/26/2023]
Abstract
The neonatal Fc receptor (FcRn) represents a transport system with the potential to facilitate absorption of biologics across the gastrointestinal barrier. How biologics interact with FcRn to enable their gastrointestinal absorption, and how these interactions might be optimized in a biological therapeutic are not well understood. Thus, we studied the absorption of Fc molecules from the intestine using three IgG4-derived Fc variants with different, pH-dependent FcRn binding and release profiles. Using several different intestinal models, we consistently observed that FcRn binding affinity correlated with transcytosis. Our findings support targeting FcRn to enable intestinal absorption of biologics and highlight additional strategic considerations for future work.
Collapse
|
12
|
Gklinos P, Papadopoulou M, Stanulovic V, Mitsikostas DD, Papadopoulos D. Monoclonal Antibodies as Neurological Therapeutics. Pharmaceuticals (Basel) 2021; 14:ph14020092. [PMID: 33530460 PMCID: PMC7912592 DOI: 10.3390/ph14020092] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 02/08/2023] Open
Abstract
Over the last 30 years the role of monoclonal antibodies in therapeutics has increased enormously, revolutionizing treatment in most medical specialties, including neurology. Monoclonal antibodies are key therapeutic agents for several neurological conditions with diverse pathophysiological mechanisms, including multiple sclerosis, migraines and neuromuscular disease. In addition, a great number of monoclonal antibodies against several targets are being investigated for many more neurological diseases, which reflects our advances in understanding the pathogenesis of these diseases. Untangling the molecular mechanisms of disease allows monoclonal antibodies to block disease pathways accurately and efficiently with exceptional target specificity, minimizing non-specific effects. On the other hand, accumulating experience shows that monoclonal antibodies may carry class-specific and target-associated risks. This article provides an overview of different types of monoclonal antibodies and their characteristics and reviews monoclonal antibodies currently in use or under development for neurological disease.
Collapse
Affiliation(s)
- Panagiotis Gklinos
- Department of Neurology, KAT General Hospital of Attica, 14561 Athens, Greece;
| | - Miranta Papadopoulou
- Center for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Vid Stanulovic
- Global Pharmacovigilance, R&D Sanofi, 91385 Chilly-Mazarin, France;
| | - Dimos D. Mitsikostas
- 1st Neurology Department, Aeginition Hospital, National and Kapodistrian University of Athens, 11521 Athens, Greece;
| | - Dimitrios Papadopoulos
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 129 Vasilissis Sophias Avenue, 11521 Athens, Greece
- Salpetriere Neuropsychiatric Clinic, 149 Papandreou Street, Metamorphosi, 14452 Athens, Greece
- Correspondence:
| |
Collapse
|
13
|
Sihombing MAEM, Safitri M, Zhou T, Wang L, McGinty S, Zhang HJ, Yin Y, Peng Q, Qiu J, Wang G. Unexpected Role of Nonimmune Cells: Amateur Phagocytes. DNA Cell Biol 2021; 40:157-171. [PMID: 33439750 DOI: 10.1089/dna.2020.5647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Effective and efficient efferocytosis of dead cells and associated cellular debris are critical to tissue homeostasis and healing of injured tissues. This important task was previously thought to be restricted to professional phagocytes (PPs). However, accumulating evidence has revealed another type of phagocyte, the amateur phagocyte (AP), which can also participate in efferocytosis. APs are non-myeloid progenitor/nonimmune cells that include differentiated cells (e.g., epithelial cells, fibroblasts, and endothelial cells [ECs]) and stem cells (e.g., neuronal progenitor cells and mesenchymal cells) and can be found throughout the human body. Studies have shown that APs have two prominent roles: identifying and removing dead cells presumably before PPs reach the site of injury and assisting PPs in the removal of cell corpses and the resolution of inflamed tissue. With respect to the engulfment and degradation of dead cells, APs are slower and less efficient than PPs. However, APs are fundamental to preventing the spread of inflammation over a large area. In this review, we present the diversity and characteristics of healthy and non-neoplastic APs in mammals. We also propose a hypothetical mechanism of the efferocytosis of immunoglobulin G (IgG)-opsonized myelin debris by ECs (APs). Furthermore, the ingestion and clearance of dead cells can induce proinflammatory or anti-inflammatory cytokine production, endothelial activation, and cellular fate transition, which contribute to the progression of disease. An understanding of the role of APs is necessary to develop effective intervention strategies, including potential molecular targets for clinical diagnosis and drug development, for inflammation-related diseases.
Collapse
Affiliation(s)
- Maic Audo Eybi Mayer Sihombing
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Maharani Safitri
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Tian Zhou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Lu Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Sean McGinty
- Division of Biomedical Engineering, University of Glasgow, Glasgow, United Kingdom
| | - Hai-Jun Zhang
- Department of Vascular and Intervention, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Yuxia Yin
- National United Engineering Laboratory for Biomedical Material Modification, Denzhou, China
| | - Qin Peng
- Institute of Systems and Physical Biology, Shenzen Bay Laboratory, Shenzhen, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
14
|
Fc Engineering Strategies to Advance IgA Antibodies as Therapeutic Agents. Antibodies (Basel) 2020; 9:antib9040070. [PMID: 33333967 PMCID: PMC7768499 DOI: 10.3390/antib9040070] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/26/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
Abstract
In the past three decades, a great interest has arisen in the use of immunoglobulins as therapeutic agents. In particular, since the approval of the first monoclonal antibody Rituximab for B cell malignancies, the progress in the antibody-related therapeutic agents has been incremental. Therapeutic antibodies can be applied in a variety of diseases, ranging from cancer to autoimmunity and allergy. All current therapeutic monoclonal antibodies used in the clinic are of the IgG isotype. IgG antibodies can induce the killing of cancer cells by growth inhibition, apoptosis induction, complement activation (CDC) or antibody-dependent cellular cytotoxicity (ADCC) by NK cells, antibody-dependent cellular phagocytosis (ADCP) by monocytes/macrophages, or trogoptosis by granulocytes. To enhance these effector mechanisms of IgG, protein and glyco-engineering has been successfully applied. As an alternative to IgG, antibodies of the IgA isotype have been shown to be very effective in tumor eradication. Using the IgA-specific receptor FcαRI expressed on myeloid cells, IgA antibodies show superior tumor-killing compared to IgG when granulocytes are employed. However, reasons why IgA has not been introduced in the clinic yet can be found in the intrinsic properties of IgA posing several technical limitations: (1) IgA is challenging to produce and purify, (2) IgA shows a very heterogeneous glycosylation profile, and (3) IgA has a relatively short serum half-life. Next to the technical challenges, pre-clinical evaluation of IgA efficacy in vivo is not straightforward as mice do not naturally express the FcαR. Here, we provide a concise overview of the latest insights in these engineering strategies overcoming technical limitations of IgA as a therapeutic antibody: developability, heterogeneity, and short half-life. In addition, alternative approaches using IgA/IgG hybrid and FcαR-engagers and the impact of engineering on the clinical application of IgA will be discussed.
Collapse
|
15
|
Kim D, Lee J, Bae I, Kim M, Huh Y, Choi J, Bae S, Choi IY, Kim HH, Kim DK. Preparation, characterization, and pharmacological study of a novel long-acting FGF21 with a potential therapeutic effect in obesity. Biologicals 2020; 69:49-58. [PMID: 33277119 DOI: 10.1016/j.biologicals.2020.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/22/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
FGF21 (Fibroblast Growth Factor 21), which is expressed in the liver, adipose tissue, and pancreas, has been widely known as a therapeutic candidate for metabolic diseases. Though FGF21 is crucial to glucose, lipid, and energy homeostasis, it is not straightforward to develop a new drug with FGF21 due to its short half-life in serum. Here, we derived a novel long-acting FGF21 (LAPS-FGF21), which is chemically conjugated to the human IgG4 Fc fragment for longer half-life in serum. The recombinant human IgG4 Fc fragment and FGF21 were prepared by the refolding of inclusion body and periplasmic expression in Escherichia coli overexpression systems, respectively. The efficacy study of LAPS-FGF21 in a Diet-Induced Obesity (DIO) mouse model revealed that LAPS-FGF21 reduced body weight effectively accompanied by improved glucose tolerance in a dose-dependent manner. The administration of LAPS-FGF21 also improved the blood profiles with a significant reduction in cholesterol and triglyceride levels. Additionally, the pharmacokinetic (PK) studies of LAPS-FGF21 using normal ICR mice demonstrated that the half-life of LAPS-FGF21 was approximately 64-fold longer than FGF21. Taken together, the LAPS-FGF21 could be a feasible drug candidate with excellent bodyweight loss efficacy and longer dosing interval by half-life increase in serum.
Collapse
Affiliation(s)
- Daejin Kim
- Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea; Hanmi Research Center, Hanmi Pharm. Co.Ltd, 550, Dongtangiheung-ro, Dongtan-myeon, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Jongsoo Lee
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea; Hanmi Research Center, Hanmi Pharm. Co.Ltd, 550, Dongtangiheung-ro, Dongtan-myeon, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - InHwan Bae
- Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea; Hanmi Research Center, Hanmi Pharm. Co.Ltd, 550, Dongtangiheung-ro, Dongtan-myeon, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Minyoung Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea; Hanmi Research Center, Hanmi Pharm. Co.Ltd, 550, Dongtangiheung-ro, Dongtan-myeon, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Youngho Huh
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea; Hanmi Research Center, Hanmi Pharm. Co.Ltd, 550, Dongtangiheung-ro, Dongtan-myeon, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Jaehyuk Choi
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea; Hanmi Research Center, Hanmi Pharm. Co.Ltd, 550, Dongtangiheung-ro, Dongtan-myeon, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Sungmin Bae
- Hanmi Research Center, Hanmi Pharm. Co.Ltd, 550, Dongtangiheung-ro, Dongtan-myeon, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - In Young Choi
- Hanmi Research Center, Hanmi Pharm. Co.Ltd, 550, Dongtangiheung-ro, Dongtan-myeon, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Ha Hyung Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Dae Kyong Kim
- Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
16
|
Bteich M. An overview of albumin and alpha-1-acid glycoprotein main characteristics: highlighting the roles of amino acids in binding kinetics and molecular interactions. Heliyon 2019; 5:e02879. [PMID: 31844752 PMCID: PMC6895661 DOI: 10.1016/j.heliyon.2019.e02879] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 01/04/2019] [Accepted: 11/15/2019] [Indexed: 12/12/2022] Open
Abstract
Although Albumin (ALB) and alpha-1-acid glycoprotein (AGP) have distinctive structural and functional characteristics, they both play a key role in binding a large variety of endogenous and exogenous ligands. An extensive binding to these plasma proteins could have a potential impact on drugs disposition (e.g. bioavailability, distribution and clearance), on their innocuity and their efficacy. This review summarizes the common knowledge about the structural and molecular characteristics of both ALB and AGP in humans, and about the most involved amino acids in their high-affinity binding pockets. However, the variability in residues found in binding pockets, for the same species, allows each plasma protein to interact differently with the ligands. The protein-ligand interaction influences differently the disposition of drugs that bind to either of these plasma proteins. The content of this review is useful for the design of new drug entities with high-binding characteristics, in qualitative and quantitative modelling (e.g. in vitro-in vivo extrapolations, 3D molecular docking, interspecies extrapolations), and for other interdisciplinary research.
