1
|
Zhang L, Ma Y, Hettinga K, Zhou P. Suckling Rat Pup Model: Do Caprine Milk Lactoferrin and Immunoglobulins Have Different Digestion and Absorption Properties from That of Human and Bovine Species? JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:3069-3079. [PMID: 39873219 DOI: 10.1021/acs.jafc.4c10539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
This study aimed to investigate the digestion and absorption properties of caprine milk serum proteins in comparison to human and bovine species by using rat pups to mimic preterm infants. The results indicate that caprine lactoferrin (LTF) had a shorter retention time in the intestine and released a greater number of fragments, resembling human milk LTF more closely. In contrast, caprine immunoglobulins (Igs) were similar to bovine Igs and both exhibited a longer retention time in the intestine. For absorption, caprine Igs could be absorbed intact, which was similar to human and bovine Igs, whereas caprine LTF fragments were found in jejunum but not in plasma of rat pups. This is similar to bovine LTF but differed from human LTF as human LTF could be absorbed intact in plasma of rat pups at 20 min. In addition, the absorption rate of peptides and amino acids from caprine milk serum was similar to that of human milk serum, which was higher than that from bovine milk serum. This study aimed to enhance our understanding of the differences in bioavailability of LTF and Igs derived from caprine, human milk, and bovine milk, thereby offering guidance for selecting protein sources for premature infants.
Collapse
Affiliation(s)
- Lina Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Dairy Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ying Ma
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Kasper Hettinga
- International Joint Research Laboratory for Dairy Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Food Quality & Design Group, Wageningen University, Wageningen 6708WG, The Netherlands
| | - Peng Zhou
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Dairy Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
2
|
Michalon A, Renaud L, Machacek M, Cortijo C, Udata C, Mercuri MF, Buller F, Hock C, Nitsch RM, Kahr PC, Grimm J. Prediction of Cardiac ATTR Depletion by NI006 (ALXN2220) Using Mechanistic PK/PD Modeling. Clin Pharmacol Ther 2025; 117:261-269. [PMID: 39410666 PMCID: PMC11652821 DOI: 10.1002/cpt.3455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/14/2024] [Indexed: 12/19/2024]
Abstract
NI006 (aka ALXN2220) is a therapeutic antibody candidate in phase III clinical development for the depletion of amyloid transthyretin (ATTR) in patients with ATTR cardiomyopathy, an infiltrative cardiomyopathy leading to increased left ventricular wall thickness (LVWT). The mode-of-action consists in removal of disease-causing amyloid accumulations by activating phagocytic immune cells, a mechanism without precedent in cardiology. To select a safe and potentially efficacious dose range and treatment duration for a combined first-in-human and proof-of-concept clinical phase Ib study, we developed a mechanistic pharmacokinetic and pharmacodynamic (PK/PD) model that can predict NI006 exposure, its effects on cardiac amyloid load and on LWVT, which is a predictor of heart failure in this disease. The PK/PD model predictions supported 0.3 mg/kg monthly dosing as a safe starting dose and identified 10-60 mg/kg monthly as the potentially efficacious dose range with substantial and dose dependent cardiac amyloid burden reduction within 4 months for 60 mg/kg and 10 months for 10 mg/kg. These predictions were in good agreement with the observed primary results of the clinical phase Ib study where amyloid burden was measured by imaging. This novel translational PK/PD model provided important predictions to guide the design of the phase Ib study of NI006, indicating the value of this approach to integrate preclinical results into clinical trial design and increase translational success.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christoph Hock
- NeurimmuneSchlierenSwitzerland
- Institute for Regenerative MedicineUniversity of ZurichSchlierenSwitzerland
| | - Roger M. Nitsch
- NeurimmuneSchlierenSwitzerland
- Institute for Regenerative MedicineUniversity of ZurichSchlierenSwitzerland
| | - Peter C. Kahr
- NeurimmuneSchlierenSwitzerland
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
| | | |
Collapse
|
3
|
Robarge JD, Budge KM, Her L, Patterson AM, Brown-Augsburger P. Rat as a Predictive Model for Human Clearance and Bioavailability of Monoclonal Antibodies. Antibodies (Basel) 2024; 14:2. [PMID: 39846610 PMCID: PMC11755617 DOI: 10.3390/antib14010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND The prediction of human clearance (CL) and subcutaneous (SC) bioavailability is a critical aspect of monoclonal antibody (mAb) selection for clinical development. While monkeys are a well-accepted model for predicting human CL, other preclinical species have been less-thoroughly explored. Unlike CL, predicting the bioavailability of SC administered mAbs in humans remains challenging as contributing factors are not well understood, and preclinical models have not been systematically evaluated. METHODS Non-clinical and clinical pharmacokinetic (PK) parameters were mined from public and internal sources for rats, cynomolgus monkeys, and humans. Intravenous (IV) and SC PK was determined in Sprague Dawley rats for fourteen mAbs without existing PK data. Together, we obtained cross-species data for 25 mAbs to evaluate CL and SC bioavailability relationships among rats, monkeys, and humans. RESULTS Rat and monkey CL significantly correlated with human CL and supported the use of species-specific exponents for body-weight-based allometric scaling. Notably, rat SC bioavailability significantly correlated with human SC bioavailability, while monkey SC bioavailability did not. Bioavailability also correlated with clearance. CONCLUSIONS The rat model enables an early assessment of mAb PK properties, allowing discrimination among molecules in the discovery pipeline and prediction of human PK. Importantly, rat SC bioavailability significantly correlated with human SC bioavailability, which has not been observed with other species. Rats are cost-effective and efficient relative to monkeys and provide a valuable tool for pharmacokinetic predictions in therapeutic antibody discovery.
Collapse
Affiliation(s)
| | | | | | | | - Patricia Brown-Augsburger
- Eli Lilly and Company, Lilly Corporate Center Indianapolis, Indianapolis, IN 46285, USA; (J.D.R.); (K.M.B.); (L.H.); (A.M.P.)
| |
Collapse
|
4
|
Franz M, Jairam RK, Kuepfer L, Hanke N. PBPK-based translation from preclinical species to humans for the full-size IgG therapeutic efalizumab. Front Pharmacol 2024; 15:1418870. [PMID: 39411068 PMCID: PMC11473394 DOI: 10.3389/fphar.2024.1418870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Animal models play a vital role in pharmaceutical research and development by supporting the planning and design of later clinical studies. To improve confidence and reliability of first in human dose estimates it is essential to assess the comparability of animal studies with the human situation. In the context of large molecules, it is particularly important to evaluate the cross-species-translatability of parameters related to neonatal fragment crystallizable receptor (FcRn) binding and target mediated drug disposition (TMDD), as they greatly influence distribution and disposition of proteins in the body of an organism. Methods Plasma pharmacokinetic data of the therapeutic protein efalizumab were obtained from literature. Physiologically based pharmacokinetic (PBPK) models were built for three different species (rabbit, non-human primate (NHP), human). Target binding was included in the NHP and human models. The assumption of similar target turnover and target-binding in NHP and human was explored, to gain insights into how these parameters might be translated between species. Results Efalizumab PBPK models were successfully developed for three species and concentration-time-profiles could be described appropriately across different intravenously administered doses. The final NHP and human models feature a common set of parameters for target turnover and drug-target-complex internalization, as well as comparable target-binding parameters. Our analyses show that different parameter values for FcRn affinity are crucial to accurately describe the concentration-time profiles. Discussion Based on the available data in rabbits, NHP and humans, parameters for FcRn affinity cannot be translated between species, but parameters related to target mediated drug disposition can be translated from NHP to human. The inclusion of additional pharmacokinetic (PK) data including different efalizumab doses would further support and confirm our findings on identifying TMDD and, thus, binding kinetics of efalizumab in NHPs. Furthermore, we suggest that information on target expression and internalization rates could make it possible to develop comprehensive human PBPK models with minimal animal testing. In this project, we compared the pharmacokinetics of a therapeutic protein in rabbit, NHP and human using an open PBPK modeling platform (Open Systems Pharmacology Suite, http://www.open-systems-pharmacology.org). Our findings could support similar translatory studies for first in human dose predictions in the future.
Collapse
Affiliation(s)
- Maria Franz
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH and Co. KG, Ingelheim, Germany
| | - Ravi Kumar Jairam
- Institute for Systems Medicine with Focus on Organ Interaction, University Hospital RWTH Aachen, Aachen, Germany
| | - Lars Kuepfer
- Institute for Systems Medicine with Focus on Organ Interaction, University Hospital RWTH Aachen, Aachen, Germany
| | - Nina Hanke
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH and Co. KG, Ingelheim, Germany
| |
Collapse
|
5
|
Chen W, Wang L, Ruan Z, Lou H, Jiang B. Translate Pharmacokinetics of PD-1/PD-L1 Monoclonal Antibodies from Cynomolgus Monkey to Human: Comparison of Different Approaches. J Pharm Sci 2024; 113:2915-2921. [PMID: 38986869 DOI: 10.1016/j.xphs.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Antibodies blocking programmed death-1 (PD-1) and its natural ligand programmed death-ligand 1 (PD-L1) have been proved to be promising strategies in recent years. Hundreds of PD-1/PD-L1 antibodies are under development worldwide. Prediction of human pharmacokinetics (PK) in the preclinical stage is critical for designing dosing regimens in first-in-human studies. This study aims to predict the PK of PD-1/PD-L1 antibodies in human by scaling of monkey data. A systematic literature search of published articles on the PK of PD-1/PD-L1 antibodies in cynomolgus monkey and in human was conducted. Allometric scaling (AS), the species time-invariant (STIV) method, as well as physiologically based pharmacokinetic (PBPK) modeling were investigated. Six antibodies (avelumab, atezolizumab, nivolumab, pembrolizumab, cemiplimab, and zimberelimab) were included for investigation. The exponents used in this study were 0.85 and 1 for clearance (CL) and distribution volume (V), respectively, both for AS and STIV methods. The generic PBPK model for macromolecules in PK-Sim was used without further modifications. The dissociation constant of the antibody for binding to FcRn (KD) in endosome space for human was assumed to be two-fold of that for monkey. Predicted human CLs for the majority of drugs were within the observed range, while Vs were not well predicted using the AS method. The STIV method and the generic PBPK model can be employed to translate concentration-time curves of PD-1/PD-L1 antibodies from cynomolgus monkey to human with comparable efficacy. The results of this study provide reference for the early development of PD-1/PD-L1 antibodies.
Collapse
Affiliation(s)
- Wenjun Chen
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lu Wang
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zourong Ruan
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Honggang Lou
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bo Jiang
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Tajiri A, Matsumoto S, Maeda S, Soga T, Kagiyama K, Ikeda H, Fukasawa K, Miyata A, Kamimura H. Prediction of human serum concentration-time profiles of therapeutic monoclonal antibodies using common marmosets ( Callithrix jacchus): initial assessment with canakinumab, adalimumab, and bevacizumab. Xenobiotica 2024; 54:648-657. [PMID: 38977390 DOI: 10.1080/00498254.2024.2371921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024]
Abstract
Cynomolgus monkeys and human FcRn transgenic mice are generally used for pharmacokinetic predictions of therapeutic monoclonal antibodies (mAbs). In the present study, the application of the common marmoset, a small nonhuman primate, as a potential animal model for prediction was evaluated for the first time.Canakinumab, adalimumab, and bevacizumab, which exhibited linear pharmacokinetics in humans, were selected as the model compounds. Marmoset pharmacokinetic data were reportedly available only for canakinumab, and those for adalimumab and bevacizumab were acquired in-house.Four pharmacokinetic parameters for a two-compartment model (i.e. clearance and volume of distribution in the central and peripheral compartments) in marmosets were extrapolated to the values in humans with allometric scaling using the average exponents of the three mAbs. As a result, the observed human serum concentration-time curves of the three mAbs following intravenous administration and those of canakinumab and adalimumab following subcutaneous injections (with an assumed absorption rate constant and bioavailability) were reasonably predicted.Although further prediction studies using a sufficient number of other mAbs are necessary to evaluate the versatility of this model, the findings indicate that marmosets can be an alternative to preceding animals for human pharmacokinetic predictions of therapeutic mAbs.
