1
|
Sardag I, Duvenci ZS, Belkaya S, Timucin E. Rational design of monomeric IL37 variants guided by stability and dynamical analyses of IL37 dimers. Comput Struct Biotechnol J 2024; 23:1854-1863. [PMID: 38882680 PMCID: PMC11177541 DOI: 10.1016/j.csbj.2024.04.037] [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: 02/20/2024] [Revised: 04/07/2024] [Accepted: 04/14/2024] [Indexed: 06/18/2024] Open
Abstract
IL37 plays important roles in the regulation of innate immunity and its oligomeric status is critical to these roles. In its monomeric state, IL37 can effectively inhibit the inflammatory response of IL18 by binding to IL18Rα, a capacity lost in its dimeric form, underlining the pivotal role of the oligomeric status of IL37 in its anti-inflammatory action. Until now, two IL37 dimer structures have been deposited in PDB, reflecting a substantial difference in their dimer interfaces. Given this discrepancy, we analyzed the PDB structures of the IL37 dimer (PDB IDs: 6ncu, 5hn1) along with a AF2-multimer prediction by molecular dynamics (MD) simulations. Results showed that the 5hn1 and AF2-predicted dimers have the same interface and stably maintained their conformations throughout simulations, while the recent IL37 dimer (PDB ID: 6ncu) with a different interface did not, proposing a possible issue with the recent IL37 dimer structure (6ncu). Next, focusing on the stable dimer structures, we have identified five critical positions of V71/Y85/I86/E89/S114, three new positions compared to the literature, that would reduce dimer stability without affecting the monomer structure. Two quintuple mutants were tested by MD simulations and showed partial or complete dissociation of the dimer. Overall, the insights gained from this study reinforce the validity of the 5hn1 and AF2 multimer structures, while also advancing our understanding of the IL37 dimer interface through the generation of monomer-locked IL37 variants.
Collapse
Affiliation(s)
- Inci Sardag
- Bogazici University, Department of Molecular Biology and Genetics, Istanbul 34342, Turkey
| | - Zeynep Sevval Duvenci
- Acibadem Mehmet Ali Aydinlar University, Institute of Health Sciences, Department of Biostatistics and Bioinformatics, Istanbul 34752, Turkey
| | - Serkan Belkaya
- Bilkent University, Department of Molecular Biology and Genetics, Ankara 06800, Turkey
- Bilkent University, The National Nanotechnology Research Center (UNAM), Ankara 06800, Turkey
| | - Emel Timucin
- Acibadem Mehmet Ali Aydinlar University, Institute of Health Sciences, Department of Biostatistics and Bioinformatics, Istanbul 34752, Turkey
- Acibadem Mehmet Ali Aydinlar University, School of Medicine, Biostatistics and Medical Informatics, Istanbul 34752, Turkey
| |
Collapse
|
2
|
Choi SM, Lee JH, Ko S, Hong SS, Jin HE. Mechanism of Action and Pharmacokinetics of Approved Bispecific Antibodies. Biomol Ther (Seoul) 2024; 32:708-722. [PMID: 39448393 PMCID: PMC11535297 DOI: 10.4062/biomolther.2024.146] [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: 08/21/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Bispecific antibodies represent a significant advancement in therapeutic antibody engineering, offering the ability to simultaneously target two distinct antigens. This dual-targeting capability enhances therapeutic efficacy, especially in complex diseases, such as cancer and autoimmune disorders, where drug resistance and incomplete target coverage are prevalent challenges. Bispecific antibodies facilitate immune cell engagement and disrupt multiple signaling pathways, providing a more comprehensive treatment approach than traditional monoclonal antibodies. However, the intricate structure of bispecific antibodies introduces unique pharmacokinetic challenges, including issues related to their absorption, distribution, metabolism, and excretion, which can significantly affect their efficacy and safety. This review provides an in-depth analysis of the structural design, mechanisms of action, and pharmacokinetics of the currently approved bispecific antibodies. It also highlights the engineering innovations that have been implemented to overcome these challenges, such as Fc modifications and advanced dimerization techniques, which enhance the stability and half-life of bispecific antibodies. Significant progress has been made in bispecific antibody technology; however, further research is necessary to broaden their clinical applications, enhance their safety profiles, and optimize their incorporation into combination therapies. Continuous advancements in this field are expected to enable bispecific antibodies to provide more precise and effective therapeutic strategies for a range of complex diseases, ultimately improving patient outcomes and advancing precision medicine.
Collapse
Affiliation(s)
- Seong Min Choi
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Soyeon Ko
- Department of Biomedical Sciences, College of Medicine & Program in Biomedicals Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine & Program in Biomedicals Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Hyo-Eon Jin
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
3
|
Jiang J, Xu L, Chai L, Guan X, Zhang L, Liu H, Yan Y, Li L, Zhao Y, Bai X, Tian L, Jia Y. Clinical pharmacokinetics and pharmacodynamics of ongericimab: A potential long-acting PCSK9 monoclonal antibody in healthy subjects and patients with hypercholesterolemia: Randomized, double-blind, placebo-controlled phase Ia and Ib/II studies. Clin Transl Sci 2024; 17:e70061. [PMID: 39498965 PMCID: PMC11536336 DOI: 10.1111/cts.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 11/07/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) increases plasma low-density lipoprotein-cholesterol (LDL-C) by decreasing the expression of the LDL-receptor on hepatic cells. Ongericimab (JS002) is a novel PCSK9 monoclonal antibody that exhibits a long-acting LDL-C lowering effect by exclusively inhibiting PCSK9 in pre-clinical studies. Two randomized, double-blind, placebo-controlled trials were conducted to evaluate the safety, tolerability, efficacy, immunogenicity, pharmacokinetic, and pharmacodynamic profiles of ongericimab in healthy subjects and patients with hypercholesterolemia. Eighty-four healthy subjects in the phase Ia study received a single dose of placebo or ongericimab (15-450 mg). Ninety patients with hypercholesterolemia in the phase Ib/II study received placebo or ongericimab 150 mg Q2W, 300 mg Q4W, or 450 mg Q4W for 12 weeks. Ongericimab exhibited non-linear kinetics. The apparent clearance decreased as the dosage increased, with terminal elimination half-life (t1/2) values of 4.5-6.5 days. Overall, ongericimab was well tolerated in both studies. A single dose of ongericimab reduced LDL-C levels by 30%-73% in healthy subjects, and repeated doses of ongericimab reduced LDL-C levels by 67%-80% in patients with hypercholesterolemia. At the end of the dosing interval in the phase Ib/II study, over 70% of patients' LDL-C levels decreased by more than 50% from baseline. The results showed that ongericimab had a significant long-acting LDL-C lowering effect with good safety and potential for clinical application.