Collapse
Affiliation(s)
- Michel Bteich
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
17
|
Mahalingaiah PK, Ciurlionis R, Durbin KR, Yeager RL, Philip BK, Bawa B, Mantena SR, Enright BP, Liguori MJ, Van Vleet TR. Potential mechanisms of target-independent uptake and toxicity of antibody-drug conjugates. Pharmacol Ther 2019; 200:110-125. [DOI: 10.1016/j.pharmthera.2019.04.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/19/2019] [Indexed: 12/13/2022]
|
18
|
Didona D, Maglie R, Eming R, Hertl M. Pemphigus: Current and Future Therapeutic Strategies. Front Immunol 2019; 10:1418. [PMID: 31293582 PMCID: PMC6603181 DOI: 10.3389/fimmu.2019.01418] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 06/05/2019] [Indexed: 12/16/2022] Open
Abstract
Pemphigus encompasses a heterogeneous group of autoimmune blistering diseases, which affect both mucous membranes and the skin. The disease usually runs a chronic-relapsing course, with a potentially devastating impact on the patients' quality of life. Pemphigus pathogenesis is related to IgG autoantibodies targeting various adhesion molecules in the epidermis, including desmoglein (Dsg) 1 and 3, major components of desmosomes. The pathogenic relevance of such autoantibodies has been largely demonstrated experimentally. IgG autoantibody binding to Dsg results in loss of epidermal keratinocyte adhesion, a phenomenon referred to as acantholysis. This in turn causes intra-epidermal blistering and the clinical appearance of flaccid blisters and erosions at involved sites. Since the advent of glucocorticoids, the overall prognosis of pemphigus has largely improved. However, mortality persists elevated, since long-term use of high dose corticosteroids and adjuvant steroid-sparing immunosuppressants portend a high risk of serious adverse events, especially infections. Recently, rituximab, a chimeric anti CD20 monoclonal antibody which induces B-cell depletion, has been shown to improve patients' survival, as early rituximab use results in higher disease remission rates, long term clinical response and faster prednisone tapering compared to conventional immunosuppressive therapies, leading to its approval as a first line therapy in pemphigus. Other anti B-cell therapies targeting B-cell receptor or downstream molecules are currently tried in clinical studies. More intriguingly, a preliminary study in a preclinical mouse model of pemphigus has shown promise regarding future therapeutic application of Chimeric Autoantibody Receptor T-cells engineered using Dsg domains to selectively target autoreactive B-cells. Conversely, previous studies from our group have demonstrated that B-cell depletion in pemphigus resulted in secondary impairment of T-cell function; this may account for the observed long-term remission following B-cell recovery in rituximab treated patients. Likewise, our data support the critical role of Dsg-specific T-cell clones in orchestrating the inflammatory response and B-cell activation in pemphigus. Monitoring autoreactive T-cells in patients may indeed provide further information on the role of these cells, and would be the starting point for designating therapies aimed at restoring the lost immune tolerance against Dsg. The present review focuses on current advances, unmet challenges and future perspectives of pemphigus management.
Collapse
Affiliation(s)
- Dario Didona
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| | - Roberto Maglie
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany.,Surgery and Translational Medicine, Section of Dermatology, University of Florence, Florence, Italy.,Section of Dermatology, Departement of Health Sciences, University of Florence, Florence, Italy
| | - Rüdiger Eming
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| | - Michael Hertl
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| |
Collapse
|
19
|
Shen X, Wang L, Xu C, Yang J, Peng R, Hu X, Wang F, Zheng H, Lao X. Fusion of thymosin alpha 1 with mutant IgG1 CH3 prolongs half-life and enhances antitumor effects in vivo. Int Immunopharmacol 2019; 74:105662. [PMID: 31220695 DOI: 10.1016/j.intimp.2019.05.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/13/2019] [Accepted: 05/23/2019] [Indexed: 10/26/2022]
Abstract
Thymosin alpha 1 (Tα1) is an immunomodulatory polypeptide secreted from the thymus. Tα1 has a wide range of biological functions, such as immunomodulation and endocrine regulation. Tα1 also displays antiviral and antitumor activities. Tα1 has been successfully used in clinical adjuvant therapy for solid tumors to improve the immune response of patients undergoing chemotherapy and radiotherapy. However, the half-life of Tα1 in the body is short, so frequent administration is required to maintain efficacy. In order to improve the pharmacokinetic profile of Tα1, we linked the mutated CH3 (mCH3) fragment of IgG1 (human) to the C-terminus of Tα1 to produce a long-acting fusion protein, Tα1-mCH3. The half-life of Tα1-mCH3 (47 h) was substantially increased compared with that of the parent molecule Tα1 (3 h). In vivo studies indicated that mCH3 fusion retained the original biological activity of Tα1, and Tα1-mCH3 showed slightly better immunomodulatory effect than Ta1. In the 4 T1 and B16F10 tumor xenograft models, Tα1-mCH3 induced a greater abundance of CD4+ and CD8+ T-cells in tumor tissues compared with Ta1. Tα1-mCH3 exhibited better effect in promoting the production of IL-2 and IFN-γ compared with Tα1. Therefore, Tα1-mCH3 more efficiently inhibited the growth of 4 T1 and B16F10 tumors than Tα1. In conclusion, fusion with mCH3 is an attractive strategy to lengthen the half-life and increase the activity of Tα1.
Collapse
Affiliation(s)
- Xutong Shen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Liping Wang
- Department of Clinical Oncology, the First City Hospital of Chenzhou, Hunan 423000, PR China
| | - Caoying Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jiahui Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Renhao Peng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xinyi Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Fanwen Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Heng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
20
|
Offeddu GS, Haase K, Gillrie MR, Li R, Morozova O, Hickman D, Knutson CG, Kamm RD. An on-chip model of protein paracellular and transcellular permeability in the microcirculation. Biomaterials 2019; 212:115-125. [PMID: 31112823 DOI: 10.1016/j.biomaterials.2019.05.022] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/23/2019] [Accepted: 05/10/2019] [Indexed: 12/19/2022]
Abstract
Recent therapeutic success of large-molecule biologics has led to intense interest in assays to measure with precision their transport across the vascular endothelium and into the target tissue. Most current in vitro endothelial models show unrealistically large permeability coefficients due to a non-physiological paracellular transport. Thus, more advanced systems are required to better recapitulate and discern the important contribution of transcellular transport (transcytosis), particularly of pharmaceutically-relevant proteins. Here, a robust platform technology for the measurement of transport through a human endothelium is presented, which utilizes in vitro microvascular networks (MVNs). The self-assembled MVNs recapitulate the morphology and junctional complexity of in vivo capillaries, and express key endothelial vesicular transport proteins. This results in measured permeabilities to large molecules comparable to those observed in vivo, which are orders of magnitude lower than those measured in transwells. The permeability of albumin and immunoglobulin G (IgG), biopharmaceutically-relevant proteins, is shown to occur primarily via transcytosis, with passage of IgG regulated by the receptor FcRn. The physiological relevance of the MVNs make it a valuable tool to assess the distribution of biopharmaceuticals into tissues, and may be used to prioritize candidate molecules from this increasingly important class of therapeutics.
Collapse
Affiliation(s)
- Giovanni S Offeddu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kristina Haase
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mark R Gillrie
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ran Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Olga Morozova
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA; Amgen Research, Amgen Inc., 360 Binney Street, Cambridge, MA, USA
| | - Dean Hickman
- Amgen Research, Amgen Inc., 360 Binney Street, Cambridge, MA, USA
| | | | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
21
|
Datta-Mannan A. Mechanisms Influencing the Pharmacokinetics and Disposition of Monoclonal Antibodies and Peptides. Drug Metab Dispos 2019; 47:1100-1110. [PMID: 31043438 DOI: 10.1124/dmd.119.086488] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/22/2019] [Indexed: 12/15/2022] Open
Abstract
Monoclonal antibodies (mAbs) and peptides are an important class of therapeutic modalities that have brought improved health outcomes in areas with limited therapeutic optionality. Presently, there more than 90 mAb and peptide therapeutics on the United States market, with over 600 more in various clinical stages of development in a broad array of therapeutic areas, including diabetes, autoimmune disorders, oncology, neuroscience, and cardiovascular and infectious diseases. Notwithstanding this potential, there is high clinical rate of attrition, with approximately 10% reaching patients. A major contributor to the failure of the molecules is often times an incomplete or poor understanding of the pharmacokinetics (PK) and disposition profiles leading to limited or diminished efficacy. Increased and thorough characterization efforts directed at disseminating mechanisms influencing the PK and disposition of mAbs and peptides can aid in improving the design for their intended pharmacological activity, and thereby their clinical success. The PK and disposition factors for mAbs and peptides are broadly influenced by target-mediated drug disposition and nontarget-related clearance mechanisms related to the interplay between the relationship of the structure and physiochemical properties of mAbs and peptides with physiologic processes. This review focuses on nontarget-related factors influencing the disposition and PK of mAbs and peptides. Contemporary considerations around the increasing in silico approaches to identify nontarget-related molecule limitations and enhancing the druggability of mAbs and peptides, including parenteral and nonparenteral delivery strategies that are geared toward improving patient experience and compliance, are also discussed.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Department of Experimental Medicine and Pharmacology, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana
| |
Collapse
|
22
|
Chung S, Nguyen V, Lin YL, Lafrance-Vanasse J, Scales SJ, Lin K, Deng R, Williams K, Sperinde G, Li JJ, Zheng K, Sukumaran S, Tesar D, Ernst JA, Fischer S, Lazar GA, Prabhu S, Song A. An in vitro FcRn- dependent transcytosis assay as a screening tool for predictive assessment of nonspecific clearance of antibody therapeutics in humans. MAbs 2019; 11:942-955. [PMID: 30982394 PMCID: PMC6601550 DOI: 10.1080/19420862.2019.1605270] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
A cell-based assay employing Madin–Darby canine kidney cells stably expressing human neonatal Fc receptor (FcRn) heavy chain and β2-microglobulin genes was developed to measure transcytosis of monoclonal antibodies (mAbs) under conditions relevant to the FcRn-mediated immunoglobulin G (IgG) salvage pathway. The FcRn-dependent transcytosis assay is modeled to reflect combined effects of nonspecific interactions between mAbs and cells, cellular uptake via pinocytosis, pH-dependent interactions with FcRn, and dynamics of intracellular trafficking and sorting mechanisms. Evaluation of 53 mAbs, including 30 marketed mAb drugs, revealed a notable correlation between the transcytosis readouts and clearance in humans. FcRn was required to promote efficient transcytosis of mAbs and contributed directly to the observed correlation. Furthermore, the transcytosis assay correctly predicted rank order of clearance of glycosylation and Fv charge variants of Fc-containing proteins. These results strongly support the utility of this assay as a cost-effective and animal-sparing screening tool for evaluation of mAb-based drug candidates during lead selection, optimization, and process development for desired pharmacokinetic properties.