Collapse
Affiliation(s)
- Ayaka Tajiri
- Drug Discovery Department, R&D Division, Meiji Seika Pharma Co., Ltd, Tokyo, Japan
| | - Shogo Matsumoto
- Drug Discovery Department, R&D Division, Meiji Seika Pharma Co., Ltd, Tokyo, Japan
| | - Satoshi Maeda
- Yaotsu Breeding Center, CLEA Japan, Inc., Gifu, Japan
| | - Takuma Soga
- Yaotsu Breeding Center, CLEA Japan, Inc., Gifu, Japan
| | | | - Hiroshi Ikeda
- Tokyo Animal and Diet Department, CLEA Japan, Inc., Tokyo, Japan
| | | | - Atsunori Miyata
- Drug Discovery Department, R&D Division, Meiji Seika Pharma Co., Ltd, Tokyo, Japan
| | | |
Collapse
|
7
|
Hentrich C, Putyrski M, Hanuschka H, Preis W, Kellmann SJ, Wich M, Cavada M, Hanselka S, Lelyveld VS, Ylera F. Engineered reversible inhibition of SpyCatcher reactivity enables rapid generation of bispecific antibodies. Nat Commun 2024; 15:5939. [PMID: 39009599 PMCID: PMC11251281 DOI: 10.1038/s41467-024-50296-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 07/05/2024] [Indexed: 07/17/2024] Open
Abstract
The precise regulation of protein function is essential in biological systems and a key goal in chemical biology and protein engineering. Here, we describe a straightforward method to engineer functional control into the isopeptide bond-forming SpyTag/SpyCatcher protein ligation system. First, we perform a cysteine scan of the structured region of SpyCatcher. Except for two known reactive and catalytic residues, none of these mutations abolish reactivity. In a second screening step, we modify the cysteines with disulfide bond-forming small molecules. Here we identify 8 positions at which modifications strongly inhibit reactivity. This inhibition can be reversed by reducing agents. We call such a reversibly inhibitable SpyCatcher "SpyLock". Using "BiLockCatcher", a genetic fusion of wild-type SpyCatcher and SpyLock, and SpyTagged antibody fragments, we generate bispecific antibodies in a single, scalable format, facilitating the screening of a large number of antibody combinations. We demonstrate this approach by screening anti-PD-1/anti-PD-L1 bispecific antibodies using a cellular reporter assay.
Collapse
Affiliation(s)
| | - Mateusz Putyrski
- Bio-Rad AbD Serotec GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Hanh Hanuschka
- Bio-Rad AbD Serotec GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Waldemar Preis
- Bio-Rad AbD Serotec GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | | | - Melissa Wich
- Bio-Rad AbD Serotec GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Manuel Cavada
- Bio-Rad AbD Serotec GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Sarah Hanselka
- Bio-Rad AbD Serotec GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Victor S Lelyveld
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Francisco Ylera
- Bio-Rad AbD Serotec GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany.
| |
Collapse
|
8
|
Miles JR, Lu P, Bai S, Aguillón-Durán GP, Rodríguez-Herrera JE, Gunn BM, Restrepo BI, Lu LL. Antigen specificity shapes antibody functions in tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597169. [PMID: 38895452 PMCID: PMC11185737 DOI: 10.1101/2024.06.03.597169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tuberculosis (TB) is the number one infectious disease cause of death worldwide due to an incomplete understanding of immunity. Emerging data highlight antibody functions mediated by the Fc domain as immune correlates. However, the mechanisms by which antibody functions impact the causative agent Mycobacterium tuberculosis (Mtb) are unclear. Here, we examine how antigen specificity determined by the Fab domain shapes Fc effector functions against Mtb. Using the critical structural and secreted virulence proteins Mtb cell wall and ESAT-6 & CFP-10, we observe that antigen specificity alters subclass, antibody post-translational glycosylation, and Fc effector functions in TB patients. Moreover, Mtb cell wall IgG3 enhances disease through opsonophagocytosis of extracellular Mtb . In contrast, polyclonal and a human monoclonal IgG1 we generated targeting ESAT-6 & CFP-10 inhibit intracellular Mtb . These data show that antibodies have multiple roles in TB and antigen specificity is a critical determinant of the protective and pathogenic capacity.
Collapse
|
9
|
Lopes JA, Garnier NE, Pei Y, Yates JGE, Campbell ESB, Goens MM, Hughes ME, Rghei AD, Stevens BAY, Guilleman MM, Thompson B, Khursigara CM, Susta L, Wootton SK. AAV-vectored expression of monospecific or bispecific monoclonal antibodies protects mice from lethal Pseudomonas aeruginosa pneumonia. Gene Ther 2024; 31:400-412. [PMID: 38678160 DOI: 10.1038/s41434-024-00453-1] [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] [Received: 11/01/2023] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024]
Abstract
Pseudomonas aeruginosa poses a significant threat to immunocompromised individuals and those with cystic fibrosis. Treatment relies on antibiotics, but persistent infections occur due to intrinsic and acquired resistance of P. aeruginosa towards multiple classes of antibiotics. To date, there are no licensed vaccines for this pathogen, prompting the urgent need for novel treatment approaches to combat P. aeruginosa infection and persistence. Here we validated AAV vectored immunoprophylaxis as a strategy to generate long-term plasma and mucosal expression of highly protective monoclonal antibodies (mAbs) targeting the exopolysaccharide Psl (Cam-003) and the PcrV (V2L2MD) component of the type-III secretion system injectosome either as single mAbs or together as a bispecific mAb (MEDI3902) in a mouse model. When administered intramuscularly, AAV-αPcrV, AAV-αPsl, and AAV-MEDI3902 significantly protected mice challenged intranasally with a lethal dose of P. aeruginosa strains PAO1 and PA14 and reduced bacterial burden and dissemination to other organs. While all AAV-mAbs provided protection, AAV-αPcrV and AAV-MEDI3902 provided 100% and 87.5% protection from a lethal challenge with 4.47 × 107 CFU PAO1 and 87.5% and 75% protection from a lethal challenge with 3 × 107 CFU PA14, respectively. Serum concentrations of MEDI3902 were ~10× lower than that of αPcrV, but mice treated with this vector showed a greater reduction in bacterial dissemination to the liver, lung, spleen, and blood compared to other AAV-mAbs. These results support further investigation into the use of AAV vectored immunoprophylaxis to prevent and treat P. aeruginosa infections and other bacterial pathogens of public health concern for which current treatment strategies are limited.
Collapse
Affiliation(s)
- Jordyn A Lopes
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Nicole E Garnier
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Yanlong Pei
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jacob G E Yates
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Elena S B Campbell
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Melanie M Goens
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Madison E Hughes
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Amira D Rghei
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Brenna A Y Stevens
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Matthew M Guilleman
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Brad Thompson
- Avamab Pharma Inc., 120, 4838 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Cezar M Khursigara
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Leonardo Susta
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Sarah K Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
10
|
Dai T, Sun H, Liban T, Vicente-Suarez I, Zhang B, Song Y, Jiang Z, Yu J, Sheng J, Lv B. A novel anti-LAG-3/TIGIT bispecific antibody exhibits potent anti-tumor efficacy in mouse models as monotherapy or in combination with PD-1 antibody. Sci Rep 2024; 14:10661. [PMID: 38724599 PMCID: PMC11082181 DOI: 10.1038/s41598-024-61477-6] [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: 02/01/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
We report the generation of a novel anti-LAG-3/TIGIT bispecific IgG4 antibody, ZGGS15, and evaluated its anti-tumor efficacy in mouse models as monotherapy or in combination with a PD-1 antibody. ZGGS15 exhibited strong affinities for human LAG-3 and TIGIT, with KDs of 3.05 nM and 2.65 nM, respectively. ZGGS15 has EC50s of 0.69 nM and 1.87 nM for binding to human LAG-3 and TIGIT on CHO-K1 cells, respectively. ZGGS15 competitively inhibited the binding of LAG-3 to MHC-II (IC50 = 0.77 nM) and the binding of TIGIT to CD155 (IC50 = 0.24 nM). ZGGS15 does not induce ADCC, CDC, or obvious cytokine production. In vivo results showed that ZGGS15 had better anti-tumor inhibition than single anti-LAG-3 or anti-TIGIT agents and demonstrated a synergistic effect when combined with nivolumab, with a significantly higher tumor growth inhibition of 95.80% (p = 0.001). The tumor volume inhibition rate for ZGGS15 at 2 mg/kg was 69.70%, and for ZGGS15 at 5 mg/kg plus nivolumab at 1 mg/kg, it was 94.03% (p < 0.001). Our data reveal that ZGGS15 exhibits potent anti-tumor efficacy without eliciting ADCC or CDC or causing cytokine production, therefore having a safe profile.
Collapse
Affiliation(s)
- Tongcheng Dai
- Suzhou Zelgen Biopharmaceuticals Co., Ltd, Kunshan, China
| | - Hao Sun
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tyler Liban
- Gensun Biopharma Inc., Thousand Oaks, CA, USA
| | | | - Bin Zhang
- Suzhou Zelgen Biopharmaceuticals Co., Ltd, Kunshan, China
| | - Yongping Song
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhongxing Jiang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jifeng Yu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | | | - Binhua Lv
- Suzhou Zelgen Biopharmaceuticals Co., Ltd, Kunshan, China.
| |
Collapse
|
11
|
Haddad G, Blaine J. Identification of Four Mouse FcRn Splice Variants and FcRn-Specific Vesicles. Cells 2024; 13:594. [PMID: 38607033 PMCID: PMC11012118 DOI: 10.3390/cells13070594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
Research into the neonatal Fc receptor (FcRn) has increased dramatically ever since Simister and Mostov first purified a rat version of the receptor. Over the years, FcRn has been shown to function not only as a receptor that transfers immunity from mother to fetus but also performs an array of different functions that include transport and recycling of immunoglobulins and albumin in the adult. Due to its important cellular roles, several clinical trials have been designed to either inhibit/enhance FcRn function or develop of non-invasive therapeutic delivery system such as fusion of drugs to IgG Fc or albumin to enhance delivery inside the cells. Here, we report the accidental identification of several FcRn alternatively spliced variants in both mouse and human cells. The four new mouse splice variants are capable of binding immunoglobulins' Fc and Fab portions. In addition, we have identified FcRn-specific vesicles in which immunoglobulins and albumin can be stored and that are involved in the endosomal-lysosomal system. The complexity of FcRn functions offers significant potential to design and develop novel and targeted therapeutics.
Collapse
Affiliation(s)
| | - Judith Blaine
- Division of Renal Disease and Hypertension, Department of Medicine, School of Medicine, University of Colorado, Aurora, CO 80045, USA;
| |
Collapse
|
12
|
Kwak H, Kim MS, Kim S, Kim J, Aoki Y, Chung SJ, Nam HJ, Lee W. Kinetic modeling of the plasma pharmacokinetic profiles of ADAMTS13 fragment and its Fc-fusion counterpart in mice. Front Pharmacol 2024; 15:1352842. [PMID: 38590637 PMCID: PMC10999626 DOI: 10.3389/fphar.2024.1352842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction: Fusion of the fragment crystallizable (Fc) to protein therapeutics is commonly used to extend the circulation time by enhancing neonatal Fc-receptor (FcRn)-mediated endosomal recycling and slowing renal clearance. This study applied kinetic modeling to gain insights into the cellular processing contributing to the observed pharmacokinetic (PK) differences between the novel recombinant ADAMTS13 fragment (MDTCS) and its Fc-fusion protein (MDTCS-Fc). Methods: For MDTCS and MDTCS-Fc, their plasma PK profiles were obtained at two dose levels following intravenous administration of the respective proteins to mice. The plasma PK profiles of MDTCS were fitted to a kinetic model with three unknown protein-dependent parameters representing the fraction recycled (FR) and the rate constants for endocytosis (kup, for the uptake into the endosomes) and for the transfer from the plasma to the interstitial fluid (kpi). For MDTCS-Fc, the model was modified to include an additional parameter for binding to FcRn. Parameter optimization was done using the Cluster Gauss-Newton Method (CGNM), an algorithm that identifies multiple sets of approximate solutions ("accepted" parameter sets) to nonlinear least-squares problems. Results: As expected, the kinetic modeling results yielded the FR of MDTCS-Fc to be 2.8-fold greater than that of MDTCS (0.8497 and 0.3061, respectively). In addition, MDTCS-Fc was predicted to undergo endocytosis (the uptake into the endosomes) at a slower rate than MDTCS. Sensitivity analyses identified the association rate constant (kon) between MDTCS-Fc and FcRn as a potentially important factor influencing the plasma half-life in vivo. Discussion: Our analyses suggested that Fc fusion to MDTCS leads to changes in not only the FR but also the uptake into the endosomes, impacting the systemic plasma PK profiles. These findings may be used to develop recombinant protein therapeutics with extended circulation time.