Collapse
Affiliation(s)
- Juanjuan Jiang
- NHC Key Laboratory of Clinical Research for Cardiovascular Medications, National Clinical Research Center of Cardiovascular DiseasesFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Li Xu
- NHC Key Laboratory of Clinical Research for Cardiovascular Medications, National Clinical Research Center of Cardiovascular DiseasesFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lin Chai
- NHC Key Laboratory of Clinical Research for Cardiovascular Medications, National Clinical Research Center of Cardiovascular DiseasesFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoyuan Guan
- NHC Key Laboratory of Clinical Research for Cardiovascular Medications, National Clinical Research Center of Cardiovascular DiseasesFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Li Zhang
- NHC Key Laboratory of Clinical Research for Cardiovascular Medications, National Clinical Research Center of Cardiovascular DiseasesFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hong Liu
- NHC Key Laboratory of Clinical Research for Cardiovascular Medications, National Clinical Research Center of Cardiovascular DiseasesFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yan Yan
- NHC Key Laboratory of Clinical Research for Cardiovascular Medications, National Clinical Research Center of Cardiovascular DiseasesFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lili Li
- Shanghai Junshi BiosciencesShanghaiChina
| | - Yi Zhao
- Shanghai Junshi BiosciencesShanghaiChina
| | | | - Lei Tian
- NHC Key Laboratory of Clinical Research for Cardiovascular Medications, National Clinical Research Center of Cardiovascular DiseasesFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Youhong Jia
- Department of CardiologyFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
4
|
Mekala JR, Nalluri HP, Reddy PN, S B S, N S SK, G V S D SK, Dhiman R, Chamarthy S, Komaragiri RR, Manyam RR, Dirisala VR. Emerging trends and therapeutic applications of monoclonal antibodies. Gene 2024; 925:148607. [PMID: 38797505 DOI: 10.1016/j.gene.2024.148607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Monoclonal antibodies (mAbs) are being used to prevent, detect, and treat a broad spectrum of malignancies and infectious and autoimmune diseases. Over the past few years, the market for mAbs has grown exponentially. They have become a significant part of many pharmaceutical product lines, and more than 250 therapeutic mAbs are undergoing clinical trials. Ever since the advent of hybridoma technology, antibody-based therapeutics were realized using murine antibodies which further progressed into humanized and fully human antibodies, reducing the risk of immunogenicity. Some of the benefits of using mAbs over conventional drugs include a drastic reduction in the chances of adverse reactions, interactions between drugs, and targeting specific proteins. While antibodies are very efficient, their higher production costs impede the process of commercialization. However, their cost factor has been improved by developing biosimilar antibodies, which are affordable versions of therapeutic antibodies. Along with biosimilars, innovations in antibody engineering have helped to design bio-better antibodies with improved efficacy than the conventional ones. These novel mAb-based therapeutics are set to revolutionize existing drug therapies targeting a wide spectrum of diseases, thereby meeting several unmet medical needs. In the future, mAbs generated by applying next-generation sequencing (NGS) are expected to become a powerful tool in clinical therapeutics. This article describes the methods of mAb production, pre-clinical and clinical development of mAbs, approved indications targeted by mAbs, and novel developments in the field of mAb research.
Collapse
Affiliation(s)
- Janaki Ramaiah Mekala
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA.
| | - Hari P Nalluri
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Prakash Narayana Reddy
- Department of Microbiology, Dr. V.S. Krishna Government College, Visakhapatnam 530013, India
| | - Sainath S B
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524320, AP, India
| | - Sampath Kumar N S
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Sai Kiran G V S D
- Santhiram Medical College and General Hospital, Nandyal, Kurnool 518501, AP, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Sciences, National Institute of Technology Rourkela-769008, India
| | - Sahiti Chamarthy
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA
| | - Raghava Rao Komaragiri
- Department of CSE, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522302, Andhra Pradesh, INDIA
| | - Rajasekhar Reddy Manyam
- Amrita School of Computing, Amrita Vishwa Vidyapeetham, Amaravati Campus, Amaravati, Andhra Pradesh, India
| | - Vijaya R Dirisala
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India.
| |
Collapse
|
5
|
An S, Zheng M, Park IG, Park SG, Noh M, Sung JH. Humanized CXCL12 antibody delays onset and modulates immune response in alopecia areata mice: insights from single-cell RNA sequencing. Front Immunol 2024; 15:1444777. [PMID: 39483478 PMCID: PMC11524852 DOI: 10.3389/fimmu.2024.1444777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
It has been demonstrated that CXCL12 inhibits hair growth via CXCR4, and its neutralizing antibody (Ab) increases hair growth in alopecia areata (AA). However, the molecular mechanisms have not been fully elucidated. In the present study, we further prepared humanized CXCL12 Ab for AA treatment and investigated underlying molecular mechanisms using single-cell RNA sequencing. Subcutaneous injection of humanized CXCL12 Ab significantly delayed AA onset in mice, and dorsal skin was analyzed. T cells and dendritic cells/macrophages were increased in the AA model, but decreased after CXCL12 Ab treatment. Pseudobulk RNA sequencing identified 153 differentially expressed genes that were upregulated in AA model and downregulated after Ab treatment. Gene ontology analysis revealed that immune cell chemotaxis and cellular response to type II interferon were upregulated in AA model but downregulated after Ab treatment. We further identified key immune cell-related genes such as Ifng, Cd8a, Ccr5, Ccl4, Ccl5, and Il21r, which were colocalized with Cxcr4 in T cells and regulated by CXCL12 Ab treatment. Notably, CD8+ T cells were significantly increased and activated via Jak/Stat pathway in the AA model but inactivated after CXCL12 Ab treatment. Collectively, these results indicate that humanized CXCL12 Ab is promising for AA treatment via immune modulatory effects.
Collapse
MESH Headings
- Animals
- Alopecia Areata/immunology
- Alopecia Areata/genetics
- Alopecia Areata/drug therapy
- Mice
- Chemokine CXCL12/genetics
- Single-Cell Analysis
- Disease Models, Animal
- Humans
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Sequence Analysis, RNA
- Receptors, CXCR4/genetics
- Receptors, CXCR4/immunology
- Receptors, CXCR4/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/drug effects
- Female
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Seungchan An
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Mei Zheng
- Epi Biotech Co., Ltd., R&D Center, Incheon, Republic of Korea
| | - In Guk Park
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Sang Gyu Park
- College of Pharmacy, Ajou University, Suwon, Republic of Korea
| | - Minsoo Noh
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Jong-Hyuk Sung
- Epi Biotech Co., Ltd., R&D Center, Incheon, Republic of Korea
| |
Collapse
|
6
|
Yang Y, Qiu H, Fan Y, Zhang Q, Qin H, Wu J, Zhang X, Liu Y, Zhou R, Zhang Q, Ye Z, Ma J, Xu Y, Feng S, Fei Y, Li N, Cui X, Dong F, Wang Q, Shen K, Shakib S, Williams J, Hu W. Safety, tolerability, pharmacokinetics and pharmacodynamics of a single intravenous dose of SHR-1707 in healthy adult subjects: two randomized, double-blind, single-ascending-dose, phase 1 studies. Alzheimers Res Ther 2024; 16:218. [PMID: 39390616 PMCID: PMC11465679 DOI: 10.1186/s13195-024-01584-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 09/29/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND SHR-1707 is a novel humanized anti-Aβ IgG1 monoclonal antibody that binds to Aβ fibrils and monomers to block the formation of Aβ plaques or to promote the microglial phagocytosis of Aβ. Preclinical studies showed that SHR-1707 reduced brain Aβ deposition in 5xFAD transgenic mice. Herein, we conducted two phase 1 studies to evaluate the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of a single intravenous dose of SHR-1707 in healthy adult subjects. METHODS Two randomized, double-blind, single-ascending-dose, phase 1 studies were conducted in China (Study CHN) and Australia (Study AUS). Study CHN consisted of 2 parts. In Part 1, eligible healthy young adults (18-45 years) were sequentially randomized 8:2 to receive SHR-1707 (five cohorts: 2, 6, 20, 40, and 60 mg/kg) or placebo in each cohort; in Part 2, elderly subjects (55-80 years) were randomized 8:4 to receive SHR-1707 (20 mg/kg) or placebo. A similar design was used in Study AUS, but with only healthy young adults enrolled across three dosing cohorts (2, 20, and 60 mg/kg). RESULTS Sixty-two (part 1/2, n = 50/12; age range, 18-42/55-63 years) and 30 subjects (age range, 18-42 years) received SHR-1707 or placebo in Study CHN and Study AUS, respectively. In Study CHN, all treatment-related adverse events (TRAEs) were mild, with the most common being transient laboratory abnormalities. In Study AUS, TRAEs were mostly mild (1 moderate event each with SHR-1707/placebo); the most common TRAEs with SHR-1707 were dysgeusia and fatigue (8.3% each). In both studies, the exposure of SHR-1707 increased in a slightly greater than dose-proportional manner over the dose range of 2-60 mg/kg in young adults; there was a dose-dependent increase in plasma Aβ42 concentration following SHR-1707 administration compared with the placebo group. The safety and PK and PD profiles of SHR-1707 in the elderly subjects were consistent with the younger counterpart at the same dose level. No ethnic difference in safety, PK and PD of SHR-1707 was observed. CONCLUSIONS A single intravenous dose of SHR-1707 at 2-60 mg/kg was safe and well tolerated in healthy young adult and elderly subjects. The PK and PD profiles are supportive for further clinical development. TRIAL REGISTRATION NCT04973189 (retrospectively registered on Jul.21, 2021) and NCT04745104 (registered on Feb.6, 2021) on clinicaltrials.gov.