Collapse
Affiliation(s)
- Shan Chung
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Van Nguyen
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Yuwen Linda Lin
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | | | - Suzie J Scales
- c Department of Molecular Biology , Genentech Inc ., South San Francisco , CA , USA
| | - Kevin Lin
- d Department of Analytical Operations , Genentech Inc ., South San Francisco , CA , USA
| | - Rong Deng
- e Department of Clinical Pharmacology , Genentech Inc ., South San Francisco , CA , USA
| | - Kathi Williams
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Gizette Sperinde
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Juan Jenny Li
- f Department of Biochemistry and Cellular Pharmacology , Genentech Inc ., South San Francisco , CA , USA
| | - Kai Zheng
- g Department of Late Stage Pharmaceutical Development , Genentech Inc ., South San Francisco , CA , USA
| | - Siddharth Sukumaran
- h Department of Pharmacokinetics & Pharmacodynamics , Genentech Inc ., South San Francisco , CA , USA
| | - Devin Tesar
- i Department of Drug Delivery , Genentech Inc ., South San Francisco , CA , USA
| | - James A Ernst
- b Department of Protein Chemistry , Genentech Inc ., South San Francisco , CA , USA
| | - Saloumeh Fischer
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Greg A Lazar
- j Department of Antibody Engineering , Genentech Inc ., South San Francisco , CA , USA
| | - Saileta Prabhu
- h Department of Pharmacokinetics & Pharmacodynamics , Genentech Inc ., South San Francisco , CA , USA
| | - An Song
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| |
Collapse
|
23
|
Neiveyans M, Melhem R, Arnoult C, Bourquard T, Jarlier M, Busson M, Laroche A, Cerutti M, Pugnière M, Ternant D, Gaborit N, Chardès T, Poupon A, Gouilleux-Gruart V, Pèlegrin A, Poul MA. A recycling anti-transferrin receptor-1 monoclonal antibody as an efficient therapy for erythroleukemia through target up-regulation and antibody-dependent cytotoxic effector functions. MAbs 2019; 11:593-605. [PMID: 30604643 PMCID: PMC6512944 DOI: 10.1080/19420862.2018.1564510] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Targeting transferrin receptor 1 (TfR1) with monoclonal antibodies is a promising therapeutic strategy in cancer as tumor cells often overexpress TfR1 and show increased iron needs. We have re-engineered six anti-human TfR1 single-chain variable fragment (scFv) antibodies into fully human scFv2-Fcγ1 and IgG1 antibodies. We selected the more promising candidate (H7), based on its ability to inhibit TfR1-mediated iron-loaded transferrin internalization in Raji cells (B-cell lymphoma). The H7 antibody displayed nanomolar affinity for its target in both formats (scFv2-Fcγ1 and IgG1), but cross-reacted with mouse TfR1 only in the scFv2-Fc format. H7 reduced the intracellular labile iron pool and, contrary to what has been observed with previously described anti-TfR1 antibodies, upregulated TfR1 level in Raji cells. H7 scFv2-Fc format elimination half-life was similar in FcRn knock-out and wild type mice, suggesting that TfR1 recycling contributes to prevent H7 elimination in vivo. In vitro, H7 inhibited the growth of erythroleukemia and B-cell lymphoma cell lines (IC50 0.1 µg/mL) and induced their apoptosis. Moreover, the Im9 B-cell lymphoma cell line, which is resistant to apoptosis induced by rituximab (anti-CD20 antibody), was sensitive to H7. In vivo, tumor regression was observed in nude mice bearing ERY-1 erythroleukemia cell xenografts treated with H7 through a mechanism that involved iron deprivation and antibody-dependent cytotoxic effector functions. Therefore, targeting TfR1 using the fully human anti-TfR1 H7 is a promising tool for the treatment of leukemia and lymphoma.
Collapse
Affiliation(s)
- Madeline Neiveyans
- a IRCM, Institut de Recherche en Cancérologie de Montpellier ; INSERM, U1194, Université de Montpellier, Montpellier , France.,b ICM , Institut régional du Cancer de Montpellier , Montpellier , France
| | - Rana Melhem
- a IRCM, Institut de Recherche en Cancérologie de Montpellier ; INSERM, U1194, Université de Montpellier, Montpellier , France.,b ICM , Institut régional du Cancer de Montpellier , Montpellier , France
| | - Christophe Arnoult
- c CNRS , GICC UMR 7292 , Tours , France.,d Université François Rabelais de Tours , Tours , France
| | - Thomas Bourquard
- e UMR INRA CNRS Physiologie de la reproduction et des comportements, Université François Rabelais de Tours , Nouzilly , France
| | - Marta Jarlier
- b ICM , Institut régional du Cancer de Montpellier , Montpellier , France
| | - Muriel Busson
- a IRCM, Institut de Recherche en Cancérologie de Montpellier ; INSERM, U1194, Université de Montpellier, Montpellier , France.,b ICM , Institut régional du Cancer de Montpellier , Montpellier , France
| | - Adrien Laroche
- a IRCM, Institut de Recherche en Cancérologie de Montpellier ; INSERM, U1194, Université de Montpellier, Montpellier , France.,b ICM , Institut régional du Cancer de Montpellier , Montpellier , France
| | | | - Martine Pugnière
- g PP2I, Plateforme Protéomique et Interactions Moléculaires , IRCM
| | - David Ternant
- c CNRS , GICC UMR 7292 , Tours , France.,d Université François Rabelais de Tours , Tours , France.,h CHRU de Tours, Department of medical pharmacology , Tours , France
| | - Nadège Gaborit
- a IRCM, Institut de Recherche en Cancérologie de Montpellier ; INSERM, U1194, Université de Montpellier, Montpellier , France.,b ICM , Institut régional du Cancer de Montpellier , Montpellier , France
| | - Thierry Chardès
- a IRCM, Institut de Recherche en Cancérologie de Montpellier ; INSERM, U1194, Université de Montpellier, Montpellier , France.,b ICM , Institut régional du Cancer de Montpellier , Montpellier , France
| | - Anne Poupon
- e UMR INRA CNRS Physiologie de la reproduction et des comportements, Université François Rabelais de Tours , Nouzilly , France
| | - Valérie Gouilleux-Gruart
- c CNRS , GICC UMR 7292 , Tours , France.,d Université François Rabelais de Tours , Tours , France
| | - Andre Pèlegrin
- a IRCM, Institut de Recherche en Cancérologie de Montpellier ; INSERM, U1194, Université de Montpellier, Montpellier , France.,b ICM , Institut régional du Cancer de Montpellier , Montpellier , France
| | - Marie-Alix Poul
- a IRCM, Institut de Recherche en Cancérologie de Montpellier ; INSERM, U1194, Université de Montpellier, Montpellier , France.,b ICM , Institut régional du Cancer de Montpellier , Montpellier , France
| |
Collapse
|
24
|
Hardiansyah D, Ng CM. Minimal physiologically-based pharmacokinetic model to investigate the effect of pH dependent FcRn affinity and the endothelial endocytosis on the pharmacokinetics of anti-VEGF humanized IgG1 antibody in cynomolgus monkey. Eur J Pharm Sci 2018; 125:130-141. [PMID: 30248389 DOI: 10.1016/j.ejps.2018.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 11/29/2022]
Abstract
In this study, we developed a first minimal physiologically-based pharmacokinetic (mPBPK) model to investigate the complex interaction effects of endocytosis rate/FcRn binding affinity at both acidic/physiological pH on the pharmacokinetics (PK) of the anti-VEGF IgG1 antibodies. The data used in this study were the PK of the native IgG and humanized anti-VEGF IgG1 antibodies with a wide range FcRn-binding at both acidic and physiological pH in the cynomolgus monkey. The basic structure of the developed mPBPK models consisted of plasma, tissue and lymph compartments. The tissue compartment was subdivided into vascular, endothelial and interstitial spaces. Non-equilibrium binding mechanism was used to describe the FcRn-IgG interaction in the endosome. The fittings in the final model with three pH systems in the endosome compartment showed a good fit based on the visualization of the fitted graphs and the coefficient of variations of the estimated parameters (CV < 50%). The quantitative endocytosis/FcRn binding affinity PK relationships was constructed using the final model to provide better understanding of complex interaction effects of endocytosis rate and FcRn binding on PK of anti-VEGF IgG1 antibodies. This result may serve as an important model-based drug discovery platform to guide the design and development of the future generation of anti-VEGF IgG1 or other therapeutic IgG1 antibodies. In addition, the mPBPK model developed in cynomolgus monkey was successfully used to predict the PK of the anti-VEGF IgG1 antibody (bevacizumab) in human subjects.