Collapse
Affiliation(s)
- Heechun Kwak
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
- Discovery Unit, Research and Early Development Department, GC Biopharma Corp, Yongin-si, Republic of Korea
| | - Min-Soo Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Suyong Kim
- Discovery Unit, Research and Early Development Department, GC Biopharma Corp, Yongin-si, Republic of Korea
| | - Jiyoung Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yasunori Aoki
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, Josai International University, Tokyo, Japan
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Suk-Jae Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyun-Ja Nam
- Discovery Unit, Research and Early Development Department, GC Biopharma Corp, Yongin-si, Republic of Korea
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
13
|
Park SA, Lee Y, Hwang H, Lee JH, Kang YJ, Kim Y, Jin C, An HJ, Oh YJ, Hinterdorfer P, Kim E, Choi S, Ko K. Fc engineered anti-virus therapeutic human IgG 1 expressed in plants with altered binding to the neonatal Fc receptor. Biotechnol J 2024; 19:e2300552. [PMID: 38528347 DOI: 10.1002/biot.202300552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 03/27/2024]
Abstract
Production of therapeutic monoclonal antibody (mAb) in transgenic plants has several advantages such as large-scale production and the absence of pathogenic animal contaminants. However, mAb with high mannose (HM) type glycans has shown a faster clearance compared to antibodies produced in animal cells. The neonatal Fc receptor (FcRn) regulates the persistence of immunoglobulin G (IgG) by the FcRn-mediated recycling pathway, which salvages IgG from lysosomal degradation within cells. In this study, Fc-engineering of antirabies virus therapeutic mAb SO57 with the endoplasmic reticulum (ER)-retention peptide signal (Lys-Asp-Glu-Leu; KDEL) (mAbpK SO57) in plant cell was conducted to enhance its binding activity to human neonatal Fc receptor (hFcRn), consequently improve its serum half-life. Enzyme-linked immunosorbent assay (ELISA) and Surface plasmon resonance assay showed altered binding affinity of the Fc region of three different mAbpK SO57 variants [M252Y/S254T/T256E (MST), M428L/N434S (MN), H433K/N434F (HN)] to hFcRn compared to wild type (WT) of mAbpK SO57. Molecular modeling data visualized the structural alterations in these mAbpK SO57. All of the mAbpK SO57 variants had HM type glycan structures similar to the WT mAbpK SO57. In addition, the neutralizing activity of the three variants against the rabies virus CVS-11 was effective as the WT mAbpK SO57. These results indicate that the binding affinity of mAbpK SO57 variants to hFcRn can be modified without alteration of N-glycan structure and neutralization activity. Taken together, this study suggests that Fc-engineering of antirabies virus mAb can be applied to enhance the efficacy of therapeutic mAbs in plant expression systems.
Collapse
Affiliation(s)
- Sol-Ah Park
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Hyunjoo Hwang
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Jeong Hwan Lee
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Yang Joo Kang
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Yerin Kim
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Caiquan Jin
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Hyun Joo An
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| | - Yoo Jin Oh
- Department of Applied Experimental Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Peter Hinterdorfer
- Department of Applied Experimental Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Eunhye Kim
- Global AI Drug Discovery Center, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Sun Choi
- Global AI Drug Discovery Center, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Kisung Ko
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| |
Collapse
|
14
|
Reusch J, Andersen JT, Rant U, Schlothauer T. Insight into the avidity-affinity relationship of the bivalent, pH-dependent interaction between IgG and FcRn. MAbs 2024; 16:2361585. [PMID: 38849969 PMCID: PMC11164218 DOI: 10.1080/19420862.2024.2361585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
Monoclonal antibodies (mAbs) as therapeutics necessitate favorable pharmacokinetic properties, including extended serum half-life, achieved through pH-dependent binding to the neonatal Fc receptor (FcRn). While prior research has mainly investigated IgG-FcRn binding kinetics with a focus on single affinity values, it has been shown that each IgG molecule can engage two FcRn molecules throughout an endosomal pH gradient. As such, we present here a more comprehensive analysis of these interactions with an emphasis on both affinity and avidity by taking advantage of switchSENSE technology, a surface-based biosensor where recombinant FcRn was immobilized via short DNA nanolevers, mimicking the membranous orientation of the receptor. The results revealed insight into the avidity-to-affinity relationship, where assessing binding through a pH gradient ranging from pH 5.8 to 7.4 showed that the half-life extended IgG1-YTE has an affinity inflection point at pH 7.2, reflecting its engineering for improved FcRn binding compared with the wild-type counterpart. Furthermore, IgG1-YTE displayed a pH switch for the avidity enhancement factor at pH 6.2, reflecting strong receptor binding to both sides of the YTE-containing Fc, while avidity was abolished at pH 7.4. When compared with classical surface plasmon resonance (SPR) technology and complementary methods, the use of switchSENSE demonstrated superior capabilities in differentiating affinity from avidity within a single measurement. Thus, the methodology provides reliable kinetic rate parameters for both binding modes and their direct relationship as a function of pH. Also, it deciphers the potential effect of the variable Fab arms on FcRn binding, in which SPR has limitations. Our study offers guidance for how FcRn binding properties can be studied for IgG engineering strategies.
Collapse
Affiliation(s)
- Johannes Reusch
- Dynamic Biosensors GmbH, Munich, Germany
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | | | - Tilman Schlothauer
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
15
|
Mahant AM, Trejo FE, Aguilan JT, Sidoli S, Permar SR, Herold BC. Antibody attributes, Fc receptor expression, gestation and maternal SARS-CoV-2 infection modulate HSV IgG placental transfer. iScience 2023; 26:107648. [PMID: 37670782 PMCID: PMC10475509 DOI: 10.1016/j.isci.2023.107648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/30/2023] [Accepted: 08/11/2023] [Indexed: 09/07/2023] Open
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is associated with protection against neonatal herpes. We hypothesized that placental transfer of ADCC-mediating herpes simplex virus (HSV) immunoglobulin G (IgG) is influenced by antigenic target, function, glycans, gestational age, and maternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Maternal and cord blood were collected from HSV-seropositive (HSV+) mothers pre-COVID and HSV+/SARS-CoV-2+ mothers during the pandemic. Transfer of HSV neutralizing IgG was significantly lower in preterm versus term dyads (transfer ratio [TR] 0.84 vs. 2.44) whereas the TR of ADCC-mediating IgG was <1.0 in both term and preterm pre-COVID dyads. Anti-glycoprotein D IgG, which had only neutralizing activity, and anti-glycoprotein B (gB) IgG, which displayed neutralizing and ADCC activity, exhibited different relative affinities for the neonatal Fc receptor (FcRn) and expressed different glycans. The transfer of ADCC-mediating IgG increased significantly in term SARS-CoV-2+ dyads. This was associated with greater placental colocalization of FcRn with FcγRIIIa. These findings have implications for strategies to prevent neonatal herpes.
Collapse
Affiliation(s)
- Aakash Mahant Mahant
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Fatima Estrada Trejo
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jennifer T. Aguilan
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Simone Sidoli
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sallie R. Permar
- Department of Pediatrics, Weil Cornell Medicine, New York, NY 10021, USA
| | - Betsy C. Herold
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
16
|
Dekkers C, El Amrani M, van Luin M, Bakker D, Plusjé L, van Wijk F, de Bruin-Weller M. Limited excretion of dupilumab into breastmilk: A case report. J Eur Acad Dermatol Venereol 2023; 37:e1154-e1155. [PMID: 37143362 DOI: 10.1111/jdv.19164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/17/2023] [Indexed: 05/06/2023]
Affiliation(s)
- Coco Dekkers
- Department of Dermatology and Allergology, National Expertise Center for Atopic Dermatitis, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mohsin El Amrani
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Matthijs van Luin
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Daphne Bakker
- Department of Dermatology and Allergology, National Expertise Center for Atopic Dermatitis, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leon Plusjé
- Department of Dermatology, Red Cross Hospital Beverwijk, Beverwijk, The Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marjolein de Bruin-Weller
- Department of Dermatology and Allergology, National Expertise Center for Atopic Dermatitis, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
17
|
Pasquiers B, Benamara S, Felices M, Ternant D, Declèves X, Puszkiel A. Translation of Monoclonal Antibodies Pharmacokinetics from Animal to Human Using Physiologically Based Modeling in Open Systems Pharmacology (OSP) Suite: A Retrospective Analysis of Bevacizumab. Pharmaceutics 2023; 15:2129. [PMID: 37631343 PMCID: PMC10459442 DOI: 10.3390/pharmaceutics15082129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Interspecies translation of monoclonal antibodies (mAbs) pharmacokinetics (PK) in presence of target-mediated drug disposition (TMDD) is particularly challenging. Incorporation of TMDD in physiologically based PK (PBPK) modeling is recent and needs to be consolidated and generalized to provide better prediction of TMDD regarding inter-species translation during preclinical and clinical development steps of mAbs. The objective of this study was to develop a generic PBPK translational approach for mAbs using the open-source software (PK-Sim® and Mobi®). The translation of bevacizumab based on data in non-human primates (NHP), healthy volunteers (HV), and cancer patients was used as a case example for model demonstration purpose. A PBPK model for bevacizumab concentration-time data was developed using data from literature and the Open Systems Pharmacology (OSP) Suite version 10. PK-sim® was used to build the linear part of bevacizumab PK (mainly FcRn-mediated), whereas MoBi® was used to develop the target-mediated part. The model was first developed for NHP and used for a priori PK prediction in HV. Then, the refined model obtained in HV was used for a priori prediction in cancer patients. A priori predictions were within 2-fold prediction error (predicted/observed) for both area under the concentration-time curve (AUC) and maximum concentration (Cmax) and all the predicted concentrations were within 2-fold average fold error (AFE) and average absolute fold error (AAFE). Sensitivity analysis showed that FcRn-mediated distribution and elimination processes must be accounted for at all mAb concentration levels, whereas the lower the mAb concentration, the more significant the target-mediated elimination. This project is the first step to generalize the full PBPK translational approach in Model-Informed Drug Development (MIDD) of mAbs using OSP Suite.
Collapse
Affiliation(s)
- Blaise Pasquiers
- Inserm UMR-S1144, Faculty of Pharmacy, Université Paris Cité, 75006 Paris, France (A.P.)
- PhinC Development, 91300 Massy, France
| | | | | | - David Ternant
- Faculty of Medicine, Université de Tours, EA 4245 T2I, 37032 Tours, France
- Service de Pharmacologie Médicale, CHRU de Tours, 37000 Tours, France
| | - Xavier Declèves
- Inserm UMR-S1144, Faculty of Pharmacy, Université Paris Cité, 75006 Paris, France (A.P.)
- Biologie du Médicament—Toxicologie, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, 75014 Paris, France
| | - Alicja Puszkiel
- Inserm UMR-S1144, Faculty of Pharmacy, Université Paris Cité, 75006 Paris, France (A.P.)
- Biologie du Médicament—Toxicologie, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, 75014 Paris, France
| |
Collapse
|
18
|
Abstract
IgGs are essential soluble components of the adaptive immune response that evolved to protect the body from infection. Compared with other immunoglobulins, the role of IgGs is distinguished and enhanced by their high circulating levels, long half-life and ability to transfer from mother to offspring, properties that are conferred by interactions with neonatal Fc receptor (FcRn). FcRn binds to the Fc portion of IgGs in a pH-dependent manner and protects them from intracellular degradation. It also allows their transport across polarized cells that separate tissue compartments, such as the endothelium and epithelium. Further, it is becoming apparent that FcRn functions to potentiate cellular immune responses when IgGs, bound to their antigens, form IgG immune complexes. Besides the protective role of IgG, IgG autoantibodies are associated with numerous pathological conditions. As such, FcRn blockade is a novel and effective strategy to reduce circulating levels of pathogenic IgG autoantibodies and curtail IgG-mediated diseases, with several FcRn-blocking strategies on the path to therapeutic use. Here, we describe the current state of knowledge of FcRn-IgG immunobiology, with an emphasis on the functional and pathological aspects, and an overview of FcRn-targeted therapy development.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lisa K Kozicky
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amit K Gandhi
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Digestive Diseases Center, Boston, MA, USA.
| |
Collapse
|
19
|
Iwasaki YW, Tharakaraman K, Subramanian V, Khongmanee A, Hatas A, Fleischer E, Rurak TT, Ngok-ngam P, Tit-oon P, Ruchirawat M, Satayavivad J, Fuangthong M, Sasisekharan R. Generation of bispecific antibodies by structure-guided redesign of IgG constant regions. Front Immunol 2023; 13:1063002. [PMID: 36703993 PMCID: PMC9871890 DOI: 10.3389/fimmu.2022.1063002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Bispecific antibodies (BsAbs) form an exciting class of bio-therapeutics owing to their multispecificity. Although numerous formats have been developed, generation of hetero-tetrameric IgG1-like BsAbs having acceptable safety and pharmacokinetics profiles from a single cell culture system remains challenging due to the heterogeneous pairing between the four chains. Herein, we employed a structure-guided approach to engineer mutations in the constant domain interfaces (CH1-CL and CH3-CH3) of heavy and κ light chains to prevent heavy-light mispairing in the antigen binding fragment (Fab) region and heavy-heavy homodimerization in the Fc region. Transient co-transfection of mammalian cells with heavy and light chains of pre-existing antibodies carrying the engineered constant domains generates BsAbs with percentage purity ranging from 78% to 85%. The engineered BsAbs demonstrate simultaneous binding of both antigens, while retaining the thermal stability, Fc-mediated effector properties and FcRn binding properties of the parental antibodies. Importantly, since the variable domains were not modified, the mutations may enable BsAb formation from antibodies belonging to different germline origins and isotypes. The rationally designed mutations reported in this work could serve as a starting point for generating optimized solutions required for large scale production.