Collapse
Affiliation(s)
- Yaru Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongyan Qiu
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Yuru Fan
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qin Zhang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huiling Qin
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Juan Wu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuan Zhang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yueyue Liu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Renpeng Zhou
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qian Zhang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zi Ye
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Jingyue Ma
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Ye Xu
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Sheng Feng
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Yue Fei
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Na Li
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Xiaojing Cui
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Fangli Dong
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Quanren Wang
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Kai Shen
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd, Shanghai, China
| | - Sepehr Shakib
- CMAX Clinical Research, Adelaide, South Australia.
- Department of Clinical Pharmacology, University of Adelaide, Adelaide, South Australia.
- Department of Clinical Pharmacology, CMAX Clinical Research, University of Adelaide, Adelaide, South Australia, Australia.
| | | | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
7
|
Bryniarski MA, Tuhin MTH, Shomin CD, Nasrollahi F, Ko EC, Soto M, Chung K, Poon-Andersen C, Primack R, Wong D, Ojeda E, Chung J, Cook KD, Conner KP. Utility of Cellular Measurements of Non-Specific Endocytosis to Assess the Target-Independent Clearance of Monoclonal Antibodies. J Pharm Sci 2024; 113:3100-3111. [PMID: 39009346 DOI: 10.1016/j.xphs.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Past studies have demonstrated higher clearance for monoclonal antibodies possessing increased rates of non-specific endocytosis. However, this metric is oftentimes evaluated indirectly using biophysical techniques or cell surface binding studies that may not provide insight into the specific rates of cellular turnover. Furthermore, few examples evaluating non-specific endocytosis have been reported for a therapeutic antibody that reached clinical assessment. In the current report, we evaluated a therapeutic human immunoglobulin G2 monoclonal antibody targeted against the interleukin-4 receptor alpha chain (IL-4Rα) that exhibited elevated target independent clearance in previous Phase 1 and 2 studies. We confirmed high non-specific clearance of the anti-IL-4Rα antibody as compared to a reference antibody during pharmacokinetic assessments in wild type mice where target-mediated disposition was absent. We then developed a cell-based method capable of measuring cellular protein endocytosis and demonstrated the anti-IL-4Rα antibody exhibited marked non-specific uptake relative to the reference compound. Antibody homology modeling identified the anti-IL-4Rα antibody possessed positive charge patches whose removal via targeted mutations substantially reduced its non-specific endocytosis. We then expanded the scope of the study by evaluating panels of both preclinical and clinically relevant monoclonal antibodies and demonstrate those with the highest rates of non-specific uptake in vitro exhibited elevated target independent clearance, low subcutaneous bioavailability, or both. Our results support the observation that high non-specific endocytosis is a negative attribute in monoclonal antibody development and demonstrate the utility of a generic cell-based screen as a quantitative tool to measure non-specific endocytosis of protein therapeutics at the single-cell level.
Collapse
Affiliation(s)
- Mark A Bryniarski
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA.
| | - Md Tariqul Haque Tuhin
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Carolyn D Shomin
- Department of Biologics, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Fatemeh Nasrollahi
- Process Development; Pre-Pivotal Drug Product Technologies, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Eunkyung Clare Ko
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Marcus Soto
- Pharmacokinetics & Drug Metabolism, Amgen Research, 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Kyu Chung
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Carrie Poon-Andersen
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Ronya Primack
- Pharmacokinetics & Drug Metabolism, Amgen Research, 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Diana Wong
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Esperanza Ojeda
- Pharmacokinetics & Drug Metabolism, Amgen Research, 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - John Chung
- Process Development; Pre-Pivotal Drug Product Technologies, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Kevin D Cook
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA
| | - Kip P Conner
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 750 Gateway Blvd, Suite 100, South San Francisco, CA, 94080, USA.
| |
Collapse
|
8
|
Boonyaratanakornkit J, Wang Q, Nader A, Kimball L, Stevens-Ayers T, Levkova M, Blazevic R, Nguyen J, Wright J, Castor J, Greninger AL, Ford E, Mielcarek M, Fordred S, Han J, Boeckh M, Waghmare A. The Effect of Gastrointestinal Graft-Versus-Host Disease and Diarrhea on the Pharmacokinetic Profile of Sotrovimab in Hematopoietic Stem Cell Transplant Recipients. J Infect Dis 2024; 230:670-679. [PMID: 38743457 DOI: 10.1093/infdis/jiae236] [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: 01/09/2024] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Monoclonal antibodies (mAbs) are utilized broadly to treat cancer and infectious diseases, and mAb exposure (serum concentration over time) is one predictor of overall treatment efficacy. Herein, we present findings from a clinical trial evaluating the pharmacokinetics of the long-acting mAb sotrovimab targeting severe acute respiratory syndrome coronavirus 2 in hematopoietic cell transplant (HCT) recipients. METHODS All participants received an intravenous infusion of sotrovimab within 1 week prior to initiating the pretransplant preparative regimen. The serum concentration of sotrovimab was measured longitudinally for up to 24 weeks posttransplant. RESULTS Compared to non-HCT participants, we found that mAb clearance was 10% and 26% higher in autologous and allogeneic HCT recipients, respectively. Overall sotrovimab exposure was approximately 15% lower in HCT recipients compared to non-HCT recipients. Exposure was significantly reduced in HCT recipients who developed diarrhea and lower gastrointestinal graft-versus-host disease (GVHD) posttransplant. CONCLUSIONS These data show that sotrovimab exposure may be reduced in HCT recipients, possibly related to increased gastrointestinal clearance in patients with GVHD. This phenomenon has implications for dose selection and duration of efficacy with sotrovimab and potentially other mAbs in this vulnerable patient population. Thus, mAb dose regimens developed in non-HCT populations may have to be optimized when applied to HCT populations.
Collapse
Affiliation(s)
- Jim Boonyaratanakornkit
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
- Department of Medicine, University of Washington, Seattle
| | | | | | - Louise Kimball
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
| | | | - Marta Levkova
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
| | - Rachel Blazevic
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
| | - Jeanette Nguyen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
| | - Jennifer Wright
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
| | - Jared Castor
- Department of Laboratory Medicine and Pathology, University of Washington
| | - Alexander L Greninger
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
- Department of Laboratory Medicine and Pathology, University of Washington
| | - Emily Ford
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
- Department of Medicine, University of Washington, Seattle
| | - Marco Mielcarek
- Department of Medicine, University of Washington, Seattle
- Clinical Research Division, Fred Hutchinson Cancer Center
| | | | | | - Michael Boeckh
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
- Department of Medicine, University of Washington, Seattle
| | - Alpana Waghmare
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center
- Infectious Diseases and Virology, Seattle Children's Hospital
- Department of Pediatrics, University of Washington, Seattle
| |
Collapse
|
9
|
Fang P, You M, Cao Y, Feng Q, Shi L, Wang J, Sun X, Yu D, Zhou W, Yin L, Mei F, Zhu X, Cheng A, Tan X. Development and validation of bioanalytical assays for the quantification of 9MW2821, a nectin-4-targeting antibody-drug conjugate. J Pharm Biomed Anal 2024; 248:116318. [PMID: 38908237 DOI: 10.1016/j.jpba.2024.116318] [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: 02/22/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
We designed and developed 9MW2821, an anti-Nectin-4 antibody-drug conjugate (ADC) with an enzymatically cleavable valine-citrulline linker and monomethyl auristatin E (MMAE) as the payload. Four bioanalytical assays for total antibodies, conjugated antibodies, conjugated payload, and free payload were then developed and validated for the comprehensive evaluation of the multiple drug forms of 9MW2821. Specific sandwich enzyme-linked immunosorbent assays were used to quantify total antibodies and conjugated antibody, showing good drug-to-antibody ratio (DAR) tolerance. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to determine free MMAE, and conjugated MMAE was quantified using a combination of ligand-binding assay (LBA) and LC-MS/MS. Based on these four assays, we studied the serum stability and monkey pharmacokinetic profiles of 9MW2821, and the in vivo DAR of 9MW2821 was calculated and dynamically monitored. In conclusion, we developed and validated series of bioanalytical assays to quantify multiple forms of 9MW2821, a new ADC, and used the assays to evaluate the serum stability and monkey pharmacokinetic characteristics. The results indicate good linker stability and suggest that the developed assays can be further used in clinical settings.