Collapse
Affiliation(s)
- Deni Hardiansyah
- College of Pharmacy, University of Kentucky, BioPharm Building, Room 341, 789 S. Limestone, Lexington, KY 40536, USA
| | - Chee Meng Ng
- College of Pharmacy, University of Kentucky, BioPharm Building, Room 341, 789 S. Limestone, Lexington, KY 40536, USA.
| |
Collapse
|
25
|
Wu H, Malone AF, Donnelly EL, Kirita Y, Uchimura K, Ramakrishnan SM, Gaut JP, Humphreys BD. Single-Cell Transcriptomics of a Human Kidney Allograft Biopsy Specimen Defines a Diverse Inflammatory Response. J Am Soc Nephrol 2018; 29:2069-2080. [PMID: 29980650 DOI: 10.1681/asn.2018020125] [Citation(s) in RCA: 279] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/05/2018] [Indexed: 01/11/2023] Open
Abstract
Background Single-cell genomics techniques are revolutionizing our ability to characterize complex tissues. By contrast, the techniques used to analyze renal biopsy specimens have changed little over several decades. We tested the hypothesis that single-cell RNA-sequencing can comprehensively describe cell types and states in a human kidney biopsy specimen.Methods We generated 8746 single-cell transcriptomes from a healthy adult kidney and a single kidney transplant biopsy core by single-cell RNA-sequencing. Unsupervised clustering analysis of the biopsy specimen was performed to identify 16 distinct cell types, including all of the major immune cell types and most native kidney cell types, in this biopsy specimen, for which the histologic read was mixed rejection.Results Monocytes formed two subclusters representing a nonclassical CD16+ group and a classic CD16- group expressing dendritic cell maturation markers. The presence of both monocyte cell subtypes was validated by staining of independent transplant biopsy specimens. Comparison of healthy kidney epithelial transcriptomes with biopsy specimen counterparts identified novel segment-specific proinflammatory responses in rejection. Endothelial cells formed three distinct subclusters: resting cells and two activated endothelial cell groups. One activated endothelial cell group expressed Fc receptor pathway activation and Ig internalization genes, consistent with the pathologic diagnosis of antibody-mediated rejection. We mapped previously defined genes that associate with rejection outcomes to single cell types and generated a searchable online gene expression database.Conclusions We present the first step toward incorporation of single-cell transcriptomics into kidney biopsy specimen interpretation, describe a heterogeneous immune response in mixed rejection, and provide a searchable resource for the scientific community.
Collapse
Affiliation(s)
- Haojia Wu
- Division of Nephrology, Department of Medicine and Departments of
| | - Andrew F Malone
- Division of Nephrology, Department of Medicine and Departments of
| | - Erinn L Donnelly
- Division of Nephrology, Department of Medicine and Departments of
| | - Yuhei Kirita
- Division of Nephrology, Department of Medicine and Departments of
| | - Kohei Uchimura
- Division of Nephrology, Department of Medicine and Departments of
| | | | | | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine and Departments of .,Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| |
Collapse
|
26
|
Datta-Mannan A, Boyles J, Huang L, Jin ZY, Peariso A, Murphy AT, Ellis B, Douglass N, Norouziyan-Cooper F, Witcher DR. Engineered FcRn Binding Fusion Peptides Significantly Enhance the Half-Life of a Fab Domain in Cynomolgus Monkeys. Biotechnol J 2018; 14:e1800007. [DOI: 10.1002/biot.201800007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/25/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Amita Datta-Mannan
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Jeffrey Boyles
- Department of Biotechnology Discovery Research; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Lihua Huang
- Department of Bioproduct Research/Development; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Zhaoyan Y. Jin
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Amber Peariso
- Department of Biotechnology Discovery Research; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Anthony T. Murphy
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Bernice Ellis
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Nicole Douglass
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Fariba Norouziyan-Cooper
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Derrick R. Witcher
- Department of Biotechnology Discovery Research; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| |
Collapse
|
27
|
Yang H, Feng Y, Cai H, Jia D, Li H, Tao Z, Zhong Y, Li Z, Shi Q, Wan L, Li L, Lu X. Endogenous IgG-based affinity-controlled release of TRAIL exerts superior antitumor effects. Am J Cancer Res 2018; 8:2459-2476. [PMID: 29721092 PMCID: PMC5928902 DOI: 10.7150/thno.23880] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/08/2018] [Indexed: 02/05/2023] Open
Abstract
The inefficiency of recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based clinical regimens has been dominantly attributed to the short half-life of TRAIL. Affinity-controlled release using endogenous long-acting proteins, such as IgG and albumin, as carriers is extremely attractive for improving the pharmacokinetics of TRAIL. Up to now, it is unclear whether IgG-binding is efficient for affinity-controlled release of TRAIL. Methods: An IgG-binding affibody, IgBD, was genetically fused to the N-terminus of TRAIL to produce IgBD-TRAIL.The IgG-binding ability, cytotoxicity, serum half-life, and in vivo antitumor effect of IgBD-TRAIL were compared with that of TRAIL. In addition, an albumin-binding affibody, ABD, was fused to TRAIL to produce ABD-TRAIL. The cytototoxicity, serum half-life, and antitumor effect of IgBD-TRAIL and ABD-TRAIL were compared. Results: IgBD fusion endowed TRAIL with high affinity (nM) for IgG without interference with its cytotoxicity. The serum half-life of IgBD-TRAIL is 50-60 times longer than that of TRAIL and the tumor uptake of IgBD-TRAIL at 8-24 h post-injection was 4-7-fold that of TRAIL. In vivo antitumor effect of IgBD-TRAIL was at least 10 times greater than that of TRAIL. Owing to the high affinity (nM) for albumin, the serum half-life of ABD-TRAIL was 80-90 times greater than that of TRAIL. However, after binding to albumin, the cytotoxicity of ABD-TRAIL was reduced more than 10 times. In contrast, binding to IgG had little impact on the cytotoxicity of IgBD-TRAIL. Consequently, intravenously injected IgBD-TRAIL showed antitumor effects superior to those of ABD-TRAIL. Conclusions: Endogenous long-acting proteins, particularly IgG-based affinity-controlled release, prolonged the serum half-life as well as significantly enhanced the antitumor effect of TRAIL. IgBD-mediated endogenous IgG binding might be a novel approach for the affinity-controlled release of other protein drugs.
Collapse
|
28
|
Devraj K, Guérit S, Macas J, Reiss Y. An In Vivo Blood-brain Barrier Permeability Assay in Mice Using Fluorescently Labeled Tracers. J Vis Exp 2018. [PMID: 29553506 DOI: 10.3791/57038] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Blood-brain barrier (BBB) is a specialized barrier that protects the brain microenvironment from toxins and pathogens in the circulation and maintains brain homeostasis. The principal sites of the barrier are endothelial cells of the brain capillaries whose barrier function results from tight intercellular junctions and efflux transporters expressed on the plasma membrane. This function is regulated by pericytes and astrocytes that together form the neurovascular unit (NVU). Several neurological diseases such as stroke, Alzheimer's disease (AD), brain tumors are associated with an impaired BBB function. Assessment of the BBB permeability is therefore crucial in evaluating the severity of the neurological disease and the success of the treatment strategies employed. We present here a simple yet robust permeability assay that have been successfully applied to several mouse models both, genetic and experimental. The method is highly quantitative and objective in comparison to the tracer fluorescence analysis by microscopy that is commonly applied. In this method, mice are injected intraperitoneally with a mix of aqueous inert fluorescent tracers followed by anesthetizing the mice. Cardiac perfusion of the animals is performed prior to harvesting brain, kidneys or other organs. Organs are homogenized and centrifuged followed by fluorescence measurement from the supernatant. Blood drawn from the cardiac puncture just before perfusion serves for normalization purpose to the vascular compartment. The tissue fluorescence is normalized to the wet weight and serum fluorescence to obtain a quantitative tracer permeability index. For additional confirmation, the contralateral hemi-brain preserved for immunohistochemistry can be utilized for tracer fluorescence visualization purposes.
Collapse
Affiliation(s)
- Kavi Devraj
- Institute of Neurology (Edinger Institute), Goethe University Hospital; Pharmazentrum Frankfurt, Institute for General Pharmacology and Toxicology, Goethe University Hospital
| | - Sylvaine Guérit
- Institute of Neurology (Edinger Institute), Goethe University Hospital
| | - Jakranka Macas
- Institute of Neurology (Edinger Institute), Goethe University Hospital
| | - Yvonne Reiss
- Institute of Neurology (Edinger Institute), Goethe University Hospital;
| |
Collapse
|
29
|
Grevys A, Nilsen J, Sand KMK, Daba MB, Øynebråten I, Bern M, McAdam MB, Foss S, Schlothauer T, Michaelsen TE, Christianson GJ, Roopenian DC, Blumberg RS, Sandlie I, Andersen JT. A human endothelial cell-based recycling assay for screening of FcRn targeted molecules. Nat Commun 2018; 9:621. [PMID: 29434196 PMCID: PMC5809500 DOI: 10.1038/s41467-018-03061-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 01/17/2018] [Indexed: 12/13/2022] Open
Abstract
Albumin and IgG have remarkably long serum half-lives due to pH-dependent FcRn-mediated cellular recycling that rescues both ligands from intracellular degradation. Furthermore, increase in half-lives of IgG and albumin-based therapeutics has the potential to improve their efficacies, but there is a great need for robust methods for screening of relative FcRn-dependent recycling ability. Here, we report on a novel human endothelial cell-based recycling assay (HERA) that can be used for such pre-clinical screening. In HERA, rescue from degradation depends on FcRn, and engineered ligands are recycled in a manner that correlates with their half-lives in human FcRn transgenic mice. Thus, HERA is a novel cellular assay that can be used to predict how FcRn-binding proteins are rescued from intracellular degradation.