Collapse
Affiliation(s)
- Yordkhwan W. Iwasaki
- Program in Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand
| | - Kannan Tharakaraman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Vidya Subramanian
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Amnart Khongmanee
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Andrew Hatas
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Eduardo Fleischer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Troy T. Rurak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Patchara Ngok-ngam
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Phanthakarn Tit-oon
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Mathuros Ruchirawat
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand,Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand
| | - Jutamaad Satayavivad
- Program in Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand,Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand,Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Mayuree Fuangthong
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand,Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand,Program in Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok, Thailand,*Correspondence: Mayuree Fuangthong, ; Ram Sasisekharan,
| | - Ram Sasisekharan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States,*Correspondence: Mayuree Fuangthong, ; Ram Sasisekharan,
| |
Collapse
|
20
|
Nelson MH, Fritzell S, Miller R, Werchau D, Van Citters D, Nilsson A, Misher L, Ljung L, Bader R, Deronic A, Chunyk AG, Schultz L, Varas LA, Rose N, Håkansson M, Gross J, Furebring C, Pavlik P, Sundstedt A, Veitonmäki N, Ramos HJ, Säll A, Dahlman A, Bienvenue D, von Schantz L, McMahan CJ, Askmyr M, Hernandez-Hoyos G, Ellmark P. The Bispecific Tumor Antigen-Conditional 4-1BB x 5T4 Agonist, ALG.APV-527, Mediates Strong T-Cell Activation and Potent Antitumor Activity in Preclinical Studies. Mol Cancer Ther 2023; 22:89-101. [PMID: 36343381 PMCID: PMC9808321 DOI: 10.1158/1535-7163.mct-22-0395] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/16/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022]
Abstract
4-1BB (CD137) is an activation-induced costimulatory receptor that regulates immune responses of activated CD8 T and natural killer cells, by enhancing proliferation, survival, cytolytic activity, and IFNγ production. The ability to induce potent antitumor activity by stimulating 4-1BB on tumor-specific cytotoxic T cells makes 4-1BB an attractive target for designing novel immuno-oncology therapeutics. To minimize systemic immune toxicities and enhance activity at the tumor site, we have developed a novel bispecific antibody that stimulates 4-1BB function when co-engaged with the tumor-associated antigen 5T4. ALG.APV-527 was built on the basis of the ADAPTIR bispecific platform with optimized binding domains to 4-1BB and 5T4 originating from the ALLIGATOR-GOLD human single-chain variable fragment library. The epitope of ALG.APV-527 was determined to be located at domain 1 and 2 on 4-1BB using X-ray crystallography. As shown in reporter and primary cell assays in vitro, ALG.APV-527 triggers dose-dependent 4-1BB activity mediated only by 5T4 crosslinking. In vivo, ALG.APV-527 demonstrates robust antitumor responses, by inhibiting growth of established tumors expressing human 5T4 followed by a long-lasting memory immune response. ALG.APV-527 has an antibody-like half-life in cynomolgus macaques and was well tolerated at 50.5 mg/kg. ALG.APV-527 is uniquely designed for 5T4-conditional 4-1BB-mediated antitumor activity with potential to minimize systemic immune activation and hepatotoxicity while providing efficacious tumor-specific responses in a range of 5T4-expressing tumor indications as shown by robust activity in preclinical in vitro and in vivo models. On the basis of the combined preclinical dataset, ALG.APV-527 has potential as a promising anticancer therapeutic for the treatment of 5T4-expressing tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jane Gross
- Aptevo Therapeutics Inc., Seattle, Washington
| | | | | | | | | | | | - Anna Säll
- Alligator Bioscience AB, Lund, Sweden
| | | | | | | | | | | | | | - Peter Ellmark
- Alligator Bioscience AB, Lund, Sweden.,Department of Immunotechnology, Lund University, Lund, Sweden.,Corresponding Author: Peter Ellmark, Alligator Bioscience, Medicon Village, 223 81 Lund, Sweden. Phone: 467-9721-2739; E-mail:
| |
Collapse
|
21
|
Pasquiers B, Benamara S, Felices M, Nguyen L, Declèves X. Review of the Existing Translational Pharmacokinetics Modeling Approaches Specific to Monoclonal Antibodies (mAbs) to Support the First-In-Human (FIH) Dose Selection. Int J Mol Sci 2022; 23:12754. [PMID: 36361546 PMCID: PMC9657028 DOI: 10.3390/ijms232112754] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 08/27/2023] Open
Abstract
The interest in therapeutic monoclonal antibodies (mAbs) has continuously growing in several diseases. However, their pharmacokinetics (PK) is complex due to their target-mediated drug disposition (TMDD) profiles which can induce a non-linear PK. This point is particularly challenging during the pre-clinical and translational development of a new mAb. This article reviews and describes the existing PK modeling approaches used to translate the mAbs PK from animal to human for intravenous (IV) and subcutaneous (SC) administration routes. Several approaches are presented, from the most empirical models to full physiologically based pharmacokinetic (PBPK) models, with a focus on the population PK methods (compartmental and minimal PBPK models). They include the translational approaches for the linear part of the PK and the TMDD mechanism of mAbs. The objective of this article is to provide an up-to-date overview and future perspectives of the translational PK approaches for mAbs during a model-informed drug development (MIDD), since the field of PK modeling has gained recently significant interest for guiding mAbs drug development.
Collapse
Affiliation(s)
- Blaise Pasquiers
- PhinC Development, 91300 Massy, France
- Université Paris Cité, Inserm UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
| | | | | | | | - Xavier Declèves
- Université Paris Cité, Inserm UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
| |
Collapse
|
22
|
Biophysical differences in IgG1 Fc-based therapeutics relate to their cellular handling, interaction with FcRn and plasma half-life. Commun Biol 2022; 5:832. [PMID: 35982144 PMCID: PMC9388496 DOI: 10.1038/s42003-022-03787-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/01/2022] [Indexed: 01/07/2023] Open
Abstract
Antibody-based therapeutics (ABTs) are used to treat a range of diseases. Most ABTs are either full-length IgG1 antibodies or fusions between for instance antigen (Ag)-binding receptor domains and the IgG1 Fc fragment. Interestingly, their plasma half-life varies considerably, which may relate to how they engage the neonatal Fc receptor (FcRn). As such, there is a need for an in-depth understanding of how different features of ABTs affect FcRn-binding and transport behavior. Here, we report on how FcRn-engagement of the IgG1 Fc fragment compare to clinically relevant IgGs and receptor domain Fc fusions, binding to VEGF or TNF-α. The results reveal FcRn-dependent intracellular accumulation of the Fc, which is in line with shorter plasma half-life than that of full-length IgG1 in human FcRn-expressing mice. Receptor domain fusion to the Fc increases its half-life, but not to the extent of IgG1. This is mirrored by a reduced cellular recycling capacity of the Fc-fusions. In addition, binding of cognate Ag to ABTs show that complexes of similar size undergo cellular transport at different rates, which could be explained by the biophysical properties of each ABT. Thus, the study provides knowledge that should guide tailoring of ABTs regarding optimal cellular sorting and plasma half-life. Analysis of clinically approved antibody-based therapeutics reveals different structural designs, such as full-length IgG1 or Fc-fusions, entail distinct biophysical properties that affect FcRn binding, intracellular transport and plasma half-life.
Collapse
|
23
|
Gurel B, Berksoz M, Capkin E, Parlar A, Pala MC, Ozkan A, Capan Y, Daglikoca DE, Yuce M. Structural and Functional Analysis of CEX Fractions Collected from a Novel Avastin® Biosimilar Candidate and Its Innovator: A Comparative Study. Pharmaceutics 2022; 14:pharmaceutics14081571. [PMID: 36015197 PMCID: PMC9415858 DOI: 10.3390/pharmaceutics14081571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Avastin® is a humanized recombinant monoclonal antibody used to treat cancer by targeting VEGF-A to inhibit angiogenesis. SIMAB054, an Avastin® biosimilar candidate developed in this study, showed a different charge variant profile than its innovator. Thus, it is fractionated into acidic, main, and basic isoforms and collected physically by Cation Exchange Chromatography (CEX) for a comprehensive structural and functional analysis. The innovator product, fractionated into the same species and collected by the same method, is used as a reference for comparative analysis. Ultra-Performance Liquid Chromatography (UPLC) ESI-QToF was used to analyze the modifications leading to charge heterogeneities at intact protein and peptide levels. The C-terminal lysine clipping and glycosylation profiles of the samples were monitored by intact mAb analysis. The post-translational modifications, including oxidation, deamidation, and N-terminal pyroglutamic acid formation, were determined by peptide mapping analysis in the selected signal peptides. The relative binding affinities of the fractionated charge isoforms against the antigen, VEGF-A, and the neonatal receptor, FcRn, were revealed by Surface Plasmon Resonance (SPR) studies. The results show that all CEX fractions from the innovator product and the SIMAB054 shared the same structural variants, albeit in different ratios. Common glycoforms and post-translational modifications were the same, but at different percentages for some samples. The dissimilarities were mostly originating from the presence of extra C-term Lysin residues, which are prone to enzymatic degradation in the body, and thus they were previously assessed as clinically irrelevant. Another critical finding was the presence of different glyco proteoforms in different charge species, such as increased galactosylation in the acidic and afucosylation in the basic species. SPR characterization of the isolated charge variants further confirmed that basic species found in the CEX analyses of the biosimilar candidate were also present in the innovator product, although at lower amounts. The charge variants’ in vitro antigen- and neonatal receptor-binding activities varied amongst the samples, which could be further investigated in vivo with a larger sample set to reveal the impact on the pharmacokinetics of drug candidates. Minor structural differences may explain antigen-binding differences in the isolated charge variants, which is a key parameter in a comparability exercise. Consequently, such a biosimilar candidate may not comply with high regulatory standards unless the binding differences observed are justified and demonstrated not to have any clinical impact.
Collapse
Affiliation(s)
- Busra Gurel
- SUNUM Nanotechnology Research and Application Center, Sabanci University, Istanbul 34956, Turkey;
| | - Melike Berksoz
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
| | - Eda Capkin
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey;
| | - Ayhan Parlar
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey;
| | - Meltem Corbacioglu Pala
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
| | - Aylin Ozkan
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
| | - Yılmaz Capan
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
| | - Duygu Emine Daglikoca
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
- Correspondence: (D.E.D.); (M.Y.)
| | - Meral Yuce
- SUNUM Nanotechnology Research and Application Center, Sabanci University, Istanbul 34956, Turkey;
- Correspondence: (D.E.D.); (M.Y.)
| |
Collapse
|
24
|
Optimized Methods for Analytical and Functional Comparison of Biosimilar mAb Drugs: A Case Study for Avastin, Mvasi, and Zirabev. Sci Pharm 2022. [DOI: 10.3390/scipharm90020036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bevacizumab is a humanized therapeutic monoclonal antibody used to reduce angiogenesis, a hallmark of cancer, by binding to VEGF-A. Many pharmaceutical companies have developed biosimilars of Bevacizumab in the last decade. The official reports provided by the FDA and EMA summarize the analytical performance of biosimilars as compared to the originators without giving detailed analytical procedures. In the current study, several key methods were optimized and reported for analytical and functional comparison of bevacizumab originators (Avastin, Altuzan) and approved commercial biosimilars (Zirabev and Mvasi). This case study presents a comparative analysis of a set of biosimilars under optimized analytical conditions for the first time in the literature. The chemical structure of all products was analyzed at intact protein and peptide levels by high-resolution mass spectrometry; the major glycoforms and posttranslational modifications, including oxidation, deamidation, N-terminal PyroGlu addition, and C-terminal Lys clipping, were compared. The SPR technique was used to reveal antigen and some receptor binding kinetics of all products, and the ELISA technique was used for C1q binding affinity analysis. Finally, the inhibition performance of the samples was evaluated by an MTS-based proliferation assay in vitro. Major glycoforms were similar, with minor differences among the samples. Posttranslational modifications, except C-terminal Lys, were determined similarly, while unclipped Lys percentage was higher in Zirabev. The binding kinetics for VEGF, FcRn, FcγRIa, and C1q were similar or in the value range of originators. The anti-proliferative effect of Zirabev was slightly higher than the originators and Mvasi. The analysis of biosimilars under the same conditions could provide a new aspect to the literature in terms of the applied analytical techniques. Further studies in this field would be helpful to better understand the inter-comparability of the biosimilars.