Collapse
Affiliation(s)
- Peng Fang
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Meng You
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Yuxia Cao
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Qingjun Feng
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Lei Shi
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Jin Wang
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Xiaowei Sun
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Dongan Yu
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Wei Zhou
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Long Yin
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Fei Mei
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Xiaohong Zhu
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Aidi Cheng
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Xiaoding Tan
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China.
| |
Collapse
|
10
|
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
|
11
|
Jairam RK, Franz M, Hanke N, Kuepfer L. Physiologically based pharmacokinetic models for systemic disposition of protein therapeutics in rabbits. Front Pharmacol 2024; 15:1427325. [PMID: 39263566 PMCID: PMC11387799 DOI: 10.3389/fphar.2024.1427325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modelling is an important tool to predict drug disposition in the body. Rabbits play a pivotal role as a highly valued small animal model, particularly in the field of ocular therapeutics, where they serve as a crucial link between preclinical research and clinical applications. In this context, we have developed PBPK models designed specifically for rabbits, with a focus on accurately predicting the pharmacokinetic profiles of protein therapeutics following intravenous administration. Our goal was to comprehend the influence of key physiological factors on systemic disposition of antibodies and their functional derivatives. For the development of the systemic PBPK models, rabbit physiological factors such as gene expression, body weight, neonatal fragment crystallizable receptor (FcRn) binding, target binding, target concentrations, and target turnover rate were meticulously considered. Additionally, key protein parameters, encompassing hydrodynamic radius, binding kinetic constants (KD, koff), internal degradation of the protein-target complex, and renal clearance, were represented in the models. Our final rabbit models demonstrated a robust correlation between predicted and observed serum concentration-time profiles after single intravenous administration in rabbits, covering IgG, Fab, F(ab)2, Fc, and Fc fusion proteins from various publications. These pharmacokinetic simulations offer a promising platform for translating preclinical findings to clinical settings. The presented rabbit intravenous PBPK models lay an important foundation for more specific applications of protein therapeutics in ocular drug development.
Collapse
Affiliation(s)
- Ravi Kumar Jairam
- Institute for Systems Medicine with Focus on Organ Interaction, University Hospital RWTH Aachen, Aachen, Germany
| | - Maria Franz
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Nina Hanke
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Lars Kuepfer
- Institute for Systems Medicine with Focus on Organ Interaction, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
12
|
Jin SE, Kim J, Sung JH. Recent approaches of antibody therapeutics in androgenetic alopecia. Front Pharmacol 2024; 15:1434961. [PMID: 39221145 PMCID: PMC11362041 DOI: 10.3389/fphar.2024.1434961] [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: 05/19/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Therapeutic antibodies (Abs) have been anticipated as promising alternatives to conventional treatments such as topical minoxidil and oral finasteride for androgenetic alopecia (AGA). Due to the high molecular weight of typical Abs, the half-life of subcutaneous Abs exceeds 2 weeks, allowing an administration intervals of once a month or longer. Direct injection into the areas of hair loss is also feasible, potentially enhancing treatment efficacy while minimizing systemic side effects. However, therapeutic Abs are rarely developed for AGA therapy due to the requirement to be responsiveness to androgens and to exist in the extracellular fluid or cell surface surrounding the hair follicle. In this review, we introduce recent progress of antibody therapeutics in AGA targeting the prolactin receptor, Interleukin-6 receptor, C-X-C motif chemokine ligand 12, and dickkopf 1. As therapeutic Abs for AGA are still in the early stages, targets need further validation and optimization for clinical application.
Collapse
Affiliation(s)
- Su-Eon Jin
- Epi Biotech Co., Ltd., Incheon, Republic of Korea
| | - Jino Kim
- New Hair Plastic Surgery Clinic, Seoul, Republic of Korea
| | | |
Collapse
|
13
|
Goraltchouk A, Lourie J, Hollander JM, Grace Rosen H, Fujishiro AA, Luppino F, Zou K, Seregin A. Development and characterization of a first-in-class adjustable-dose gene therapy system. Gene 2024; 919:148500. [PMID: 38663689 DOI: 10.1016/j.gene.2024.148500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
INTRODUCTION Despite significant potential, gene therapy has been relegated to the treatment of rare diseases, due in part to an inability to adjust dosage following initial administration. Other significant constraints include cost, specificity, antigenicity, and systemic toxicity of current generation technologies. To overcome these challenges, we developed a first-in-class adjustable-dose gene therapy system, with optimized biocompatibility, localization, durability, and cost. METHODS A lipid nanoparticle (LNP) delivery system was developed and characterized by dynamic light scattering for size, zeta potential, and polydispersity. Cytocompatibility and transfection efficiency were optimized in vitro using primary human adipocytes and preadipocytes. Durability, immunogenicity, and adjustment of expression were evaluated in C57BL/6 and B6 albino mice using in vivo bioluminescence imaging. Biodistribution was assessed by qPCR and immunohistochemistry; therapeutic protein expression was quantified by ELISA. RESULTS Following LNP optimization, in vitro transfection efficiency of primary human adipocytes reached 81.3 % ± 8.3 % without compromising cytocompatibility. Critical physico-chemical properties of the system (size, zeta potential, polydispersity) remained stable over a broad range of genetic cassette sizes (1,871-6,203 bp). Durable expression was observed in vivo over 6 months, localizing to subcutaneous adipose tissues at the injection site with no detectable transgene in the liver, heart, spleen, or kidney. Gene expression was adjustable using several physical and pharmacological approaches, including cryolipolysis, focused ultrasound, and pharmacologically inducible apoptosis. The ability of transfected adipocytes to express therapeutic transgenes ranging from peptides to antibodies, at potentially clinically relevant levels, was confirmed in vitro and in vivo. CONCLUSION We report the development of a novel, low-cost therapeutic platform, designed to enable the replacement of subcutaneously administered protein treatments with a single-injection, adjustable-dose gene therapy.
Collapse
Affiliation(s)
- Alex Goraltchouk
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA 02492, United States of America
| | - Jared Lourie
- Department of Exercise and Health Sciences, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125, United States of America
| | - Judith M Hollander
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA 02492, United States of America
| | - H Grace Rosen
- Department of Biology, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125, United States of America
| | - Atsutaro A Fujishiro
- Department of Exercise and Health Sciences, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125, United States of America
| | - Francesco Luppino
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA 02492, United States of America
| | - Kai Zou
- Department of Exercise and Health Sciences, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125, United States of America
| | - Alexey Seregin
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA 02492, United States of America.
| |
Collapse
|
14
|
Yoshijima C, Suzuki Y, Tanaka R, Ono H, Oda A, Ozaki T, Shibata H, Itoh H, Ohno K. Evaluation of intra- and inter-individual variations in plasma belimumab concentrations in adult patients with systemic lupus erythematosus. Pharmacol Res Perspect 2024; 12:e1255. [PMID: 39096038 PMCID: PMC11297282 DOI: 10.1002/prp2.1255] [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: 05/07/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
In this study, plasma belimumab concentrations were measured over the course of treatment in systemic lupus erythematosus (SLE) patients on belimumab therapy, and intra- and interindividual variations in plasma belimumab concentration were evaluated. A single-center prospective study was conducted at Oita University Hospital to evaluate trough plasma concentrations over the course of treatment in 13 SLE patients treated with intravenous belimumab. Plasma belimumab concentrations were measured by a validated ultra-high performance liquid chromatography with tandem mass spectrometry method. The median age of the patients was 40 (interquartile range: 35-51) years and the median weight was 51.8 (47.0-58.1) kg. A mean of 9.4 (range: 1-13) blood samples was collected per patient at routine visits. The mean (± SD) plasma belimumab concentration was 33.4 ± 11.9 μg/mL in the patient with the lowest concentration and 170.0 ± 16.6 μg/mL in the patient with the highest concentration, indicating a 5-fold difference between patients. On the other hand, the within-patient coefficient of variation ranged from 7.1% to 35.7%, showing no large variations. No significant correlation was observed between plasma belimumab concentration and belimumab dose (mg/kg) (Spearman's rank correlation coefficient = 0.22, p = .54). Examinations of trough plasma belimumab concentrations over the course of treatment in patients with SLE showed small intraindividual variation but large interindividual variation. Plasma belimumab trough concentration varied widely among patients administered the approved dose.