Collapse
Affiliation(s)
- Algirdas Grevys
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Jeannette Nilsen
- CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, N-0450 Oslo, Norway
| | - Kine M K Sand
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Muluneh B Daba
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Inger Øynebråten
- Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo, N-0424, Oslo, Norway
| | - Malin Bern
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Martin B McAdam
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Stian Foss
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Tilman Schlothauer
- Biochemical and Analytical Research, Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center, DE-82377 Munich, Germany
| | - Terje E Michaelsen
- School of Pharmacy, University of Oslo, N-0371, Oslo, Norway.,Norwegian Institute of Public Health, Infection Immunology, N-0403, Oslo, Norway
| | | | | | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Inger Sandlie
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Jan Terje Andersen
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway. .,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway. .,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, N-0424, Oslo, Norway.
| |
Collapse
|
30
|
Two-Pore Minimum Physiologically-based Pharmacokinetic Model to Describe the Disposition of Therapeutic Monoclonal IgG Antibody in Humans. Pharm Res 2018; 35:47. [PMID: 29411151 DOI: 10.1007/s11095-017-2292-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/23/2017] [Indexed: 12/26/2022]
Abstract
PURPOSE The aim of this study was to develop a two-pore minimum physiologically-based pharmacokinetic (mPBPK) model in describing the pharmacokinetic (PK) of therapeutic monoclonal antibody (TMAb) in human subjects. METHODS PK data used in this study were endogenous/exogenous native IgG and two TMAbs (palivizumab and Motavizumab-YTE) in normal volunteer or familial hypercatabolic hypoproteinemia (FIHH) patient. Several important components were implemented to overcome the limitations of the early mPBPK model, e.g. two-pore model to describe the transcapillary transport of IgG from vascular to interstitial space. Six mPBPK models with different osmotic reflection coefficient (OFC) of transcapillary transport, endocytosis rates (ETR) and plasma clearance for the TMAbs/IgG were tested and the best model was selected using AICc values. RESULTS The final model consisted of different OFC and ETR values for native IgG and TMAbs, supporting the hypothesis that the dynamics in the endosomal space had an important role in the compliant FcRn salvage mechanism to determine the clearance of TMAbs. The estimated FcRn concentration of FIHH subjects was 2.72 μmol/l. The final two-pore mPBPK model has a better performance for native IgG than previously developed mPBPK model. CONCLUSIONS The final two-pore mPBPK model not only overcome the limitations of the early mPBPK model but also has a better performance to describe the disposition of the IgG antibody in human subjects.
Collapse
|
31
|
Ahmed AR, Carrozzo M, Caux F, Cirillo N, Dmochowski M, Alonso AE, Gniadecki R, Hertl M, López-Zabalza MJ, Lotti R, Pincelli C, Pittelkow M, Schmidt E, Sinha AA, Sprecher E, Grando SA. Monopathogenic vs multipathogenic explanations of pemphigus pathophysiology. Exp Dermatol 2018; 25:839-846. [PMID: 27305362 DOI: 10.1111/exd.13106] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2016] [Indexed: 01/31/2023]
Abstract
This viewpoint highlights major, partly controversial concepts about the pathogenesis of pemphigus. The monopathogenic theory explains intra-epidermal blistering through the "desmoglein (Dsg) compensation" hypothesis, according to which an antibody-dependent disabling of Dsg 1- and/or Dsg 3-mediated cell-cell attachments of keratinocytes (KCs) is sufficient to disrupt epidermal integrity and cause blistering. The multipathogenic theory explains intra-epidermal blistering through the "multiple hit" hypothesis stating that a simultaneous and synchronized inactivation of the physiological mechanisms regulating and/or mediating intercellular adhesion of KCs is necessary to disrupt epidermal integrity. The major premise for a multipathogenic theory is that a single type of autoantibody induces only reversible changes, so that affected KCs can recover due to a self-repair. The damage, however, becomes irreversible when the salvage pathway and/or other cell functions are altered by a partnering autoantibody and/or other pathogenic factors. Future studies are needed to (i) corroborate these findings, (ii) characterize in detail patient populations with non-Dsg-specific autoantibodies, and (iii) determine the extent of the contribution of non-Dsg antibodies in disease pathophysiology.
Collapse
Affiliation(s)
- A Razzaque Ahmed
- Department of Dermatology of Tufts University and Center for Blistering Diseases, Boston, MA, USA
| | - Marco Carrozzo
- School of Dental Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Frédéric Caux
- Department of Dermatology, University Paris 13, Avicenne Hospital, APHP, Bobigny, France
| | - Nicola Cirillo
- Melbourne Dental School and Oral Health CRC, The University of Melbourne, Melbourne, Vic., Australia
| | - Marian Dmochowski
- Autoimmune Blistering Dermatoses Section, Department of Dermatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Agustín España Alonso
- Department of Dermatology, School of Medicine, University Clinic of Navarra, University of Navarra, Navarra, Spain
| | - Robert Gniadecki
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - Michael Hertl
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| | | | - Roberta Lotti
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Pincelli
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Mark Pittelkow
- Department of Dermatology, Mayo Clinic, Scottsdale, AZ, USA
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Animesh A Sinha
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Eli Sprecher
- Department of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Sergei A Grando
- Institute for Immunology and Departments of Dermatology and Biological Chemistry, University of California, Irvine, CA, USA.
| |
Collapse
|
32
|
Stamou M, Grodzki AC, van Oostrum M, Wollscheid B, Lein PJ. Fc gamma receptors are expressed in the developing rat brain and activate downstream signaling molecules upon cross-linking with immune complex. J Neuroinflammation 2018; 15:7. [PMID: 29306331 PMCID: PMC5756609 DOI: 10.1186/s12974-017-1050-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Background Exposure of the developing brain to immune mediators, including antibodies, is postulated to increase risk for neurodevelopmental disorders and neurodegenerative disease. It has been suggested that immunoglobulin G-immune complexes (IgG-IC) activate Fc gamma receptors (FcγR) expressed on neurons to modify signaling events in these cells. However, testing this hypothesis is hindered by a paucity of data regarding neuronal FcγR expression and function. Methods FcγR transcript expression in the hippocampus, cortex, and cerebellum of neonatal male and female rats was investigated ex vivo and in mixed cultures of primary hippocampal and cortical neurons and astrocytes using quantitative PCR analyses. Expression at the protein level in mixed cultures of primary hippocampal and cortical neurons and astrocytes was determined by immunocytochemistry, western blotting, proteotype analysis, and flow cytometry. The functionality of these receptors was assessed by measuring changes in intracellular calcium levels, Erk phosphorylation, and IgG internalization following stimulation with IgG-immune complexes. Results FcgrIa, FcgrIIa, FcgrIIb, FcgrIIIa, and Fcgrt transcripts were detectable in the cortex, hippocampus, and cerebellum at postnatal days 1 and 7. These transcripts were also present in primary hippocampal and cortical cell cultures, where their expression was modulated by IFNγ. Expression of FcγRIa, FcγRIIb, and FcγRIIIa, but not FcγRIIa or FcRn proteins, was confirmed in cultured hippocampal and cortical neurons and astrocytes at the single cell level. A subpopulation of these cells co-expressed the activating FcγRIa and the inhibitory FcγRIIb. Functional analyses demonstrated that exposure of hippocampal and cortical cell cultures to IgG-IC increases intracellular calcium and Erk phosphorylation and triggers FcγR-mediated internalization of IgG. Conclusions Our data demonstrate that developing neurons and astrocytes in the hippocampus and the cortex express signaling competent FcγR. These findings suggest that IgG antibodies may influence normal neurodevelopment or function via direct interactions with FcγR on non-immune cells in the brain. Electronic supplementary material The online version of this article (10.1186/s12974-017-1050-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marianna Stamou
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Ana Cristina Grodzki
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Marc van Oostrum
- Department of Health Sciences and Technology, Institute of Molecular Systems Biology, ETH Zurich, 8093, Zürich, Switzerland
| | - Bernd Wollscheid
- Department of Health Sciences and Technology, Institute of Molecular Systems Biology, ETH Zurich, 8093, Zürich, Switzerland
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA.
| |
Collapse
|
33
|
Brady JM, Baltimore D, Balazs AB. Antibody gene transfer with adeno-associated viral vectors as a method for HIV prevention. Immunol Rev 2017; 275:324-333. [PMID: 28133808 DOI: 10.1111/imr.12478] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Broadly neutralizing antibodies (bNAbs) against human immunodeficiency virus (HIV) show great promise in HIV prevention as they are capable of potently neutralizing a considerable breadth of genetically diverse strains. Passive transfer of monoclonal bNAb proteins can confer protection in animal models of HIV infection at modest concentrations, inspiring efforts to develop an HIV vaccine capable of eliciting bNAb responses. However, these antibodies demonstrate high degrees of somatic mutation and other unique characteristics that may hinder the ability of conventional approaches to consistently and effectively produce bNAb analogs. As an alternative strategy, we and others have proposed vector-mediated gene transfer to generate long-term, systemic production of bNAbs in the absence of immunization. Herein, we review the use of adeno-associated virus (AAV) vectors for delivery of HIV bNAbs and antibody-like proteins and summarize both the advantages and disadvantages of this strategy as a method for HIV prevention.
Collapse
Affiliation(s)
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | |
Collapse
|
34
|
|
35
|
Nelms B, Dalomba NF, Lencer W. A targeted RNAi screen identifies factors affecting diverse stages of receptor-mediated transcytosis. J Cell Biol 2017; 216:511-525. [PMID: 28069747 PMCID: PMC5294788 DOI: 10.1083/jcb.201609035] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/22/2016] [Accepted: 12/20/2016] [Indexed: 11/22/2022] Open
Abstract
Transcytosis plays an important role in establishing cell polarity and in mediating transport of large cargo across epithelial barriers, but its molecular basis is unclear. Nelms et al. present a new dataset of genes involved in receptor-mediated transcytosis and show that the apical and basolateral recycling and transcytotic pathways are genetically separable. Endosome transport by transcytosis is the primary mechanism by which proteins and other large cargo traverse epithelial barriers in normal tissue. Transcytosis is also essential for establishing and maintaining membrane polarity in epithelia and other polarized cells. To identify novel components of this pathway, we conducted a high-throughput RNA interference screen for factors necessary for the bidirectional transcytosis of IgG by the Fcγ receptor FcRn. This screen identified 23 genes whose suppression resulted in a reproducible decrease in FcRn-mediated transcytosis. Pulse-chase kinetic transport assays on four of the top-ranking genes (EXOC2, EXOC7, PARD6B, and LEPROT) revealed distinct effects on the apical and basolateral recycling and transcytotic pathways, demonstrating that these pathways are genetically separable. We also found a strong dependence on PARD6B for apical, but not basolateral, recycling, implicating this cell polarity gene in assembly or maintenance of the apical endosomal system. This dataset yields insights into how vesicular transport is adapted to the specialized functions of differentiated cell types and opens new research avenues into epithelial trafficking.