Collapse
|
25
|
Development and Functional Characterization of a Versatile Radio-/Immunotheranostic Tool for Prostate Cancer Management. Cancers (Basel) 2022; 14:cancers14081996. [PMID: 35454902 PMCID: PMC9027777 DOI: 10.3390/cancers14081996] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In previous studies, we described a modular Chimeric Antigen Receptor (CAR) T cell platform which we termed UniCAR. In contrast to conventional CARs, the interaction of UniCAR T cells does not occur directly between the CAR T cell and the tumor cell but is mediated via bispecific adaptor molecules so-called target modules (TMs). Here we present the development and functional characterization of a novel IgG4-based TM, directed to the tumor-associated antigen (TAA) prostate stem cell antigen (PSCA), which is overexpressed in prostate cancer (PCa). We show that this anti-PSCA IgG4-TM cannot only be used for (i) redirection of UniCAR T cells to PCa cells but also for (ii) positron emission tomography (PET) imaging, and (iii) alpha particle-based endoradiotherapy. For radiolabeling, the anti-PSCA IgG4-TM was conjugated with the chelator DOTAGA. PET imaging was performed using the 64Cu-labeled anti-PSCA IgG4-TM. According to PET imaging, the anti-PSCA IgG4-TM accumulates with high contrast in the PSCA-positive tumors of experimental mice without visible uptake in other organs. For endoradiotherapy the anti-PSCA IgG4-TM-DOTAGA conjugate was labeled with 225Ac3+. Targeted alpha therapy resulted in tumor control over 60 days after a single injection of the 225Ac-labeled TM. The favorable pharmacological profile of the anti-PSCA IgG4-TM, and its usage for (i) imaging, (ii) targeted alpha therapy, and (iii) UniCAR T cell immunotherapy underlines the promising radio-/immunotheranostic capabilities for the diagnostic imaging and treatment of PCa. Abstract Due to its overexpression on the surface of prostate cancer (PCa) cells, the prostate stem cell antigen (PSCA) is a potential target for PCa diagnosis and therapy. Here we describe the development and functional characterization of a novel IgG4-based anti-PSCA antibody (Ab) derivative (anti-PSCA IgG4-TM) that is conjugated with the chelator DOTAGA. The anti-PSCA IgG4-TM represents a multimodal immunotheranostic compound that can be used (i) as a target module (TM) for UniCAR T cell-based immunotherapy, (ii) for diagnostic positron emission tomography (PET) imaging, and (iii) targeted alpha therapy. Cross-linkage of UniCAR T cells and PSCA-positive tumor cells via the anti-PSCA IgG4-TM results in efficient tumor cell lysis both in vitro and in vivo. After radiolabeling with 64Cu2+, the anti-PSCA IgG4-TM was successfully applied for high contrast PET imaging. In a PCa mouse model, it showed specific accumulation in PSCA-expressing tumors, while no uptake in other organs was observed. Additionally, the DOTAGA-conjugated anti-PSCA IgG4-TM was radiolabeled with 225Ac3+ and applied for targeted alpha therapy. A single injection of the 225Ac-labeled anti-PSCA IgG4-TM was able to significantly control tumor growth in experimental mice. Overall, the novel anti-PSCA IgG4-TM represents an attractive first member of a novel group of radio-/immunotheranostics that allows diagnostic imaging, endoradiotherapy, and CAR T cell immunotherapy.
Collapse
|
26
|
Tsai TI, Khalili JS, Gilchrist M, Waight AB, Cohen D, Zhuo S, Zhang Y, Ding M, Zhu H, Mak ANS, Zhu Y, Goulet DR. ACE2-Fc fusion protein overcomes viral escape by potently neutralizing SARS-CoV-2 variants of concern. Antiviral Res 2022; 199:105271. [PMID: 35240221 PMCID: PMC8882475 DOI: 10.1016/j.antiviral.2022.105271] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/06/2022] [Accepted: 02/23/2022] [Indexed: 01/01/2023]
Abstract
COVID-19, an infectious disease caused by the SARS-CoV-2 virus, emerged globally in early 2020 and has remained a serious public health issue. To date, although several preventative vaccines have been approved by FDA and EMA, vaccinated individuals increasingly suffer from breakthrough infections. Therapeutic antibodies may provide an alternative strategy to neutralize viral infection and treat serious cases; however, the clinical data and our experiments show that some FDA-approved monoclonal antibodies lose function against COVID-19 variants such as Omicron. Therefore, in this study, we present a novel therapeutic agent, SI-F019, an ACE2-Fc fusion protein whose neutralization efficiency is not compromised, but actually strengthened, by the mutations of dominant variants including Omicron. Comprehensive biophysical analyses revealed the mechanism of increased inhibition to be enhanced interaction of SI-F019 with all the tested spike variants, in contrast to monoclonal antibodies which tended to show weaker binding to some variants. The results imply that SI-F019 may be a broadly useful agent for treatment of COVID-19.
Collapse
Affiliation(s)
- Tsung-I Tsai
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | | | - Mark Gilchrist
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | | | - Daniella Cohen
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | - Shi Zhuo
- Chengdu Strait Technology Industry Development Park, Wenjiang Dist., Chengdu, Sichuan, 611130, China
| | - Yong Zhang
- Chengdu Strait Technology Industry Development Park, Wenjiang Dist., Chengdu, Sichuan, 611130, China
| | - Muran Ding
- Chengdu Strait Technology Industry Development Park, Wenjiang Dist., Chengdu, Sichuan, 611130, China
| | - Hai Zhu
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | | | - Yi Zhu
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA
| | - Dennis R Goulet
- SystImmune Inc., 15318 NE 95th St., Redmond, WA, 98052, USA.
| |
Collapse
|
27
|
Slavetinsky CJ, Hauser JN, Gekeler C, Slavetinsky J, Geyer A, Kraus A, Heilingbrunner D, Wagner S, Tesar M, Krismer B, Kuhn S, Ernst CM, Peschel A. Sensitizing Staphylococcus aureus to antibacterial agents by decoding and blocking the lipid flippase MprF. eLife 2022; 11:66376. [PMID: 35044295 PMCID: PMC8806190 DOI: 10.7554/elife.66376] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
The pandemic of antibiotic resistance represents a major human health threat demanding new antimicrobial strategies. MprF is the synthase and flippase of the phospholipid lysyl-phosphatidylglycerol that increases virulence and resistance of methicillin-resistant Staphylococcus aureus (MRSA) and other pathogens to cationic host defense peptides and antibiotics. With the aim to design MprF inhibitors that could sensitize MRSA to antimicrobial agents and support the clearance of staphylococcal infections with minimal selection pressure, we developed MprF-targeting monoclonal antibodies, which bound and blocked the MprF flippase subunit. Antibody M-C7.1 targeted a specific loop in the flippase domain that proved to be exposed at both sides of the bacterial membrane, thereby enhancing the mechanistic understanding of bacterial lipid translocation. M-C7.1 rendered MRSA susceptible to host antimicrobial peptides and antibiotics such as daptomycin, and it impaired MRSA survival in human phagocytes. Thus, MprF inhibitors are recommended for new anti-virulence approaches against MRSA and other bacterial pathogens.
Collapse
Affiliation(s)
| | | | - Cordula Gekeler
- Department of Infection Biology, Eberhard Karls University Tübingen
| | | | - André Geyer
- Department of Infection Biology, Eberhard Karls University Tübingen
| | | | | | - Samuel Wagner
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen
| | | | - Bernhard Krismer
- Department of Infection Biology, Eberhard Karls University Tübingen
| | - Sebastian Kuhn
- Department of Infection Biology, Eberhard Karls University Tübingen
| | - Christoph M Ernst
- Department of Molecular Biology and Center for Computational and Integrative Biology, Broad Institute
| | - Andreas Peschel
- Department of Infection Biology, Eberhard Karls University Tübingen
| |
Collapse
|
28
|
Ménochet K, Yu H, Wang B, Tibbitts J, Hsu CP, Kamath AV, Richter WF, Baumann A. Non-human primates in the PKPD evaluation of biologics: Needs and options to reduce, refine, and replace. A BioSafe White Paper. MAbs 2022; 14:2145997. [PMID: 36418217 PMCID: PMC9704389 DOI: 10.1080/19420862.2022.2145997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
Monoclonal antibodies (mAbs) deliver great benefits to patients with chronic and/or severe diseases thanks to their strong specificity to the therapeutic target. As a result of this specificity, non-human primates (NHP) are often the only preclinical species in which therapeutic antibodies cross-react with the target. Here, we highlight the value and limitations that NHP studies bring to the design of safe and efficient early clinical trials. Indeed, data generated in NHPs are integrated with in vitro information to predict the concentration/effect relationship in human, and therefore the doses to be tested in first-in-human trials. The similarities and differences in the systems defining the pharmacokinetics and pharmacodynamics (PKPD) of mAbs in NHP and human define the nature and the potential of the preclinical investigations performed in NHPs. Examples have been collated where the use of NHP was either pivotal to the design of the first-in-human trial or, inversely, led to the termination of a project prior to clinical development. The potential impact of immunogenicity on the results generated in NHPs is discussed. Strategies to optimize the use of NHPs for PKPD purposes include the addition of PD endpoints in safety assessment studies and the potential re-use of NHPs after non-terminal studies or cassette dosing several therapeutic agents of interest. Efforts are also made to reduce the use of NHPs in the industry through the use of in vitro systems, alternative in vivo models, and in silico approaches. In the case of prediction of ocular PK, the body of evidence gathered over the last two decades renders the use of NHPs obsolete. Expert perspectives, advantages, and pitfalls with these alternative approaches are shared in this review.
Collapse
Affiliation(s)
| | - Hongbin Yu
- R&D Project Management and Development Strategies, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Bonnie Wang
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Inc, Princeton, NJ, USA
| | - Jay Tibbitts
- Nonclinical Development, South San Francisco, CA, USA
| | - Cheng-Pang Hsu
- Preclinical Development and Clinical Pharmacology, AskGene Pharma Inc, Camarillo, CA, USA
| | - Amrita V. Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, South San Francisco, CA, USA
| | - Wolfgang F. Richter
- Roche Pharma Research and Early Development, Roche Innovation, Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Andreas Baumann
- R&D, Bayer Pharma AG, Berlin, Germany & Non-clinical Biotech Consulting, Potsdam, Germany °(° present affiliation)
| |
Collapse
|
29
|
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: 7.7] [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
|
30
|
Gstöttner C, Hook M, Christopeit T, Knaupp A, Schlothauer T, Reusch D, Haberger M, Wuhrer M, Domínguez-Vega E. Affinity Capillary Electrophoresis-Mass Spectrometry as a Tool to Unravel Proteoform-Specific Antibody-Receptor Interactions. Anal Chem 2021; 93:15133-15141. [PMID: 34739220 PMCID: PMC8600502 DOI: 10.1021/acs.analchem.1c03560] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Monoclonal antibody (mAb) pharmaceuticals consist of a plethora of different proteoforms with different functional characteristics, including pharmacokinetics and pharmacodynamics, requiring their individual assessment. Current binding techniques do not distinguish between coexisting proteoforms requiring tedious production of enriched proteoforms. Here, we have developed an approach based on mobility shift-affinity capillary electrophoresis-mass spectrometry (ACE-MS), which permitted us to determine the binding of coexisting mAb proteoforms to Fc receptors (FcRs). For high-sensitivity MS analysis, we used a sheathless interface providing adequate mAb sensitivity allowing functional characterization of mAbs with a high sensitivity and dynamic range. As a model system, we focused on the interaction with the neonatal FcR (FcRn), which determines the half-life of mAbs. Depending on the oxidation status, proteoforms exhibited different electrophoretic mobility shifts in the presence of FcRn, which could be used to determine their affinity. We confirmed the decrease of the FcRn affinity with antibody oxidation and observed a minor glycosylation effect, with higher affinities for galactosylated glycoforms. Next to relative binding, the approach permits the determination of individual KD values in solution resulting in values of 422 and 139 nM for double-oxidized and non-oxidized variants. Hyphenation with native MS provides unique capabilities for simultaneous heterogeneity assessment for mAbs, FcRn, and complexes formed. The latter provides information on binding stoichiometry revealing 1:1 and 1:2 for antibody/FcRn complexes. The use of differently engineered Fc-only constructs allowed distinguishing between symmetric and asymmetric binding. The approach opens up unique possibilities for proteoform-resolved antibody binding studies to FcRn and can be extended to other FcRs and protein interactions.
Collapse
Affiliation(s)
- Christoph Gstöttner
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| | - Michaela Hook
- Pharma Technical Development Penzberg, Roche Diagnostics GmbH, Penzberg 82377, Germany
| | - Tony Christopeit
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Alexander Knaupp
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Tilman Schlothauer
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Dietmar Reusch
- Pharma Technical Development Penzberg, Roche Diagnostics GmbH, Penzberg 82377, Germany
| | - Markus Haberger
- Pharma Technical Development Penzberg, Roche Diagnostics GmbH, Penzberg 82377, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| | - Elena Domínguez-Vega
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| |
Collapse
|
31
|
Nath N, Godat B, Flemming R, Urh M. Deciphering the Interaction between Neonatal Fc Receptor and Antibodies Using a Homogeneous Bioluminescent Immunoassay. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:1211-1221. [PMID: 34312257 PMCID: PMC8358922 DOI: 10.4049/jimmunol.2100181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/22/2021] [Indexed: 11/19/2022]
Abstract
Long half-life of therapeutic Abs and Fc fusion proteins is crucial to their efficacy and is, in part, regulated by their interaction with neonatal Fc receptor (FcRn). However, the current methods (e.g., surface plasmon resonance and biolayer interferometry) for measurement of interaction between IgG and FcRn (IgG/FcRn) require either FcRn or IgG to be immobilized on the surface, which is known to introduce experimental artifacts and have led to conflicting data. To study IgG/FcRn interactions in solution, without a need for surface immobilization, we developed a novel (to our knowledge), solution-based homogeneous binding immunoassay based on NanoBiT luminescent protein complementation technology. We optimized the assay (NanoBiT FcRn assay) for human FcRn, mouse FcRn, rat FcRn, and cynomolgus FcRn and used them to determine the binding affinities of a panel of eight Abs. Assays could successfully capture the modulation in IgG/FcRn binding based on changes in Fc fragment of the Abs. We also looked at the individual contribution of Fc and F(ab)2 on the IgG/FcRn interaction and found that Fc is the main driver for the interaction at pH 6. Our work highlights the importance of using orthogonal methods to validate affinity data generated using biosensor platforms. Moreover, the simple add-and-read format of the NanoBiT FcRn assay is amenable for high-throughput screening during early Ab discovery phase.