Collapse
MESH Headings
- Humans
- Lupus Erythematosus, Systemic/drug therapy
- Lupus Erythematosus, Systemic/blood
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/blood
- Adult
- Female
- Middle Aged
- Prospective Studies
- Male
- Immunosuppressive Agents/pharmacokinetics
- Immunosuppressive Agents/blood
- Immunosuppressive Agents/administration & dosage
- Immunosuppressive Agents/therapeutic use
- Tandem Mass Spectrometry
- Chromatography, High Pressure Liquid
- Drug Monitoring/methods
Collapse
Affiliation(s)
- Chisato Yoshijima
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| | - Yosuke Suzuki
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| | - Ryota Tanaka
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Hiroyuki Ono
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Ayako Oda
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| | - Takashi Ozaki
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of MedicineOita UniversityYufuOitaJapan
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of MedicineOita UniversityYufuOitaJapan
| | - Hiroki Itoh
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Keiko Ohno
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| |
Collapse
|
15
|
Tao X, Sukumaran S, Sperinde G, Liu C, Beardsley MI, Day P, Kalo M, Ayewoh E, Cai H, Wang Y, Jun I, Hirst K, Nguyen V, Chung S, Lee D, Lekkerkerker A, Stefanich E. Sialic Acid Mediated Endothelial and Hepatic Uptake: A Mechanism based Mathematic Model Elucidating the Complex Pharmacokinetics and Pharmacodynamics of Efmarodocokin Alfa, a Variably Glycosylated Fusion Protein. J Pharm Sci 2024; 113:1975-1986. [PMID: 38561054 DOI: 10.1016/j.xphs.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024]
Abstract
Sialic acid (SA) is crucial for protecting glycoproteins from clearance. Efmarodocokin alfa (IL-22Fc), a fusion protein agonist that links IL-22 to the crystallizable fragment (Fc) of human IgG4, contains 8 N-glycosylation sites and exhibits heterogeneous and variable terminal sialylation biodistribution. This presents a unique challenge for Pharmacokinetic (PK) and Pharmacodynamic (PD) analysis and cross-species translation. In this study, we sought to understand how varying SA levels and heterogeneous distribution contribute to IL-22Fc's complex PKPD properties. We initially used homogenous drug material with varying SA levels to examine PKPD in mice. Population PKPD analysis based on mouse data revealed that SA was a critical covariate simultaneously accounting for the substantial between subject variability (BSV) in clearance (CL), distribution clearance (CLd), and volume of distribution (Vd). In addition to the well-established mechanism by which SA inhibits ASGPR activity, we hypothesized a novel mechanism by which decrease in SA increases the drug uptake by endothelial cells. This decrease in SA, leading to more endothelial uptake, was supported by the neonatal Fc receptor (FcRn) dependent cell-based transcytosis assay. The population analysis also suggested in vivo EC50 (IL-22Fc stimulating Reg3β) was independent on SA, while the in-vitro assay indicated a contradictory finding of SA-in vitro potency relationship. We created a mechanism based mathematical (MBM) PKPD model incorporating the decrease in SA mediated endothelial and hepatic uptake, and successfully characterized the SA influence on IL-22Fc PK, as well as the increased PK exposure being responsible for increased PD. Thereby, the MBM model supported that SA has no direct impact on EC50, aligning with the population PKPD analysis. Subsequently, using the MBM PKPD model, we employed 5 subpopulation simulations to reconstitute the heterogeneity of drug material. The simulation accurately predicted the PKPD of heterogeneously and variably sialylated drug in mouse, monkey and human. The successful prospective validation confirmed the MBM's ability to predict IL-22Fc PK across variable SA levels, homogenous to heterogeneous material, and across species (R2=0.964 for clearance prediction). Our model prediction suggests an average of 1 mol/mol SA increase leads to a 50% increase in drug exposure. This underlines the significance of controlling sialic acid levels during lot-to-lot manufacturing.
Collapse
Affiliation(s)
- Xun Tao
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Siddharth Sukumaran
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA; Now at Janssen: Pharmaceutical Companies of Johnson & Johnson, 1125 Trenton-Harbourton Road, Titusville, NJ 08560, USA
| | | | - Chang Liu
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Peter Day
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Matt Kalo
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Hao Cai
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yehong Wang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA; Now at Gilead Sciences, Inc, 333 Lakeside Drive. Foster City, CA 94404, USA
| | - Inyoung Jun
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA; Now at University of Florida, Gainesville, FL 32611, USA
| | - Kyle Hirst
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Van Nguyen
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Shan Chung
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Donna Lee
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Eric Stefanich
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
16
|
Kátai CB, Smithline SJ, Thalhauser CJ, Bosgra S, Elassaiss-Schaap J. An asymptotic description of a basic FcRn-regulated clearance mechanism and its implications for PBPK modelling of large antibodies. J Pharmacokinet Pharmacodyn 2024:10.1007/s10928-024-09925-8. [PMID: 38914910 DOI: 10.1007/s10928-024-09925-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/07/2024] [Indexed: 06/26/2024]
Abstract
A basic FcRn-regulated clearance mechanism is investigated using the method of matched asymptotic expansions. The broader aim of the work is to obtain further insight on the mechanism, thereby providing theoretical support for future pharmacologically-based pharmacokinetic modelling efforts. The corresponding governing equations are first non-dimensionalised and the order of magnitudes of the model parameters are assessed based on their values reported in the literature. Under the assumption of high FcRn-binding affinity, analytical approximations are derived that are valid over the characteristic phases of the problem. Additionally, relatively simple equations relating clearance and AUC to physiological model parameters are derived, which are valid over the longest characteristic time scale of the problem. For lower to moderate doses clearance is effectively linear, whereas for higher doses it is nonlinear. It is shown that for all doses sufficiently high the leading-order approximation for the IgG concentration in plasma, over the longest characteristic time scale, is independent of the initial dose. This is because IgG that is in 'excess' of FcRn is eliminated over a time scale much shorter than that of the terminal phase. In conclusion, analytical approximations of the basic FcRn mechanism have been derived using matched asymptotic expansions, leading to a simple equation relating clearance to FcRn binding affinity, the ratio of degradation and FcRn concentration, and the volumes of the system.
Collapse
Affiliation(s)
- Csaba B Kátai
- PD-value B.V., Yalelaan 1, 3584, Utrecht, CL, The Netherlands.
| | | | | | - Sieto Bosgra
- Genmab B.V., Uppsalalaan 15, 3584, Utrecht, CT, The Netherlands
| | | |
Collapse
|
17
|
Broer LN, Knapen DG, de Groot DJA, Mol PG, Kosterink JG, de Vries EG, Lub-de Hooge MN. Monoclonal antibody biosimilars for cancer treatment. iScience 2024; 27:110115. [PMID: 38974466 PMCID: PMC11225859 DOI: 10.1016/j.isci.2024.110115] [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] [Indexed: 07/09/2024] Open
Abstract
Monoclonal antibodies are important cancer medicines. The European Medicines Agency (EMA) approved 48 and the Food and Drug Administration (FDA) 56 anticancer monoclonal antibody-based therapies. Their high prices burden healthcare systems and hamper global drug access. Biosimilars could retain costs and expand the availability of monoclonal antibodies. In Europe, five rituximab biosimilars, six trastuzumab biosimilars, and eight bevacizumab biosimilars are available as anti-cancer drugs. To gain insight into the biosimilar landscape for cancer treatment, we performed a literature search and analysis. In this review, we summarize cancer monoclonal antibodies' properties crucial for the desired pharmacology and point out sources of variability. The analytical assessment of all EMA-approved bevacizumab biosimilars is highlighted to illustrate this variability. The global landscape of investigational and approved biosimilars is mapped, and the challenges for access to cancer biosimilars are identified.