Collapse
Affiliation(s)
- Bradlee Nelms
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115.,Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138
| | - Natasha Furtado Dalomba
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Wayne Lencer
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115 .,Harvard Digestive Diseases Center, Boston, MA 02115
| |
Collapse
|
36
|
Wang X, McKay P, Yee LT, Dutina G, Hass PE, Nijem I, Allison D, Cowan KJ, Lin K, Quarmby V, Yang J. Impact of SPR biosensor assay configuration on antibody: Neonatal Fc receptor binding data. MAbs 2016; 9:319-332. [PMID: 28001487 DOI: 10.1080/19420862.2016.1261774] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Binding interactions with the neonatal Fc receptor (FcRn) are one determinant of pharmacokinetic properties of recombinant human monoclonal antibody (rhumAb) therapeutics, and a conserved binding motif in the crystallizable fragment (Fc) region of IgG molecules interacts with FcRn. Surface plasmon resonance (SPR) biosensor assays are often used to characterize interactions between FcRn and rhumAb therapeutics. In such assays, generally either the rhumAb (format 1) or the FcRn protein (format 2) is immobilized on a biosensor chip. However, because evidence suggests that, in some cases, the variable domains of a rhumAb may also affect FcRn binding, we evaluated the effect of SPR assay configuration on binding data. We sought to assess FcRn binding properties of 2 rhumAbs (rhumAb1 and rhumAb2) to FcRn proteins using these 2 biosensor assay formats. The two rhumAbs have greater than 99% sequence identity in the Fc domain but differ in their Fab regions. rhumAb2 contains a positively charged patch in the variable domain that is absent in rhumAb1. Our results showed that binding of rhumAb1 to FcRn was independent of biosensor assay configuration, while binding of rhumAb2 to FcRn was highly SPR assay configuration dependent. Further investigations revealed that the format dependency of rhumAb2-FcRn binding is linked to the basic residues that form a positively charged patch in the variable domain of rhumAb2. Our work highlights the importance of analyzing rhumAb-FcRn binding interactions using 2 alternate SPR biosensor assay configurations. This approach may also provide a simple way to identify the potential for non-Fc-driven FcRn binding interactions in otherwise typical IgGs.
Collapse
Affiliation(s)
- Xiangdan Wang
- a BioAnalytical Sciences, Genentech , South San Francisco , CA , USA
| | - Patrick McKay
- b Technical Development, Genentech , South San Francisco , CA , USA
| | - Liliana T Yee
- b Technical Development, Genentech , South San Francisco , CA , USA
| | - George Dutina
- c Department of Early Stage Cell Culture , Genentech , South San Francisco , CA , USA
| | - Philip E Hass
- d Protein Chemistry, Genentech , South San Francisco , CA , USA
| | - Ihsan Nijem
- a BioAnalytical Sciences, Genentech , South San Francisco , CA , USA
| | - David Allison
- e Clinical Pharmacology, Genentech , South San Francisco , CA , USA
| | - Kyra J Cowan
- a BioAnalytical Sciences, Genentech , South San Francisco , CA , USA
| | - Kevin Lin
- f Analytical Operations, Genentech , South San Francisco , CA , USA
| | - Valerie Quarmby
- a BioAnalytical Sciences, Genentech , South San Francisco , CA , USA
| | - Jihong Yang
- a BioAnalytical Sciences, Genentech , South San Francisco , CA , USA
| |
Collapse
|
37
|
Deissler HL, Lang GK, Lang GE. Neonatal Fc receptor FcRn is involved in intracellular transport of the Fc fusion protein aflibercept and its transition through retinal endothelial cells. Exp Eye Res 2016; 154:39-46. [PMID: 27836572 DOI: 10.1016/j.exer.2016.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/18/2016] [Accepted: 11/07/2016] [Indexed: 12/15/2022]
Abstract
Retinal endothelial cells (REC) likely contribute to the clearance of intravitreally injected IgG. Because this is of high relevance to the pharmacokinetic assessment of the widely used therapeutic Fc fusion protein aflibercept, we studied its transport through immortalized bovine REC (iBREC) in detail. For shuttling of IgG or Fc fusion proteins like aflibercept, endothelial cells use the highly conserved neonatal Fc receptor (FcRn) also expressed in iBREC where it is down regulated by serum depletion. Therefore, we focused on studying intracellular localization and transport of aflibercept under conditions affecting its interaction with the FcRn. Intracellular localization of aflibercept was assessed by Western-blot analyses of subcellular protein fractions or by immunofluorescence staining. After uptake in a temperature-dependent process, aflibercept co-localized with early endosomes, which harbor FcRn. Similar amounts of aflibercept were co-extracted with proteins from membranes/organelles irrespectively of the amount of FBS in the culture medium. Lowering the concentration of FBS resulted in a strong, but reversible association with cytoskeletal proteins suggesting a block in intracellular transport. In accordance with this finding, aflibercept's transport through an iBREC monolayer grown on porous membrane inserts was markedly delayed in the absence of FBS in the culture medium indicating that aflibercept is taken up but not exocytosed under these conditions. Transcytosis of aflibercept was also strongly delayed by inhibition of phosphatidylinositol 3-kinase with LY294002, which affects FcRn-mediated IgG transport. A similar inhibition of aflibercept's transport was observed with IgG-binding proteins (i.e. protein A or protein G) that block interaction between FcRn and aflibercept. Interfering with aflibercept's binding to the FcRn with protein A (or protein G) or the inhibitory FcRn-specific monoclonal antibody 1G3 resulted in a reduced amount of intracellular aflibercept. Taken together, our results strongly suggest that FcRn is involved in transport of aflibercept through REC in vitro.
Collapse
Affiliation(s)
- Heidrun L Deissler
- Department of Ophthalmology, University of Ulm, Prittwitzstrasse 43, 89075 Ulm, Germany.
| | - Gerhard K Lang
- Department of Ophthalmology, University of Ulm, Prittwitzstrasse 43, 89075 Ulm, Germany
| | - Gabriele E Lang
- Department of Ophthalmology, University of Ulm, Prittwitzstrasse 43, 89075 Ulm, Germany
| |
Collapse
|
38
|
Bruce VJ, Ta AN, McNaughton BR. Minimalist Antibodies and Mimetics: An Update and Recent Applications. Chembiochem 2016; 17:1892-1899. [DOI: 10.1002/cbic.201600303] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Virginia J. Bruce
- Department of Chemistry; Colorado State University; Fort Collins CO 80523 USA
| | - Angeline N. Ta
- Department of Chemistry; Colorado State University; Fort Collins CO 80523 USA
| | - Brian R. McNaughton
- Department of Chemistry; Colorado State University; Fort Collins CO 80523 USA
- Department of Biochemistry and Molecular Biology; Colorado State University; Fort Collins CO 80523 USA
| |
Collapse
|
39
|
Frigo G, Tramentozzi E, Orso G, Ceolotto G, Pagetta A, Stagni C, Menin C, Rosato A, Finotti P. Human IgGs induce synthesis and secretion of IgGs and neonatal Fc receptor in human umbilical vein endothelial cells. Immunobiology 2016; 221:1329-1342. [PMID: 27523744 DOI: 10.1016/j.imbio.2016.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/16/2016] [Accepted: 08/04/2016] [Indexed: 11/17/2022]
Abstract
Human IgGs are increasingly used in the therapy of many different immune and inflammatory diseases, however their mechanism of action still remains unclear in most diseases. To gain insight into the mechanism by which IgGs might also exert their effects on endothelial cells, we tested human IgGs on human umbilical vein endothelial cells (HUVECs). IgGs induced a time-dependent increase in the synthesis and secretion of IgGs, together with a marked angiogenic-like transformation of HUVECs that was maximal after a 20-h incubation. IgGs stimulated IG gene transcription without affecting the process of gene rearrangement, already present in control HUVECs. The mechanism involved the activation of transcription factors with the increased expression of HSP90, HSP70 and inactive MMP-9 responsible for the phenotypic differentiation associated with the most intense IgG synthesis and secretion. However, even a short incubation with IgGs followed by recovery of cells was sufficient to trigger and sustain in time the synthesis and secretion of new IgGs, independently of the angiogenic-like transformation visible only when cells were continuously exposed to IgGs. Under the stimulus of IgGs, specific secretory pathways were also activated in HUVECs together with the expression of FcRn, which was always associated with IgGs of new synthesis, forming complexes that were also secreted. Our results disclose a so far unknown and unexpected mechanism of IgGs on HUVECs that behave as Ig-producing immune cells. Results might have relevance for the effects that IgGs also exert in vivo in physiological conditions.