Collapse
Affiliation(s)
- Nidhi Nath
- Research and Development Department, Promega Corp., Madison, WI
| | - Becky Godat
- Research and Development Department, Promega Corp., Madison, WI
| | - Rod Flemming
- Research and Development Department, Promega Corp., Madison, WI
| | - Marjeta Urh
- Research and Development Department, Promega Corp., Madison, WI
| |
Collapse
|
32
|
Development of novel immunoprophylactic agents against multidrug resistant Gram-negative bacterial infections. Antimicrob Agents Chemother 2021; 65:e0098521. [PMID: 34370589 DOI: 10.1128/aac.00985-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The widespread emergence of antibiotic resistance including multidrug resistance in Gram negative (G-) bacterial pathogens poses a critical challenge to the current antimicrobial armamentarium. Antibody-drug conjugates (ADCs), primarily used in anti-cancer therapy, offer a promising treatment alternative due to their ability to deliver a therapeutic molecule while simultaneously activating the host immune response. The Cloudbreak® platform is being used to develop ADCs to treat infectious diseases, composed of a therapeutic targeting moiety (TM) attached via a non-cleavable linker to an effector moiety (EM) to treat infectious diseases. In this proof-of-concept study, 21 novel dimeric peptidic molecules (TMs) were evaluated for activity against a screening panel of G- pathogens. The activity of the TMs were not impacted by existing drug resistance. Potent TMs were conjugated to the Fc fragment of human IgG1 (EM) resulting in 4 novel ADCs. These ADCs were evaluated for immunoprophylactic efficacy in a neutropenic mouse model of deep thigh infection. In colistin-sensitive infections, 3 of the 4 ADCs offered similar protection as therapeutically dosed colistin while CTC-171 offered enhanced protection. The efficacy of these ADCs was unchanged in colistin-resistant infections. Together, these results indicate that the ADCs used here are capable of potent binding to G- pathogens regardless of LPS modifications that otherwise lead to antibiotic resistance and support further exploration of ADCs in the treatment of drug resistant G- bacterial infections.
Collapse
|
33
|
Shmool TA, Martin LK, Bui-Le L, Moya-Ramirez I, Kotidis P, Matthews RP, Venter GA, Kontoravdi C, Polizzi KM, Hallett JP. An experimental approach probing the conformational transitions and energy landscape of antibodies: a glimmer of hope for reviving lost therapeutic candidates using ionic liquid. Chem Sci 2021; 12:9528-9545. [PMID: 34349928 PMCID: PMC8278930 DOI: 10.1039/d1sc02520a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/26/2021] [Indexed: 12/26/2022] Open
Abstract
Understanding protein folding in different environmental conditions is fundamentally important for predicting protein structures and developing innovative antibody formulations. While the thermodynamics and kinetics of folding and unfolding have been extensively studied by computational methods, experimental methods for determining antibody conformational transition pathways are lacking. Motivated to fill this gap, we prepared a series of unique formulations containing a high concentration of a chimeric immunoglobin G4 (IgG4) antibody with different excipients in the presence and absence of the ionic liquid (IL) choline dihydrogen phosphate. We determined the effects of different excipients and IL on protein thermal and structural stability by performing variable temperature circular dichroism and bio-layer interferometry analyses. To further rationalise the observations of conformational changes with temperature, we carried out molecular dynamics simulations on a single antibody binding fragment from IgG4 in the different formulations, at low and high temperatures. We developed a methodology to study the conformational transitions and associated thermodynamics of biomolecules, and we showed IL-induced conformational transitions. We showed that the increased propensity for conformational change was driven by preferential binding of the dihydrogen phosphate anion to the antibody fragment. Finally, we found that a formulation containing IL with sugar, amino acids and surfactant is a promising candidate for stabilising proteins against conformational destabilisation and aggregation. We hope that ultimately, we can help in the quest to understand the molecular basis of the stability of antibodies and protein misfolding phenomena and offer new candidate formulations with the potential to revive lost therapeutic candidates.
Collapse
Affiliation(s)
- Talia A Shmool
- Department of Chemical Engineering, Imperial College London South Kensington Campus London SW7 2AZ UK +44 (0)20 7594 5388
| | - Laura K Martin
- Department of Engineering Science, University of Oxford Parks Road Oxford OX1 3PJ UK
| | - Liem Bui-Le
- Department of Chemical Engineering, Imperial College London South Kensington Campus London SW7 2AZ UK +44 (0)20 7594 5388
| | - Ignacio Moya-Ramirez
- Department of Chemical Engineering, Imperial College London South Kensington Campus London SW7 2AZ UK +44 (0)20 7594 5388
| | - Pavlos Kotidis
- Department of Chemical Engineering, Imperial College London South Kensington Campus London SW7 2AZ UK +44 (0)20 7594 5388
| | - Richard P Matthews
- Department of Chemical Engineering, Imperial College London South Kensington Campus London SW7 2AZ UK +44 (0)20 7594 5388
| | - Gerhard A Venter
- Scientific Computing Research Unit, Department of Chemistry, University of Cape Town Rondebosch Cape Town 7701 South Africa
| | - Cleo Kontoravdi
- Department of Chemical Engineering, Imperial College London South Kensington Campus London SW7 2AZ UK +44 (0)20 7594 5388
| | - Karen M Polizzi
- Department of Chemical Engineering, Imperial College London South Kensington Campus London SW7 2AZ UK +44 (0)20 7594 5388
| | - Jason P Hallett
- Department of Chemical Engineering, Imperial College London South Kensington Campus London SW7 2AZ UK +44 (0)20 7594 5388
| |
Collapse
|
34
|
Fieux M, Le Quellec S, Bartier S, Coste A, Louis B, Giroudon C, Nourredine M, Bequignon E. FcRn as a Transporter for Nasal Delivery of Biologics: A Systematic Review. Int J Mol Sci 2021; 22:6475. [PMID: 34204226 PMCID: PMC8234196 DOI: 10.3390/ijms22126475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022] Open
Abstract
FcRn plays a major role in regulating immune homeostasis, but it is also able to transport biologics across cellular barriers. The question of whether FcRn could be an efficient transporter of biologics across the nasal epithelial barrier is of particular interest, as it would allow a less invasive strategy for the administration of biologics in comparison to subcutaneous, intramuscular or intravenous administrations, which are often used in clinical practice. A focused systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. It was registered on the international prospective register of systematic reviews PROSPERO, which helped in identifying articles that met the inclusion criteria. Clinical and preclinical studies involving FcRn and the nasal delivery of biologics were screened, and the risk of bias was assessed across studies using the Oral Health Assessment Tool (OHAT). Among the 12 studies finally included in this systematic review (out of the 758 studies screened), 11 demonstrated efficient transcytosis of biologics through the nasal epithelium. Only three studies evaluated the potential toxicity of biologics' intranasal delivery, and they all showed that it was safe. This systematic review confirmed that FcRn is expressed in the nasal airway and the olfactory epithelium, and that FcRn may play a role in IgG and/or IgG-derived molecule-transcytosis across the airway epithelium. However, additional research is needed to better characterize the pharmacokinetic and pharmacodynamic properties of biologics after their intranasal delivery.
Collapse
Affiliation(s)
- Maxime Fieux
- Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Service d’ORL, D’otoneurochirurgie et de Chirurgie Cervico-Faciale, Pierre Bénite, CEDEX, F-69495 Lyon, France
- Université de Lyon, Université Lyon 1, F-69003 Lyon, France; (S.L.Q.); (M.N.)
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
| | - Sandra Le Quellec
- Université de Lyon, Université Lyon 1, F-69003 Lyon, France; (S.L.Q.); (M.N.)
- Hospices Civils de Lyon, Hôpital Cardiologique Louis Pradel, Unité D’hémostase Clinique, CEDEX, F-69500 Bron, France
- EA 4609 Hémostase et Cancer, Université Claude Bernard Lyon 1, F-69372 Lyon, France
- Hospices Civils de Lyon, Centre de Biologie et de Pathologie Est, Service D’hématologie Biologique, CEDEX, F-69500 Bron, France
| | - Sophie Bartier
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
- Service d’ORL, de Chirurgie Cervico Faciale, Hôpital Henri Mondor, Assistance Publique des Hôpitaux de Paris, F-94000 Créteil, France
| | - André Coste
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
- Service d’ORL, de Chirurgie Cervico Faciale, Centre Hospitalier Intercommunal de Créteil, F-94010 Créteil, France
| | - Bruno Louis
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
| | - Caroline Giroudon
- Hospices Civils de Lyon, Service de la Documentation Centrale, CEDEX, F-69424 Lyon, France;
| | - Mikail Nourredine
- Université de Lyon, Université Lyon 1, F-69003 Lyon, France; (S.L.Q.); (M.N.)
- Hospices Civils de Lyon, Service de Biostatistique et Bioinformatique, F-69003 Lyon, France
- CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR 5558, F-69100 Villeurbanne, France
| | - Emilie Bequignon
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
- Service d’ORL, de Chirurgie Cervico Faciale, Centre Hospitalier Intercommunal de Créteil, F-94010 Créteil, France
| |
Collapse
|
35
|
Singh R, Moreno M, Stanimirovic D. Comparison of Various Approaches to Translate Non-Linear Pharmacokinetics of Monoclonal Antibodies from Cynomolgus Monkey to Human. Eur J Drug Metab Pharmacokinet 2021; 46:555-567. [PMID: 34120326 DOI: 10.1007/s13318-021-00691-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVES The prediction of pharmacokinetics of monoclonal antibodies (mAbs) exhibiting non-linear pharmacokinetics in preclinical species to human is challenging, and very limited scientific work has been published in this field of research. Therefore, we have conducted an elaborate comparative assessment to determine the most reliable preclinical to clinical scaling strategy for mAbs with non-linear pharmacokinetics. METHODS We have compared three different scaling approaches to predict human pharmacokinetics from cynomolgus monkey. In the first approach, cynomolgus monkey pharmacokinetic parameters estimated using a two-compartment model with parallel linear and non-linear elimination were allometrically scaled to simulate human pharmacokinetics. In the second approach, allometric exponents were integrated with a minimal physiologically based pharmacokinetic (mPBPK) model to translate human pharmacokinetics. In the third approach, we have employed a species time-invariant method, wherein a two-compartment model with parallel linear and non-linear elimination was used as a framework model for simulation of the human profile. RESULTS Human exposure parameters projected by an integrated allometric method were only within two fold for approximately 45-70% of predictions at different doses of five mAbs evaluated, while approximately 70-80% of Cmax and AUC predictions by integrated mPBPK modelling as well as the species time-invariant method were within two-fold error. The average fold error for clearance predictions by the integrated mPBPK method was 1.10-1.45 fold, whilst for the species time-variant and integrated allometric methods, the average fold error was between 1.04 and 1.37 fold and 1.24 and 2.13 fold, respectively. CONCLUSIONS Our findings suggest that the species time-variant method and mPBPK proposed by us can be employed to reliably translate non-linear pharmacokinetics of mAbs from cynomolgus monkey to human.
Collapse
Affiliation(s)
- Renu Singh
- Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada. .,Global DMPK, Research IVIVT, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA. .,Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Montreal, QC, Canada.
| | - Maria Moreno
- Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Danica Stanimirovic
- Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| |
Collapse
|
36
|
Matsuda Y, Chakrabarti A, Takahashi K, Yamada K, Nakata K, Okuzumi T, Mendelsohn BA. Chromatographic analysis of site-specific antibody-drug conjugates produced by AJICAP first-generation technology using a recombinant FcγIIIa receptor-ligand affinity column. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1177:122753. [PMID: 34098178 DOI: 10.1016/j.jchromb.2021.122753] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/24/2021] [Accepted: 04/29/2021] [Indexed: 01/08/2023]
Abstract
Commercially approved conventional antibody-drug conjugates (ADCs) are produced as heterogeneous mixtures containing a stochastic distribution of payloads decorating the antibody molecules resulting in decreased efficacy and thus lowering their therapeutic index. Control of the DAR and conjugation site in the development of next-generation ADCs is believed to assist in increasing the therapeutic index of these targeted biologics leading to overall enhanced clinical efficacy and reduced toxicity. A chemical site-specific conjugation technology termed AJICAP® allows ADC developers to control both the location and quantity of the payload conjugation to an antibody. Furthermore, this simplified ADC composition enables a streamlined chemical analysis. Here we report the chromatographic separation of site-specific ADCs produced by AJICAP® technology using an analytical affinity chromatography HPLC column containing a recombinant FcγIIIa receptor-ligand immobilized on a non-porous polymer resin (NPR). These HPLC analyses provided visually clear chromatogram results reflecting the heterogeneity of each ADC. The affinity strength was also measured by biolayer interferometry (BLI) and predicted by molecular structure analysis. The results indicate that AJICAP® technology is a promising solution to link hydrophobic payloads to antibodies without compromising antibody receptor function. This study also shows that FcγIIIa-NPR column can be used to characterize site-specific conjugated ADCs compared to ADCs synthesized using conventional methods.