Collapse
Affiliation(s)
- Linda N. Broer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Daan G. Knapen
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Derk-Jan A. de Groot
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Peter G.M. Mol
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jos G.W. Kosterink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Pharmaco-, Therapy-, Epidemiology- and Economy, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Elisabeth G.E. de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marjolijn N. Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
18
|
Zheng Y, Chen G, Liu G, Rana GE, Wang C. A single-step high-throughput bioassay for quantifying Fc-containing recombinant proteins based on non-classical calculation of fluorescence polarization. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:3917-3926. [PMID: 38832468 DOI: 10.1039/d4ay00372a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
The titer of recombinant proteins is one of the key parameters in biopharmaceutical manufacturing processes. The fluorescence polarization (FP)-based assay, a homogeneous, high-throughput and real-time analytical method, had emerged as a powerful tool for biochemical analysis and environmental monitoring. In this study, an FP-based bioassay was utilized to quantify antibody fragment crystallizable (Fc)-containing proteins, such as recombinant monoclonal antibodies (mAbs) and mAb derivatives, in the cell culture supernatant, and the impacts of tracer molecular weight and FITC-coupling conditions on fluorescence polarization were methodically examined. Distinct from the fluorescence polarization potency calculated by classical formula, we for the first time proposed a new concept and calculation of fluorescence polarization intensity, based on which an analytical method with broader detection range and analysis window was established for quantifying Fc-containing proteins. This provided new ideas for the practical application of fluorescence polarization theory. The established method could detect 96 samples within 30 minutes, with dynamic titer range of 2.5-400 mg L-1, and a linear fitting R2 between the measured and actual concentration reaching 0.99. The method had great application prospects in determining the titer of recombinant proteins with Fc fragments, especially when applied to large-scale screening of high-yield and stable expression CHO cell lines commonly used in biopharmaceutical industry.
Collapse
Affiliation(s)
- Yujuan Zheng
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ganjun Chen
- Dartsbio Pharmaceuticals Co., Ltd, Zhongshan 528400, China
| | - Guojian Liu
- Shanghai Mabstone Biotechnology Co., Ltd, Shanghai 201203, China
| | - Gul E Rana
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunhe Wang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
- Dartsbio Pharmaceuticals Co., Ltd, Zhongshan 528400, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
19
|
Daneels W, Van Parys A, Huyghe L, Rogge E, De Rouck S, Christiaen R, Zabeau L, Taveirne S, Van Dorpe J, Kley N, Cauwels A, Depla E, Tavernier J, Offner F. High efficacy of huCD20-targeted AcTaferon in humanized patient derived xenograft models of aggressive B cell lymphoma. Exp Hematol Oncol 2024; 13:59. [PMID: 38831452 PMCID: PMC11145843 DOI: 10.1186/s40164-024-00524-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 05/13/2024] [Indexed: 06/05/2024] Open
Abstract
Type I interferon (IFN) is a potent antitumoral drug, with an important history in the treatment of hematologic malignancies. However, its pleiotropic nature leads to severe dose-limiting toxicities that blunt its therapeutic potential. To achieve selective targeting of specific immune or tumor cells, AcTakines (Activity-on-Target Cytokines), i.e., immunocytokines utilizing attenuated cytokines, and clinically optimized A-Kines™ were developed. In syngeneic murine models, the CD20-targeted murine IFNα2-based AcTaferons (AFNs) have demonstrated clear antitumoral effects, with excellent tolerability. The current study explores the antitumoral potential of the humanized huCD20-Fc-AFN in 5 different humanized patient derived xenograft (PDX) models of huCD20+ aggressive B non-Hodgkin lymphomas (B-NHLs). The huCD20-Fc-AFN consists of a huCD20-specific single-domain antibody (VHH) linked through a heterodimeric 'knob-in-hole' human IgG1 Fc molecule to an attenuated huIFNα2 sequence. An in vitro targeting efficacy of up to 1.000-fold could be obtained, without detectable in vivo toxicities, except for selective (on-target) and reversible B cell depletion. Treatment with huCD20-Fc-AFN significantly increased the median overall survival (mOS) in both non-humanized (mOS 31 to 45 days; HR = 0.26; p = 0.001), and humanized NSG/NOG mice (mOS 34 to 80 days; HR = 0.37; p < 0.0001). In humanized mice, there was a trend for increased survival when compared to equimolar rituximab (mOS 49 to 80 days; HR = 0.73; p = 0.09). The antitumoral effects of huCD20-Fc-AFN were partly due to direct effects of type I IFN on the tumor cells, but additional effects via the human immune system are essential to obtain long-term remissions. To conclude, huCD20-Fc-AFN could provide a novel therapeutic strategy for huCD20-expressing aggressive B-NHLs.
Collapse
Affiliation(s)
- Willem Daneels
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.
- Department of Hematology, Ghent University Hospital, C. Heymanslaan 10, 9000, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium.
| | - Alexander Van Parys
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Orionis Biosciences BV, Ghent, Belgium
| | - Leander Huyghe
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Orionis Biosciences BV, Ghent, Belgium
| | - Elke Rogge
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Orionis Biosciences BV, Ghent, Belgium
| | - Steffi De Rouck
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Orionis Biosciences BV, Ghent, Belgium
| | | | | | | | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Niko Kley
- Orionis Biosciences BV, Ghent, Belgium
| | - Anje Cauwels
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Orionis Biosciences BV, Ghent, Belgium
| | | | - Jan Tavernier
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Orionis Biosciences BV, Ghent, Belgium
| | - Fritz Offner
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Hematology, Ghent University Hospital, C. Heymanslaan 10, 9000, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| |
Collapse
|
20
|
Rosenstock J, Juneja R, Beals JM, Moyers JS, Ilag L, McCrimmon RJ. The Basis for Weekly Insulin Therapy: Evolving Evidence With Insulin Icodec and Insulin Efsitora Alfa. Endocr Rev 2024; 45:379-413. [PMID: 38224978 PMCID: PMC11091825 DOI: 10.1210/endrev/bnad037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Indexed: 01/17/2024]
Abstract
Basal insulin continues to be a vital part of therapy for many people with diabetes. First attempts to prolong the duration of insulin formulations were through the development of suspensions that required homogenization prior to injection. These insulins, which required once- or twice-daily injections, introduced wide variations in insulin exposure contributing to unpredictable effects on glycemia. Advances over the last 2 decades have resulted in long-acting, soluble basal insulin analogues with prolonged and less variable pharmacokinetic exposure, improving their efficacy and safety, notably by reducing nocturnal hypoglycemia. However, adherence and persistence with once-daily basal insulin treatment remains low for many reasons including hypoglycemia concerns and treatment burden. A soluble basal insulin with a longer and flatter exposure profile could reduce pharmacodynamic variability, potentially reducing hypoglycemia, have similar efficacy to once-daily basal insulins, simplify dosing regimens, and improve treatment adherence. Insulin icodec (Novo Nordisk) and insulin efsitora alfa (basal insulin Fc [BIF], Eli Lilly and Company) are 2 such insulins designed for once-weekly administration, which have the potential to provide a further advance in basal insulin replacement. Icodec and efsitora phase 2 clinical trials, as well as data from the phase 3 icodec program indicate that once-weekly insulins provide comparable glycemic control to once-daily analogues, with a similar risk of hypoglycemia. This manuscript details the technology used in the development of once-weekly basal insulins. It highlights the clinical rationale and potential benefits of these weekly insulins while also discussing the limitations and challenges these molecules could pose in clinical practice.