Collapse
Affiliation(s)
- Giulia Frigo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Pharmacology Building, L.go E. Meneghetti 2, 35131 Padua, Italy
| | - Elisa Tramentozzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Pharmacology Building, L.go E. Meneghetti 2, 35131 Padua, Italy
| | - Genny Orso
- Scientific Institute, IRCCS Eugenio Medea, Conegliano, Treviso, Italy
| | - Giulio Ceolotto
- Department of Medicine, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Andrea Pagetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Pharmacology Building, L.go E. Meneghetti 2, 35131 Padua, Italy
| | - Camilla Stagni
- Department of Surgery Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, Padua, Italy
| | - Chiara Menin
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Via Gattamelata 64, Padua, Italy
| | - Antonio Rosato
- Department of Surgery Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, Padua, Italy; Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Via Gattamelata 64, Padua, Italy.
| | - Paola Finotti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Pharmacology Building, L.go E. Meneghetti 2, 35131 Padua, Italy.
| |
Collapse
|
40
|
Avery LB, Wang M, Kavosi MS, Joyce A, Kurz JC, Fan YY, Dowty ME, Zhang M, Zhang Y, Cheng A, Hua F, Jones HM, Neubert H, Polzer RJ, O'Hara DM. Utility of a human FcRn transgenic mouse model in drug discovery for early assessment and prediction of human pharmacokinetics of monoclonal antibodies. MAbs 2016; 8:1064-78. [PMID: 27232760 DOI: 10.1080/19420862.2016.1193660] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Therapeutic antibodies continue to develop as an emerging drug class, with a need for preclinical tools to better predict in vivo characteristics. Transgenic mice expressing human neonatal Fc receptor (hFcRn) have potential as a preclinical pharmacokinetic (PK) model to project human PK of monoclonal antibodies (mAbs). Using a panel of 27 mAbs with a broad PK range, we sought to characterize and establish utility of this preclinical animal model and provide guidance for its application in drug development of mAbs. This set of mAbs was administered to both hemizygous and homozygous hFcRn transgenic mice (Tg32) at a single intravenous dose, and PK parameters were derived. Higher hFcRn protein tissue expression was confirmed by liquid chromatography-high resolution tandem mass spectrometry in Tg32 homozygous versus hemizygous mice. Clearance (CL) was calculated using non-compartmental analysis and correlations were assessed to historical data in wild-type mouse, non-human primate (NHP), and human. Results show that mAb CL in hFcRn Tg32 homozygous mouse correlate with human (r(2) = 0.83, r = 0.91, p < 0.01) better than NHP (r(2) = 0.67, r = 0.82, p < 0.01) for this dataset. Applying simple allometric scaling using an empirically derived best-fit exponent of 0.93 enabled the prediction of human CL from the Tg32 homozygous mouse within 2-fold error for 100% of mAbs tested. Implementing the Tg32 homozygous mouse model in discovery and preclinical drug development to predict human CL may result in an overall decreased usage of monkeys for PK studies, enhancement of the early selection of lead molecules, and ultimately a decrease in the time for a drug candidate to reach the clinic.
Collapse
Affiliation(s)
- Lindsay B Avery
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Mengmeng Wang
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Mania S Kavosi
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Alison Joyce
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Jeffrey C Kurz
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Yao-Yun Fan
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Martin E Dowty
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Minlei Zhang
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Yiqun Zhang
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Aili Cheng
- b Pharmaceutical Sciences Analytical R&D, Pfizer Inc. , Andover , MA , USA
| | - Fei Hua
- c PharmaTherapeutics Clinical R&D, Pfizer Inc. , Technology Square, Cambridge , MA , USA
| | - Hannah M Jones
- d Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Cambridge , MA , USA
| | - Hendrik Neubert
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| | - Robert J Polzer
- d Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Cambridge , MA , USA
| | - Denise M O'Hara
- a Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc. , Andover , MA , USA
| |
Collapse
|
41
|
Wagner MC, Myslinski J, Pratap S, Flores B, Rhodes G, Campos-Bilderback SB, Sandoval RM, Kumar S, Patel M, Ashish, Molitoris BA. Mechanism of increased clearance of glycated albumin by proximal tubule cells. Am J Physiol Renal Physiol 2016; 310:F1089-102. [PMID: 26887834 PMCID: PMC4889321 DOI: 10.1152/ajprenal.00605.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/10/2016] [Indexed: 11/22/2022] Open
Abstract
Serum albumin is the most abundant plasma protein and has a long half-life due to neonatal Fc receptor (FcRn)-mediated transcytosis by many cell types, including proximal tubule cells of the kidney. Albumin also interacts with, and is modified by, many small and large molecules. Therefore, the focus of the present study was to address the impact of specific known biological albumin modifications on albumin-FcRn binding and cellular handling. Binding at pH 6.0 and 7.4 was performed since FcRn binds albumin strongly at acidic pH and releases it after transcytosis at physiological pH. Equilibrium dissociation constants were measured using microscale thermophoresis. Since studies have shown that glycated albumin is excreted in the urine at a higher rate than unmodified albumin, we studied glucose and methylgloxal modified albumins (21 days). All had reduced affinity to FcRn at pH 6.0, suggesting these albumins would not be returned to the circulation via the transcytotic pathway. To address why modified albumin has reduced affinity, we analyzed the structure of the modified albumins using small-angle X-ray scattering. This analysis showed significant structural changes occurring to albumin with glycation, particularly in the FcRn-binding region, which could explain the reduced affinity to FcRn. These results offer an explanation for enhanced proximal tubule-mediated sorting and clearance of abnormal albumins.
Collapse
Affiliation(s)
- Mark C Wagner
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Jered Myslinski
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Shiv Pratap
- The Council of Scientific and Industrial Research Institute of Microbial Technology, Chandigarh, India
| | - Brittany Flores
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - George Rhodes
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Silvia B Campos-Bilderback
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Ruben M Sandoval
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Sudhanshu Kumar
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Monika Patel
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Ashish
- The Council of Scientific and Industrial Research Institute of Microbial Technology, Chandigarh, India
| | - Bruce A Molitoris
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana; Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana; and
| |
Collapse
|
42
|
Martins JP, Kennedy PJ, Santos HA, Barrias C, Sarmento B. A comprehensive review of the neonatal Fc receptor and its application in drug delivery. Pharmacol Ther 2016; 161:22-39. [PMID: 27016466 DOI: 10.1016/j.pharmthera.2016.03.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Advances in the understanding of neonatal Fc receptor (FcRn) biology and function have demonstrated that this receptor, primarily identified for the transfer of passive immunity from mother infant, is involved in several biological and immunological processes. In fact, FcRn is responsible for the long half-life of IgG and albumin in the serum, by creating an intracellular protein reservoir, which is protected from lysosomal degradation and, importantly, trafficked across the cell. Such discovery has led researchers to hypothesize the role for this unique receptor in the controlled delivery of therapeutic agents. A great amount of FcRn-based strategies are already under extensive investigation, in which FcRn reveals to have profound impact on the biodistribution and half-life extension of therapeutic agents. This review summarizes the main findings on FcRn biology, function and distribution throughout different tissues, together with the main advances on the FcRn-based therapeutic opportunities and model systems, which indicate that this receptor is a potential target for therapeutic regimen modification.
Collapse
Affiliation(s)
- João Pedro Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo 228, 4150-180 Porto, Portugal
| | - Patrick J Kennedy
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo 228, 4150-180 Porto, Portugal; Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI -00014 Helsinki, Finland
| | - Cristina Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde and Instituto Universitário de Ciências da Saúde, 4585-116 Gandra, Portugal.
| |
Collapse
|
43
|
Ziemssen F, Sobolewska B, Deissler H, Deissler H. Safety of monoclonal antibodies and related therapeutic proteins for the treatment of neovascular macular degeneration: addressing outstanding issues. Expert Opin Drug Saf 2015; 15:75-87. [PMID: 26568279 DOI: 10.1517/14740338.2016.1121232] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The vascular endothelial growth factor (VEGF) inhibitors most widely used to treat neovascular age-dependent macular degeneration (nAMD) are different proteins with structural features potentially relevant to adverse effects (AEs). Two of these are also established in cancer therapy (with higher dosages and AEs). The importance of ocular AE and extraocular activities is still a subject of controversy and ongoing research. AREAS COVERED Potential risks of intraocular VEGF inhibition based on prospective studies, in vitro investigations, pharmacokinetics, and hints from anti-cancer treatment. EXPERT OPINION nAMD is a frequently observed chronic clinical condition severely affecting the visual function of elderly persons. Intravitreal injection of VEGF-inactivating proteins is highly effective to prevent loss of vision. Anti-VEGF therapy is well tolerated, and low rates of ocular and systemic AEs in smaller trials suggest a very high benefit/risk ratio. The proteins established in nAMD therapy show similar efficacies. In the controversy over the off-label use of bevacizumab purely on grounds of much lower cost, the small, but potentially relevant differences between the available drugs are easily either dramatized (by pharmaceutical companies) or trivialized (by health insurances) and even political interference is involved. Facing the lack of a convincing body of evidence regarding safety, further long-term study results seem necessary.
Collapse
Affiliation(s)
- Focke Ziemssen
- a Center for Ophthalmology , Eberhard Karl University , D-72076 Tuebingen , Germany
| | - Bianka Sobolewska
- a Center for Ophthalmology , Eberhard Karl University , D-72076 Tuebingen , Germany
| | - Heidrun Deissler
- b Department of Ophthalmology , Univeristy of Ulm , D-89075 Ulm , Germany
| | | |
Collapse
|
44
|
Deissler HL, Lang GK, Lang GE. Internalization of bevacizumab by retinal endothelial cells and its intracellular fate: Evidence for an involvement of the neonatal Fc receptor. Exp Eye Res 2015; 143:49-59. [PMID: 26481553 DOI: 10.1016/j.exer.2015.10.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/25/2015] [Accepted: 10/05/2015] [Indexed: 12/27/2022]
Abstract
Bevacizumab is one of the VEGF-binding proteins that are established in clinical practice to treat various ocular diseases. In view of therapeutic long-term application, potential accumulation of the antibody in retinal cells gave reason for safety concerns. Internalization of considerable amounts of bevacizumab by retinal endothelial (REC) and pigment epithelial cells has been observed which may affect their important functions. Therefore we investigated the transport and intracellular localization of bevacizumab in immortalized bovine REC (iBREC) in detail, considering possible roles of vesicles and receptors mediating uptake and intracellular transport. By performing transcytosis assays with iBREC monolayers cultivated on porous membrane inserts, we demonstrated that bevacizumab was transported efficiently through a tight monolayer from the lower to the upper chamber or vice versa. When added to the lower chamber in excess, the internalized antibody was transported through the cells, but it was also recycled to be set free at the same side of the cell into a bevacizumab-free environment. The rates of both processes strongly depended on the concentration of fetal bovine serum (FBS) in the environment. This observation is important because in vivo REC might be exposed to varying amounts of serum, e.g. in patients with macular edema. FBS also affected the intracellular localization of bevacizumab as shown by analyses of subcellular fractions and direct immunofluorescence staining. When iBREC were cultivated in low-serum medium, most of the antibody was found in the fraction of cytoskeleton proteins and spots of high intensity of bevacizumab-specific staining close to the nuclei were observed. Cultivation in medium with FBS resulted in internalized bevacizumab predominately found in the membrane/organelle fraction in addition to its weaker association with proteins from the cytoskeleton and uniform staining of the cell. Bevacizumab-specific staining close to the cytoskeleton proteins α-tubulin or vimentin was also observed. Accumulation and association of the antibody with the cytoskeleton induced by serum reduction could be reversed by subsequent FBS addition. In uptake and transport of bevacizumab vesicles and binding to a receptor seems to be involved: Internalization was strongly temperature-dependent which ruled out paracellular passage and a fraction of the internalized bevacizumab was associated with early endosomes. Protein A inhibited transcytosis and affected intracellular localization suggesting a key role of the neonatal Fc receptor (FcRn). Interestingly, FcRn expression was decreased when iBREC were cultivated without FBS. Our results suggest this pathway of bevacizumab uptake and transition through iBREC: Independent of serum, bevacizumab is taken up through a nonspecific mechanism. The subsequent sorting into transport vesicles depends on the presence of serum as regulator of FcRn expression. Without sufficient amounts of the receptor being expressed, a likely obstructed exocytosis results in intracellular accumulation and an increased association with cytoskeleton proteins. Interaction of substantial amounts of bevacizumab with the cytoskeleton may be the reason for under these conditions suppressed migration of iBREC. If long-term therapies by intravitreal injection lead to accumulation of bevacizumab in REC in vivo and potentially harmful consequences, will have to be revealed by future investigations.