Collapse
Affiliation(s)
- Yutaka Matsuda
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan.
| | - Atis Chakrabarti
- Tosoh Bioscience, 3604 Horizon Drive, Suite 100, King of Prussia, PA 19406, USA
| | | | - Kei Yamada
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Kunio Nakata
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Tatsuya Okuzumi
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Brian A Mendelsohn
- Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, CA 92121, USA
| |
Collapse
|
37
|
Qi T, Cao Y. In Translation: FcRn across the Therapeutic Spectrum. Int J Mol Sci 2021; 22:3048. [PMID: 33802650 PMCID: PMC8002405 DOI: 10.3390/ijms22063048] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
As an essential modulator of IgG disposition, the neonatal Fc receptor (FcRn) governs the pharmacokinetics and functions many therapeutic modalities. In this review, we thoroughly reexamine the hitherto elucidated biological and thermodynamic properties of FcRn to provide context for our assessment of more recent advances, which covers antigen-binding fragment (Fab) determinants of FcRn affinity, transgenic preclinical models, and FcRn targeting as an immune-complex (IC)-clearing strategy. We further comment on therapeutic antibodies authorized for treating SARS-CoV-2 (bamlanivimab, casirivimab, and imdevimab) and evaluate their potential to saturate FcRn-mediated recycling. Finally, we discuss modeling and simulation studies that probe the quantitative relationship between in vivo IgG persistence and in vitro FcRn binding, emphasizing the importance of endosomal transit parameters.
Collapse
Affiliation(s)
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA;
| |
Collapse
|
38
|
Patel DD, Bussel JB. Neonatal Fc receptor in human immunity: Function and role in therapeutic intervention. J Allergy Clin Immunol 2021; 146:467-478. [PMID: 32896307 DOI: 10.1016/j.jaci.2020.07.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/08/2023]
Abstract
The humoral immune response provides specific, long-lived protection against invading pathogens, via immunoglobulin production and other memory functions. IgG, the most abundant immunoglobulin isotype, has the longest half-life and protects against bacterial and viral infections. The neonatal Fc receptor (FcRn) transports IgG across barriers, for example, the placenta, enhancing fetal humoral immunity to levels similar to their mothers'. Importantly, FcRn, by protecting IgG from intracellular degradation, results in an approximately 21-day circulating IgG half-life and high plasma levels; similarly, FcRn recycles albumin and is the portal of entry for enteric cytopathic human orphan (echo) virus infection. Dysregulated immune responses may lead to antibodies against self-antigens (autoantibodies), resulting in organ-specific or systemic autoimmune diseases. Autoantibody-mediated diseases have been treated by nonspecific immunoglobulin-lowering/modulating therapies, including immunoadsorption, plasma exchange, and high-dose intravenous immunoglobulin. However, targeting FcRn with specific inhibitors results in reduction in only IgG levels. The effectiveness of FcRn inhibitors in autoimmune diseases, including myasthenia gravis and immune thrombocytopenia, provides further evidence that IgG is a primary driver in these autoantibody-mediated diseases. We describe the role of FcRn in human biology, including insights that clinical testing of FcRn inhibitors have provided into FcRn biology and autoimmune disease mechanisms, allowing fact-based speculation on their therapeutic potential.
Collapse
Affiliation(s)
- Dhavalkumar D Patel
- UCB Pharma, Brussels, Belgium; University of North Carolina, Chapel Hill, NC.
| | | |
Collapse
|
39
|
Kraft TE, Richter WF, Emrich T, Knaupp A, Schuster M, Wolfert A, Kettenberger H. Heparin chromatography as an in vitro predictor for antibody clearance rate through pinocytosis. MAbs 2021; 12:1683432. [PMID: 31769731 PMCID: PMC6927760 DOI: 10.1080/19420862.2019.1683432] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pharmacokinetic (PK) properties of therapeutic antibodies directly affect efficacy, dose and dose intervals, application route and tissue penetration. In indications where health-care providers and patients can choose between several efficacious and safe therapeutic options, convenience (determined by dosing interval or route of application), which is mainly driven by PK properties, can affect drug selection. Therapeutic antibodies can have greatly different PK even if they have identical Fc domains and show no target-mediated drug disposition. Biophysical properties like surface charge or hydrophobicity, and binding to surrogates for high abundant off-targets (e.g., baculovirus particles, Chinese hamster ovary cell membrane proteins) were proposed to be responsible for these differences. Here, we used heparin chromatography to separate a polyclonal mix of endogenous human IgGs (IVIG) into fractions that differ in their PK properties. Heparin was chosen as a surrogate for highly negatively charged glycocalyx components on endothelial cells, which are among the main contributors to nonspecific clearance. By directly correlating heparin retention time with clearance, we identified heparin chromatography as a tool to assess differences in unspecific cell-surface interaction and the likelihood for increased pinocytotic uptake and degradation. Building on these results, we combined predictors for FcRn-mediated recycling and cell-surface interaction. The combination of heparin and FcRn chromatography allow identification of antibodies with abnormal PK by mimicking the major root causes for fast, non-target-mediated, clearance of therapeutic, Fc-containing proteins.
Collapse
Affiliation(s)
- Thomas E Kraft
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Wolfgang F Richter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Thomas Emrich
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Alexander Knaupp
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Michaela Schuster
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Andreas Wolfert
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Hubert Kettenberger
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
40
|
Ladel S, Maigler F, Flamm J, Schlossbauer P, Handl A, Hermann R, Herzog H, Hummel T, Mizaikoff B, Schindowski K. Impact of Glycosylation and Species Origin on the Uptake and Permeation of IgGs through the Nasal Airway Mucosa. Pharmaceutics 2020; 12:E1014. [PMID: 33114132 PMCID: PMC7690786 DOI: 10.3390/pharmaceutics12111014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/18/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022] Open
Abstract
Although we have recently reported the involvement of neonatal Fc receptor (FcRn) in intranasal transport, the transport mechanisms are far from being elucidated. Ex vivo porcine olfactory tissue, primary cells from porcine olfactory epithelium (OEPC) and the human cell line RPMI 2650 were used to evaluate the permeation of porcine and human IgG antibodies through the nasal mucosa. IgGs were used in their wild type and deglycosylated form to investigate the impact of glycosylation. Further, the expression of FcRn and Fc-gamma receptor (FCGR) and their interaction with IgG were analyzed. Comparable permeation rates for human and porcine IgG were observed in OEPC, which display the highest expression of FcRn. Only traces of porcine IgGs could be recovered at the basolateral compartment in ex vivo olfactory tissue, while human IgGs reached far higher levels. Deglycosylated human IgG showed significantly higher permeation in comparison to the wild type in RPMI 2650 and OEPC, but insignificantly elevated in the ex vivo model. An immunoprecipitation with porcine primary cells and tissue identified FCGR2 as a potential interaction partner in the nasal mucosa. Glycosylation sensitive receptors appear to be involved in the uptake, transport, but also degradation of therapeutic IgGs in the airway epithelial layer.
Collapse
Affiliation(s)
- Simone Ladel
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Frank Maigler
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Johannes Flamm
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Patrick Schlossbauer
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
| | - Alina Handl
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Rebecca Hermann
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
| | - Helena Herzog
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
- Faculty of Natural Science, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Thomas Hummel
- Smell & Taste Clinic, Department of Otorhinolaryngology, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany;
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany;
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany; (S.L.); (F.M.); (J.F.); (P.S.); (A.H.); (R.H.); (H.H.)
| |
Collapse
|
41
|
Hubbard JJ, Pyzik M, Rath T, Kozicky LK, Sand KM, Gandhi AK, Grevys A, Foss S, Menzies SC, Glickman JN, Fiebiger E, Roopenian DC, Sandlie I, Andersen JT, Sly LM, Baker K, Blumberg RS. FcRn is a CD32a coreceptor that determines susceptibility to IgG immune complex-driven autoimmunity. J Exp Med 2020; 217:e20200359. [PMID: 32658257 PMCID: PMC7537387 DOI: 10.1084/jem.20200359] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/21/2020] [Accepted: 05/29/2020] [Indexed: 12/18/2022] Open
Abstract
IgG immune complexes (ICs) promote autoimmunity through binding fragment crystallizable (Fc) γ-receptors (FcγRs). Of these, the highly prevalent FcγRIIa (CD32a) histidine (H)-131 variant (CD32aH) is strongly linked to human autoimmune diseases through unclear mechanisms. We show that, relative to the CD32a arginine (R)-131 (CD32aR) variant, CD32aH more avidly bound human (h) IgG1 IC and formed a ternary complex with the neonatal Fc receptor (FcRn) under acidic conditions. In primary human and mouse cells, both CD32a variants required FcRn to induce innate and adaptive immune responses to hIgG1 ICs, which were augmented in the setting of CD32aH. Conversely, FcRn induced responses to IgG IC independently of classical FcγR, but optimal responses required FcRn and FcγR. Finally, FcRn blockade decreased inflammation in a rheumatoid arthritis model without reducing circulating autoantibody levels, providing support for FcRn's direct role in IgG IC-associated inflammation. Thus, CD32a and FcRn coregulate IgG IC-mediated immunity in a manner favoring the CD32aH variant, providing a novel mechanism for its disease association.
Collapse
Affiliation(s)
- Jonathan J. Hubbard
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Michal Pyzik
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Timo Rath
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Lisa K. Kozicky
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Kine M.K. Sand
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Amit K. Gandhi
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Algirdas Grevys
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Stian Foss
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Susan C. Menzies
- Division of Gastroenterology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan N. Glickman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Edda Fiebiger
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | | | - Inger Sandlie
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan Terje Andersen
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Laura M. Sly
- Division of Gastroenterology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kristi Baker
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Richard S. Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Harvard Digestive Diseases Center, Boston, MA
| |
Collapse
|
42
|
Ruano-Salguero JS, Lee KH. Antibody transcytosis across brain endothelial-like cells occurs nonspecifically and independent of FcRn. Sci Rep 2020; 10:3685. [PMID: 32111886 PMCID: PMC7048754 DOI: 10.1038/s41598-020-60438-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/04/2020] [Indexed: 11/09/2022] Open
Abstract
The blood-brain barrier (BBB) hinders the brain delivery of therapeutic immunoglobulin γ (IgG) antibodies. Evidence suggests that IgG-specific processing occurs within the endothelium of the BBB, but any influence on transcytosis remains unclear. Here, involvement of the neonatal Fc receptor (FcRn), which mediates IgG recycling and transcytosis in peripheral endothelium, was investigated by evaluating the transcytosis of IgGs with native or reduced FcRn engagement across human induced pluripotent stem cell-derived brain endothelial-like cells. Despite differential trafficking, the permeability of all tested IgGs were comparable and remained constant irrespective of concentration or competition with excess IgG, suggesting IgG transcytosis occurs nonspecifically and originates from fluid-phase endocytosis. Comparison with the receptor-enhanced permeability of transferrin indicates that the phenomena observed for IgG is ubiquitous for most macromolecules. However, increased permeability was observed for macromolecules with biophysical properties known to engage alternative endocytosis mechanisms, highlighting the importance of biophysical characterizations in assessing transcytosis mechanisms.
Collapse
Affiliation(s)
- John S Ruano-Salguero
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, 19711, USA
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, 19716, USA.
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, 19711, USA.
| |
Collapse
|
43
|
Assessment of Structural and Functional Comparability of Biosimilar Products: Trastuzumab as a Case Study. BioDrugs 2020; 34:209-223. [DOI: 10.1007/s40259-020-00404-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
44
|
Huang H, Fang L, Xue L, Zhang T, Kim K, Hou S, Zheng F, Zhan CG. PEGylation but Not Fc-Fusion Improves in Vivo Residence Time of a Thermostable Mutant of Bacterial Cocaine Esterase. Bioconjug Chem 2019; 30:3021-3027. [PMID: 31661952 DOI: 10.1021/acs.bioconjchem.9b00622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
It is very popular to fuse a protein drug or drug candidate to the Fc domain of immunoglobulin G (IgG) in order to prolong the in vivo half-life. In this study, we have designed, prepared, and tested an Fc-fused thermostable cocaine esterase (CocE) mutant (known as E196-301, with the T172R/G173Q/L196C/I301C substitutions on CocE) expressed in E. coli. As expected, Fc-fusion does not affect the in vitro enzyme activity and thermal stability of the enzyme and that Fc-E196-301 can favorably bind FcRn with Kd = 386 ± 35 nM. However, Fc-fusion does not prolong the in vivo half-life of E196-301 at all; Fc-E196-301 and E196-301 have essentially the same PK profile (t1/2 = 0.4 ± 0.1 h) in rats. This is the first time demonstrating that Fc-fusion does not prolong in vivo half-life of a protein. This finding is consistent with the mechanistic understanding that E196-301 and Fc-E196-301 are all degraded primarily through rapid proteolysis in the body. The Fc fusion cannot protect E196-301 from the proteolysis in the body. Nevertheless, it has been demonstrated that PEGylation can effectively protect E196-301, as the PEGylated E196-301, i.e., PEG-E196-301, has a significantly prolonged in vivo half-life. It has also been demonstrated that both E196-301 and PEG-E196-301 have dose-dependent in vivo half-lives (e.g., 19.9 ± 6.4 h for the elimination t1/2 of 30 mg/kg PEG-E196-301), as the endogenous proteolytic enzymes responsible for proteolysis of E196-301 (PEGylated or not) are nearly saturated by the high plasma concentration produced by a high dose of E196-301 or PEG-E196-301.