Collapse
Affiliation(s)
- Julio Rosenstock
- Velocity Clinical Research at Medical City,
Dallas, TX 75230, USA
| | - Rattan Juneja
- Lilly Diabetes and Obesity, Eli Lilly and Company,
Indianapolis, IN 46225, USA
| | - John M Beals
- Lilly Diabetes and Obesity, Eli Lilly and Company,
Indianapolis, IN 46225, USA
| | - Julie S Moyers
- Lilly Diabetes and Obesity, Eli Lilly and Company,
Indianapolis, IN 46225, USA
| | - Liza Ilag
- Lilly Diabetes and Obesity, Eli Lilly and Company,
Indianapolis, IN 46225, USA
| | - Rory J McCrimmon
- School of Medicine, University of Dundee, Dundee
DD1 9SY, Scotland, UK
| |
Collapse
|
21
|
Janković SM, Janković SV. Anti-calcitonin Gene-Related Peptide Monoclonal Antibodies in Migraine: Focus on Clinical Pharmacokinetics. Eur J Drug Metab Pharmacokinet 2024; 49:277-293. [PMID: 38461486 DOI: 10.1007/s13318-024-00885-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2024] [Indexed: 03/12/2024]
Abstract
The calcitonin gene-related peptide transmission was the target for recent development of drugs that effectively prevent attacks of both episodic and chronic migraine. The aim of this narrative review was to offer deeper insight into pharmacokinetics of monoclonal antibodies approved for prevention of migraine attacks. For this narrative review, relevant literature was searched for in MEDLINE and Google Scholar databases, covering periods 1966-2023 and 2006-2023, respectively. The ClinicalTrials.gov database was also searched for relevant clinical studies whose results had not been published previously in medical journals, covering the period 2000-2023. The monoclonal antibodies from this group are distributed mainly in the plasma and part of the extracellular space; they are neither metabolized in the liver nor excreted via the kidneys. The elimination of galcanezumab, eptinezumab and fremanezumab takes place only by a non-specific linear process via the reticuloendothelial system in the liver, while erenumab is eliminated by a non-specific process and by a specific, saturable process because of binding to receptors located on the cell membrane. Since the elimination processes do not have a large capacity, the half-life is about 2 weeks for erenumab and about 4 weeks for other monoclonal antibodies. Variability in the pharmacokinetics of these monoclonal antibodies is small in different subpopulations, and body weight is the only parameter to consider when choosing the dose of these drugs.
Collapse
Affiliation(s)
- Slobodan M Janković
- Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića Street, 69, 34000, Kragujevac, Serbia.
| | - Snežana V Janković
- Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića Street, 69, 34000, Kragujevac, Serbia
| |
Collapse
|
22
|
Wang H, Song M, Xu J, Liu Z, Peng M, Qin H, Wang S, Wang Z, Liu K. Long-Acting Strategies for Antibody Drugs: Structural Modification, Controlling Release, and Changing the Administration Route. Eur J Drug Metab Pharmacokinet 2024; 49:295-316. [PMID: 38635015 DOI: 10.1007/s13318-024-00891-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2024] [Indexed: 04/19/2024]
Abstract
Because of their high specificity, high affinity, and targeting, antibody drugs have been widely used in the treatment of many diseases and have become the most favored new drugs for research in the world. However, some antibody drugs (such as small-molecule antibody fragments) have a short half-life and need to be administered frequently, and are often associated with injection-site reactions and local toxicities during use. Increasing attention has been paid to the development of antibody drugs that are long-acting and have fewer side effects. This paper reviews existing strategies to achieve long-acting antibody drugs, including modification of the drug structure, the application of drug delivery systems, and changing their administration route. Among these, microspheres have been studied extensively regarding their excellent tolerance at the injection site, controllable loading and release of drugs, and good material safety. Subcutaneous injection is favored by most patients because it can be quickly self-administered. Subcutaneous injection of microspheres is expected to become the focus of developing long-lasting antibody drug strategies in the near future.
Collapse
Affiliation(s)
- Hao Wang
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai Ocean University, Hucheng Ring Road, Shanghai, 201306, China
| | - Mengdi Song
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai Ocean University, Hucheng Ring Road, Shanghai, 201306, China
| | - Jiaqi Xu
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai Ocean University, Hucheng Ring Road, Shanghai, 201306, China
| | - Zhenjing Liu
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai Ocean University, Hucheng Ring Road, Shanghai, 201306, China
| | - Mingyue Peng
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai Ocean University, Hucheng Ring Road, Shanghai, 201306, China
| | - Haoqiang Qin
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai Ocean University, Hucheng Ring Road, Shanghai, 201306, China
| | - Shaoqian Wang
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai Ocean University, Hucheng Ring Road, Shanghai, 201306, China
| | - Ziyang Wang
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai Ocean University, Hucheng Ring Road, Shanghai, 201306, China
| | - Kehai Liu
- College of Food, Shanghai Ocean University, 999 Hucheng Ring Road, Nanhui New Town, Pudong New Area, Shanghai, 201306, China.
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai Ocean University, Hucheng Ring Road, Shanghai, 201306, China.
| |
Collapse
|
23
|
Courville J, Roupe K, Arold G. Re-discover the value of protein binding assessments in hepatic and renal impairment studies and its contributions in drug labels and dose decisions. Clin Transl Sci 2024; 17:e13810. [PMID: 38716900 PMCID: PMC11077687 DOI: 10.1111/cts.13810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/05/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
One of the key pharmacokinetic properties of most small molecule drugs is their ability to bind to serum proteins. Unbound or free drug is responsible for pharmacological activity while the balance between free and bound drug can impact drug distribution, elimination, and other safety parameters. In the hepatic impairment (HI) and renal impairment (RI) clinical studies, unbound drug concentration is often assessed; however, the relevance and impact of the protein binding (PB) results is largely limited. We analyzed published clinical safety and pharmacokinetic studies in subjects with HI or RI with PB assessment up to October 2022 and summarized the contribution of PB results on their label dose recommendations. Among drugs with HI publication, 32% (17/53) associated product labels include PB results in HI section. Of these, the majority (9/17, 53%) recommend dose adjustments consistent with observed PB change. Among drugs with RI publication, 27% (12/44) of associated product labels include PB results in RI section with the majority (7/12, 58%) recommending no dose adjustment, consistent with the reported absence of PB change. PB results were found to be consistent with a tailored dose recommendation in 53% and 58% of the approved labels for HI and RI section, respectively. We further discussed the interpretation challenges of PB results, explored treatment decision factors including total drug concentration, exposure-response relationships, and safety considerations in these case examples. Collectively, comprehending the alterations in free drug levels in HI and RI informs treatment decision through a risk-based approach.
Collapse
Affiliation(s)
- Jocelyn Courville
- Clinical Pharmacology—Drug Development SolutionICON plcBlue BellPennsylvaniaUSA
| | - Kathryn Roupe
- Clinical Pharmacology, PharmacokineticsWorldwide Clinical TrialsAustinTexasUSA
| | - Gerhard Arold
- Clinical Pharmacology—Drug Development SolutionICON plcLangenGermany
| |
Collapse
|
24
|
Wu R, Kahl DM, Kloberdanz R, Rohilla KJ, Balasubramanian S. Demonstration of a robust high cell density transient CHO platform yielding mAb titers of up to 2 g/L without medium exchange. Biotechnol Prog 2024; 40:e3435. [PMID: 38329375 DOI: 10.1002/btpr.3435] [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: 10/04/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/09/2024]
Abstract
Biopharmaceuticals like therapeutic monoclonal antibodies (mAbs) and other derived proteins are popular for treating various diseases. Transient gene expression (TGE) is typically used as a fast yet efficient method to generate moderate amounts of material. It has been used to support early stage research and discovery processes. Introduction of a robust high yielding and predictive TGE platform in Chinese hamster ovary (CHO) is crucial. It maintains the consistency in cell lines and processes throughout the early drug discovery and downstream manufacturing processes. This helps researchers to identify the issues at an early stage for timely resolution. In this study, we have demonstrated a simple high-titer platform for TGE in CHO based on a dilution process of seeding cells. We achieved titers ranging from 0.8 to 1.9 g/L for eight model mAbs at three scales (1, 30, 100 mL) in 10 days using our new platform. The ability to seed by dilution significantly streamlined the process and dramatically enhanced platform throughput. We observed a modest reduction in titer ranging from 11% to 28% when cells were seeded using dilution compared to when cells were seeded using medium exchange. Further studies revealed that carry over of spent medium into transfection negatively affected the DNA uptake and transcription processes, while the translation and secretion was minimally impacted. In summary, our transient CHO platform using cells prepared by dilution at high densities can achieve high titers of up to 1.9 g/L, which can be further improved by targeting the bottlenecks of transfection and transcription.