Collapse
Affiliation(s)
| | - Gerhard K Lang
- Department of Ophthalmology, University of Ulm, 89075 Ulm, Germany
| | - Gabriele E Lang
- Department of Ophthalmology, University of Ulm, 89075 Ulm, Germany
| |
Collapse
|
45
|
Meyer S, Nederend M, Jansen JHM, Reiding KR, Jacobino SR, Meeldijk J, Bovenschen N, Wuhrer M, Valerius T, Ubink R, Boross P, Rouwendal G, Leusen JHW. Improved in vivo anti-tumor effects of IgA-Her2 antibodies through half-life extension and serum exposure enhancement by FcRn targeting. MAbs 2015; 8:87-98. [PMID: 26466856 PMCID: PMC4966554 DOI: 10.1080/19420862.2015.1106658] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Antibody therapy is a validated treatment approach for several malignancies. All currently clinically applied therapeutic antibodies (Abs) are of the IgG isotype. However, not all patients respond to this therapy and relapses can occur. IgA represents an alternative isotype for antibody therapy that engages FcαRI expressing myeloid effector cells, such as neutrophils and monocytes. IgA Abs have been shown to effectively kill tumor cells both in vitro and in vivo. However, due to the short half-life of IgA Abs in mice, daily injections are required to reach an effect comparable to IgG Abs. The relatively long half-life of IgG Abs and serum albumin arises from their capability of interacting with the neonatal Fc receptor (FcRn). As IgA Abs lack a binding site for FcRn, we generated IgA Abs with the variable regions of the Her2-specific Ab trastuzumab and attached an albumin-binding domain (ABD) to the heavy or light chain (HCABD/LCABD) to extend their serum half-life. These modified Abs were able to bind albumin from different species in vitro. Furthermore, tumor cell lysis of IgA-Her2-LCABD Abs in vitro was similar to unmodified IgA-Her2 Abs. Pharmacokinetic studies in mice revealed that the serum exposure and half-life of the modified IgA-Her2 Abs was extended. In a xenograft mouse model, the modified IgA1 Abs exhibited a slightly, but significantly, improved anti-tumor response compared to the unmodified Ab. In conclusion, empowering IgA Abs with albumin-binding capacity results in in vitro and in vivo functional Abs with an enhanced exposure and prolonged half-life.
Collapse
Affiliation(s)
- Saskia Meyer
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - Maaike Nederend
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - J H Marco Jansen
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - Karli R Reiding
- b Center for Proteomics and Metabolomics; Leiden University Medical Center ; Leiden , The Netherlands
| | - Shamir R Jacobino
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - Jan Meeldijk
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - Niels Bovenschen
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands.,c Department of Pathology ; UMC Utrecht ; Utrecht , The Netherlands
| | - Manfred Wuhrer
- b Center for Proteomics and Metabolomics; Leiden University Medical Center ; Leiden , The Netherlands
| | - Thomas Valerius
- d Division of Stem Cell Transplantation and Immunotherapy ; Department of Internal Medicine II; Christian-Albrechts-University ; Kiel , Germany
| | - Ruud Ubink
- e Synthon Biopharmaceuticals BV ; Nijmegen , The Netherlands
| | - Peter Boross
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | | | - Jeanette H W Leusen
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| |
Collapse
|
46
|
Redman JM, Hill EM, AlDeghaither D, Weiner LM. Mechanisms of action of therapeutic antibodies for cancer. Mol Immunol 2015; 67:28-45. [PMID: 25911943 PMCID: PMC4529810 DOI: 10.1016/j.molimm.2015.04.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/29/2015] [Accepted: 04/03/2015] [Indexed: 02/06/2023]
Abstract
The therapeutic utility of antibodies and their derivatives is achieved by various means. The FDA has approved several targeted antibodies that disrupt signaling of various growth factor receptors for the treatment of a number of cancers. Rituximab, and other anti-CD20 monoclonal antibodies are active in B cell malignancies. As more experience has been gained with anti-CD20 monoclonal antibodies, the multifactorial nature of their anti-tumor mechanisms has emerged. Other targeted antibodies function to dampen inhibitory checkpoints. These checkpoint inhibitors have recently achieved dramatic results in several cancers, including melanoma. These and related antibodies continue to be investigated in the clinical and pre-clinical settings. Novel antibody structures that target two or more antigens have also made their way into clinical use. Tumor targeted antibodies can also be conjugated to chemo- or radiotherapeutic agents, or catalytic toxins, as a means to deliver toxic payloads to cancer cells. Here we provide a review of these mechanisms and a discussion of their relevance to current and future clinical applications.
Collapse
Affiliation(s)
- J M Redman
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - E M Hill
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - D AlDeghaither
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - L M Weiner
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States.
| |
Collapse
|
47
|
|
48
|
Abstract
The evolution of care in hemophilia is a remarkable story. Over the last 60 years, advances in protein purification, protein chemistry, donor screening, viral inactivation, gene sequencing, gene cloning, and recombinant protein production have dramatically enhanced the treatment and lives of patients with hemophilia. Recent efforts have produced enhanced half-life (EHL) clotting factors to better support prophylaxis and decrease the frequency of infusions. Medical needs remain in the areas of alternate modes of administration to decrease the need for venous access, better treatment, and prophylaxis for patients who form antibodies to clotting factors, and ultimately a cure of the underlying genetic defect. In this brief review, the authors summarize data on EHL clotting factors, introduce agents whose mode of action is not clotting factor replacement, and list current gene therapy efforts.
Collapse
Affiliation(s)
- Marcus E Carr
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA ; Worldwide Research and Development, Pfizer, Inc., Cambridge, MA, USA
| | - Bartholomew J Tortella
- Drexel University College of Medicine, Philadelphia, PA, USA ; Global Innovative Pharma, Pfizer, Inc., Collegeville, PA, USA
| |
Collapse
|
49
|
Chen Y, Chernyavsky A, Webber RJ, Grando SA, Wang PH. Critical Role of the Neonatal Fc Receptor (FcRn) in the Pathogenic Action of Antimitochondrial Autoantibodies Synergizing with Anti-desmoglein Autoantibodies in Pemphigus Vulgaris. J Biol Chem 2015; 290:23826-37. [PMID: 26260795 DOI: 10.1074/jbc.m115.668061] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 01/23/2023] Open
Abstract
Pemphigus vulgaris (PV) is a life-long, potentially fatal IgG autoantibody-mediated blistering disease targeting mucocutaneous keratinocytes (KCs). PV patients develop pathogenic anti-desmoglein (Dsg) 3 ± 1 and antimitochondrial antibodies (AMA), but it remained unknown whether and how AMA enter KCs and why other cell types are not affected in PV. Therefore, we sought to elucidate mechanisms of cell entry, trafficking, and pathogenic action of AMA in PV. We found that PVIgGs associated with neonatal Fc receptor (FcRn) on the cell membrane, and the PVIgG-FcRn complexes entered KCs and reached mitochondria where they dissociated. The liberated AMA altered mitochondrial membrane potential, respiration, and ATP production and induced cytochrome c release, although the lack or inactivation of FcRn abolished the ability of PVIgG to reach and damage mitochondria and to cause detachment of KCs. The assays of mitochondrial functions and keratinocyte adhesion demonstrated that although the pathobiological effects of AMA on KCs are reversible, they become irreversible, leading to epidermal blistering (acantholysis), when AMA synergize with anti-Dsg antibodies. Thus, it appears that AMA enter a keratinocyte in a complex with FcRn, become liberated from the endosome in the cytosol, and are trafficked to the mitochondria, wherein they trigger pro-apoptotic events leading to shrinkage of basal KCs uniquely expressing FcRn in epidermis. During recovery, KCs extend their cytoplasmic aprons toward neighboring cells, but anti-Dsg antibodies prevent assembly of nascent desmosomes due to steric hindrance, thus rendering acantholysis irreversible. In conclusion, FcRn is a common acceptor protein for internalization of AMA and, perhaps, for PV autoantibodies to other intracellular antigens, and PV is a novel disease paradigm for investigating and elucidating the role of FcRn in this autoimmune disease and possibly other autoimmune diseases.
Collapse
Affiliation(s)
- Yumay Chen
- From the Irvine Diabetes Center, Department of Medicine, and
| | | | | | - Sergei A Grando
- Departments of Dermatology and Biological Chemistry, and the Institute for Immunology, University of California at Irvine, Irvine, California 92967 and
| | - Ping H Wang
- From the Irvine Diabetes Center, Department of Medicine, and Biological Chemistry, and
| |
Collapse
|
50
|
Ward ES, Ober RJ. Commentary: "There's been a Flaw in Our Thinking". Front Immunol 2015; 6:351. [PMID: 26236309 PMCID: PMC4503918 DOI: 10.3389/fimmu.2015.00351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/28/2015] [Indexed: 12/16/2022] Open
Affiliation(s)
- E Sally Ward
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center , College Station, TX , USA ; Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center , Bryan, TX , USA ; Department of Immunology, University of Texas Southwestern Medical Center , Dallas, TX , USA
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center , College Station, TX , USA ; Department of Biomedical Engineering, Texas A&M University , College Station, TX , USA
| |
Collapse
|