Collapse
Affiliation(s)
- Haifeng Huang
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States.,Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Lei Fang
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Liu Xue
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Ting Zhang
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States.,Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Kyungbo Kim
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States.,Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Shurong Hou
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States.,Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Fang Zheng
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States.,Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States.,Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| |
Collapse
|
45
|
Yuan D, Rode F, Cao Y. A systems pharmacokinetic/pharmacodynamic model for concizumab to explore the potential of anti-TFPI recycling antibodies. Eur J Pharm Sci 2019; 138:105032. [PMID: 31394258 PMCID: PMC6824202 DOI: 10.1016/j.ejps.2019.105032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 07/22/2019] [Accepted: 08/01/2019] [Indexed: 01/31/2023]
Abstract
Concizumab is a humanized monoclonal antibody in clinical investigation directed against membrane-bound and soluble tissue factor pathway inhibitor (mTFPI and sTFPI) for treatment of hemophilia. Concizumab displays a non-linear pharmacokinetic (PK) profile due to mTFPI-mediated endocytosis and necessitates a high dose and frequent dosing to suppress the abundant sTFPI, a negative regulator of coagulation. Recycling antibodies that can dissociate bound mTFPI/sTFPI in endosomes for degradation and rescue antibody from degradation have a potential in reducing the dose by extending antibody systemic persistence and sTFPI suppression. We developed a systems PK/pharmacodynamics (PD) model with nested endosome compartments to simulate the effect of decreased antibody binding to mTFPI/sTFPI in endosomes on antibody clearance and sTFPI suppression for exploring the potential of anti-TFPI recycling antibodies in reducing the dose. A dynamic model-building strategy was taken. A reduced PK/PD model without the endosome compartments was developed to optimize unknown target turnover parameters using concizumab PK data. The optimized parameters were then employed in the systems PK/PD model for simulations. The obtained systems PK/PD model adequately described the PK of concizumab in rabbits, monkeys, and humans and the PD in humans. The systems PK/PD model predicted that an anti-TFPI recycling antibody with a 100-fold higher mTFPI/sTFPI dissociation constant in endosomes than concizumab can extend sTFPI suppression from 12 days to 1 month. Thus, the systems PK/PD model provides a quantitative platform for guiding the engineering and translational development of anti-TFPI recycling antibodies.
Collapse
Affiliation(s)
- Dongfen Yuan
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frederik Rode
- Novo Nordisk, Translational DMPK, H. Lundbeck A/S, Denmark
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
46
|
Pyzik M, Sand KMK, Hubbard JJ, Andersen JT, Sandlie I, Blumberg RS. The Neonatal Fc Receptor (FcRn): A Misnomer? Front Immunol 2019; 10:1540. [PMID: 31354709 PMCID: PMC6636548 DOI: 10.3389/fimmu.2019.01540] [Citation(s) in RCA: 292] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Antibodies are essential components of an adaptive immune response. Immunoglobulin G (IgG) is the most common type of antibody found in circulation and extracellular fluids. Although IgG alone can directly protect the body from infection through the activities of its antigen binding region, the majority of IgG immune functions are mediated via proteins and receptors expressed by specialized cell subsets that bind to the fragment crystallizable (Fc) region of IgG. Fc gamma (γ) receptors (FcγR) belong to a broad family of proteins that presently include classical membrane-bound surface receptors as well as atypical intracellular receptors and cytoplasmic glycoproteins. Among the atypical FcγRs, the neonatal Fc receptor (FcRn) has increasingly gained notoriety given its intimate influence on IgG biology and its ability to also bind to albumin. FcRn functions as a recycling or transcytosis receptor that is responsible for maintaining IgG and albumin in the circulation, and bidirectionally transporting these two ligands across polarized cellular barriers. More recently, it has been appreciated that FcRn acts as an immune receptor by interacting with and facilitating antigen presentation of peptides derived from IgG immune complexes (IC). Here we review FcRn biology and focus on newer advances including how emerging FcRn-targeted therapies may affect the immune responses to IgG and IgG IC.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Kine M K Sand
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jonathan J Hubbard
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Harvard Digestive Diseases Center, Boston, MA, United States
| |
Collapse
|
47
|
Hirasawa S, Kitahara Y, Okamatsu Y, Fujii T, Nakayama A, Ueno S, Ijichi C, Futaki F, Nakata K, Taki M. Facile and Efficient Chemoenzymatic Semisynthesis of Fc-Fusion Compounds for Half-Life Extension of Pharmaceutical Components. Bioconjug Chem 2019; 30:2323-2331. [PMID: 31038930 DOI: 10.1021/acs.bioconjchem.9b00235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The formation of Fc-fusions, in which biologically active molecules and the Fc fragment of antibodies are linked to each other, is one of the most efficient and successful half-life extension technologies to be developed and applied to peptide and protein pharmaceuticals thus far. Fc-fusion compounds are generally produced by recombinant methods. However, these cannot be applied to artificial middle molecules, such as peptides with non-natural amino acids, unnatural cyclic peptides, or pharmaceutical oligonucleotides. Here, we developed a simple, efficient, semisynthetic method for Fc-fusion production involving our previously developed enzymatic N-terminal extension reaction (i.e., NEXT-A reaction) and strain-promoted azide-alkyne cycloaddition, achieving quantitative conversion and high selectivity for the N-terminus of the Fc protein. An Fc-fusion compound prepared by this method showed comparable biological activity to that of the original peptide and a long-circulating plasma half-life. Thus, the proposed method is potentially applicable for the conjugation of a wide range of pharmaceutical components.
Collapse
Affiliation(s)
- Shigeo Hirasawa
- Department of Engineering Science, Graduate School of Informatics and Engineering , The University of Electro-Communications (UEC) , 1-5-1 Chofugaoka , Chofu , Tokyo 182-8585 , Japan
| | | | | | | | | | | | | | | | | | - Masumi Taki
- Department of Engineering Science, Graduate School of Informatics and Engineering , The University of Electro-Communications (UEC) , 1-5-1 Chofugaoka , Chofu , Tokyo 182-8585 , Japan
| |
Collapse
|
48
|
Vaniotis G, Moffett S, Sulea T, Wang N, Elahi SM, Lessard E, Baardsnes J, Perrino S, Durocher Y, Frystyk J, Massie B, Brodt P. Enhanced anti-metastatic bioactivity of an IGF-TRAP re-engineered to improve physicochemical properties. Sci Rep 2018; 8:17361. [PMID: 30478273 PMCID: PMC6255772 DOI: 10.1038/s41598-018-35407-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/07/2018] [Indexed: 01/22/2023] Open
Abstract
The insulin-like growth factor (IGF) axis has been implicated in the progression of malignant disease and identified as a clinically important therapeutic target. Several IGF-1 receptor (IGF-1R) targeting drugs including humanized monoclonal antibodies have advanced to phase II/III clinical trials, but to date, have not progressed to clinical use, due, at least in part, to interference with insulin receptor signalling. We previously reported on the production of a soluble fusion protein consisting of the extracellular domain of human IGF-1R fused to the Fc portion of human IgG1 (first generation IGF-TRAP) that bound human IGF-1 and IGF-2 with a 3 log higher affinity than insulin. We showed that the IGF-TRAP had potent anti-cancer activity in several pre-clinical models of aggressive carcinomas. Here we report on the re-engineering of the IGF-TRAP with the aim of improving physicochemical properties and suitability for clinical applications. We show that cysteine-serine substitutions in the Fc hinge region of IGF-TRAP eliminated high-molecular-weight oligomerized species, while a further addition of a flexible linker, not only improved the pharmacokinetic profile, but also enhanced the therapeutic profile of the IGF-TRAP, as evaluated in an experimental colon carcinoma metastasis model. Dose-response profiles of the modified IGF-TRAPs correlated with their bio-availability profiles, as measured by the IGF kinase-receptor-activation (KIRA) assay, providing a novel, surrogate biomarker for drug efficacy. This study provides a compelling example of structure-based re-engineering of Fc-fusion-based biologics for better manufacturability that also significantly improved pharmacological parameters. It identifies the re-engineered IGF-TRAP as a potent anti-cancer therapeutic.
Collapse
Affiliation(s)
- George Vaniotis
- Department of Surgery, McGill University, Montreal Quebec, Canada
| | - Serge Moffett
- Department of Surgery, McGill University, Montreal Quebec, Canada
| | - Traian Sulea
- Institute of Parasitology, McGill University, Montreal Quebec, Canada
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Ni Wang
- Department of Surgery, McGill University, Montreal Quebec, Canada
| | - S Mehdy Elahi
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Etienne Lessard
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | | | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Jan Frystyk
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Bernard Massie
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Pnina Brodt
- Department of Surgery, McGill University, Montreal Quebec, Canada.
- Department of Medicine, McGill University, Montreal Quebec, Canada.
- Department of Oncology, McGill University, Montreal Quebec, Canada.
- Cancer Research Program, Research Institute of the McGill University Health Center, Montreal Quebec, Canada.
| |
Collapse
|
49
|
Improvement of pharmacokinetic properties of therapeutic antibodies by antibody engineering. Drug Metab Pharmacokinet 2018; 34:25-41. [PMID: 30472066 DOI: 10.1016/j.dmpk.2018.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/13/2018] [Accepted: 10/23/2018] [Indexed: 01/17/2023]
Abstract
Monoclonal antibodies (mAbs) have become an important therapeutic option for several diseases. Since several mAbs have shown promising efficacy in clinic, the competition to develop mAbs has become severe. In efforts to gain a competitive advantage over other mAbs and provide significant benefits to patients, innovations in antibody engineering have aimed at improving the pharmacokinetic properties of mAbs. Because engineering can provide therapeutics that are more convenient, safer, and more efficacious for patients in several disease areas, it is an attractive approach to provide significant benefits to patients. Further advances in engineering mAbs to modulate their pharmacokinetics were driven by the increase of total soluble target antigen concentration that is often observed after injecting a mAb, which then requires a high dosage to antagonize. To decrease the required dosage, several antibody engineering techniques have been invented that reduce the total concentration of soluble target antigen. Here, we review the various ways that antibody engineering can improve the pharmacokinetic properties of mAbs.
Collapse
|
50
|
Smith B, Kiessling A, Lledo-Garcia R, Dixon KL, Christodoulou L, Catley MC, Atherfold P, D'Hooghe LE, Finney H, Greenslade K, Hailu H, Kevorkian L, Lightwood D, Meier C, Munro R, Qureshi O, Sarkar K, Shaw SP, Tewari R, Turner A, Tyson K, West S, Shaw S, Brennan FR. Generation and characterization of a high affinity anti-human FcRn antibody, rozanolixizumab, and the effects of different molecular formats on the reduction of plasma IgG concentration. MAbs 2018; 10:1111-1130. [PMID: 30130439 PMCID: PMC6291300 DOI: 10.1080/19420862.2018.1505464] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Rozanolixizumab (UCB7665), a humanized high-affinity anti-human neonatal Fc receptor (FcRn) monoclonal antibody (IgG4P), has been developed to reduce pathogenic IgG in autoimmune and alloimmune diseases. We document the antibody isolation and compare rozanolixizumab with the same variable region expressed in various mono-, bi- and trivalent formats. We report activity data for rozanolixizumab and the different molecular formats in human cells, FcRn-transgenic mice, and cynomolgus monkeys. Rozanolixizumab, considered the most effective molecular format, dose-dependently and selectively reduced plasma IgG concentrations in an FcRn-transgenic mouse model (no effect on albumin). Intravenous (IV) rozanolixizumab dosing in cynomolgus monkeys demonstrated non-linear pharmacokinetics indicative of target-mediated drug disposition; single IV rozanolixizumab doses (30 mg/kg) in cynomolgus monkeys reduced plasma IgG concentration by 69% by Day 7 post-administration. Daily IV administration of rozanolixizumab (initial 30 mg/kg loading dose; 5 mg/kg daily thereafter) reduced plasma IgG concentrations in all cynomolgus monkeys, with low concentrations maintained throughout the treatment period (42 days). In a 13-week toxicology study in cynomolgus monkeys, supra-pharmacological subcutaneous and IV doses of rozanolixizumab (≤ 150 mg/kg every 3 days) were well tolerated, inducing sustained (but reversible) reductions in IgG concentrations by up to 85%, with no adverse events observed. We have demonstrated accelerated natural catabolism of IgG through inhibition of IgG:FcRn interactions in mice and cynomolgus monkeys. Inhibition of FcRn with rozanolixizumab may provide a novel therapeutic approach to reduce pathogenic IgG in human autoimmune disease. Rozanolixizumab is being investigated in patients with immune thrombocytopenia (NCT02718716) and myasthenia gravis (NCT03052751).
Collapse
|