Collapse
Affiliation(s)
- Rigumula Wu
- Department of Cell Culture and Bioprocess Operations, Genentech, Inc, San Francisco, California, USA
| | - Danielle M Kahl
- Department of Cell Culture and Bioprocess Operations, Genentech, Inc, San Francisco, California, USA
| | - Ronald Kloberdanz
- Department of Cell Culture and Bioprocess Operations, Genentech, Inc, San Francisco, California, USA
| | - Kushal J Rohilla
- Department of Cell Culture and Bioprocess Operations, Genentech, Inc, San Francisco, California, USA
| | - Sowmya Balasubramanian
- Department of Cell Culture and Bioprocess Operations, Genentech, Inc, San Francisco, California, USA
| |
Collapse
|
25
|
Go EB, Lee JH, Cho JH, Kwon NH, Choi JI, Kwon I. Enhanced therapeutic potential of antibody fragment via IEDDA-mediated site-specific albumin conjugation. J Biol Eng 2024; 18:23. [PMID: 38576037 PMCID: PMC10996255 DOI: 10.1186/s13036-024-00418-3] [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: 11/11/2023] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND The use of single-chain variable fragments (scFvs) for treating human diseases, such as cancer and immune system disorders, has attracted significant attention. However, a critical drawback of scFv is its extremely short serum half-life, which limits its therapeutic potential. Thus, there is a critical need to prolong the serum half-life of the scFv for clinical applications. One promising serum half-life extender for therapeutic proteins is human serum albumin (HSA), which is the most abundant protein in human serum, known to have an exceptionally long serum half-life. However, conjugating a macromolecular half-life extender to a small protein, such as scFv, often results in a significant loss of its critical properties. RESULTS In this study, we conjugated the HSA to a permissive site of scFv to improve pharmacokinetic profiles. To ensure minimal damage to the antigen-binding capacity of scFv upon HSA conjugation, we employed a site-specific conjugation approach using a heterobifunctional crosslinker that facilitates thiol-maleimide reaction and inverse electron-demand Diels-Alder reaction (IEDDA). As a model protein, we selected 4D5scFv, derived from trastuzumab, a therapeutic antibody used in human epithermal growth factor 2 (HER2)-positive breast cancer treatment. We introduced a phenylalanine analog containing a very reactive tetrazine group (frTet) at conjugation site candidates predicted by computational methods. Using the linker TCO-PEG4-MAL, a single HSA molecule was site-specifically conjugated to the 4D5scFv (4D5scFv-HSA). The 4D5scFv-HSA conjugate exhibited HER2 binding affinity comparable to that of unmodified 4D5scFv. Furthermore, in pharmacokinetic profile in mice, the serum half-life of 4D5scFv-HSA was approximately 12 h, which is 85 times longer than that of 4D5scFv. CONCLUSIONS The antigen binding results and pharmacokinetic profile of 4D5scFv-HSA demonstrate that the site-specifically albumin-conjugated scFv retained its binding affinity with a prolonged serum half-life. In conclusion, we developed an effective strategy to prepare site-specifically albumin-conjugated 4D5scFv, which can have versatile clinical applications with improved efficacy.
Collapse
Affiliation(s)
- Eun Byeol Go
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jae Hun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jeong Haeng Cho
- ProAbTech, Gwangju, 61005, Republic of Korea
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Na Hyun Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jong-Il Choi
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Inchan Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
| |
Collapse
|
26
|
Biesdorf C, Guan X, Siddani SR, Hoffman D, Boehm N, Medeiros BC, Doi T, de Jonge M, Rasco D, Menon RM, Polepally AR. Pharmacokinetics and immunogenicity of eftozanermin alfa in subjects with previously-treated solid tumors or hematologic malignancies: results from a phase 1 first-in-human study. Cancer Chemother Pharmacol 2024; 93:329-339. [PMID: 38036720 DOI: 10.1007/s00280-023-04613-9] [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: 07/25/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023]
Abstract
PURPOSE Eftozanermin alfa is a second-generation tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor agonist that enhances death receptor 4/5 clustering on tumor cells to induce apoptosis. We report the pharmacokinetics and immunogenicity of eftozanermin alfa administered intravenously to 153 adults with previously-treated solid tumors or hematologic malignancies from the first-in-human, open-label, dose-escalation and dose-optimization study. METHODS Dose escalation evaluated eftozanermin alfa monotherapy 2.5-15 mg/kg on Day 1 or Days 1/8 of a 21-day cycle. Dose optimization evaluated eftozanermin alfa monotherapy or combination therapy with either oral venetoclax 400-800 mg daily (eftozanermin alfa 1.25-7.5 mg/kg Days 1/8/15 of a 21-day cycle) or chemotherapy (eftozanermin alfa 3.75 or 7.5 mg/kg Days 1/8/15/22 of a 28-day cycle and FOLFIRI regimen [leucovorin, 5-fluorouracil, and irinotecan] with/without bevacizumab on Days 1/15 of a 28-day cycle). RESULTS Systemic exposures (maximum observed concentration [Cmax] and area under the concentration-time curve [AUC]) of eftozanermin alfa were approximately dose-proportional across the entire dose escalation range with minimal to no accumulation in Cycle 3 versus Cycle 1 exposures. Comparable exposures and harmonic mean half-lives (35.1 h [solid tumors], 31.3 h [hematologic malignancies]) were observed between malignancy types. Exposures (dose-normalized Cmax and AUC) in Japanese subjects were similar to non-Japanese subjects. Furthermore, eftozanermin alfa/venetoclax combination therapy did not have an impact on the exposures of either agent. Treatment-emergent anti-drug antibodies were observed in 9.4% (13/138) of subjects. CONCLUSIONS The study results, including a pharmacokinetic profile consistent with weekly dosing and low incidence of immunogenicity, support further investigation of eftozanermin alfa. TRIAL REGISTRATION ID NCT03082209.
Collapse
Affiliation(s)
- Carla Biesdorf
- Clinical Pharmacology, AbbVie Inc., 1 North Waukegan Road, Bldg. AP31-3, North Chicago, IL, 60064, USA.
| | - Xiaowen Guan
- AbbVie Biotherapeutics Inc., South San Francisco, CA, USA
| | - Satya R Siddani
- Clinical Pharmacology, AbbVie Inc., 1 North Waukegan Road, Bldg. AP31-3, North Chicago, IL, 60064, USA
| | - David Hoffman
- Clinical Pharmacology, AbbVie Inc., 1 North Waukegan Road, Bldg. AP31-3, North Chicago, IL, 60064, USA
| | | | | | - Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | | | - Drew Rasco
- South Texas Accelerated Research Therapeutics (START), San Antonio, TX, USA
| | - Rajeev M Menon
- Clinical Pharmacology, AbbVie Inc., 1 North Waukegan Road, Bldg. AP31-3, North Chicago, IL, 60064, USA
| | | |
Collapse
|
27
|
Harris CT, Cohen S. Reducing Immunogenicity by Design: Approaches to Minimize Immunogenicity of Monoclonal Antibodies. BioDrugs 2024; 38:205-226. [PMID: 38261155 PMCID: PMC10912315 DOI: 10.1007/s40259-023-00641-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 01/24/2024]
Abstract
Monoclonal antibodies (mAbs) have transformed therapeutic strategies for various diseases. Their high specificity to target antigens makes them ideal therapeutic agents for certain diseases. However, a challenge to their application in clinical practice is their potential risk to induce unwanted immune response, termed immunogenicity. This challenge drives the continued efforts to deimmunize these protein therapeutics while maintaining their pharmacokinetic properties and therapeutic efficacy. Because mAbs hold a central position in therapeutic strategies against an array of diseases, the importance of conducting comprehensive immunogenicity risk assessment during the drug development process cannot be overstated. Such assessment necessitates the employment of in silico, in vitro, and in vivo strategies to evaluate the immunogenicity risk of mAbs. Understanding the intricacies of the mechanisms that drive mAb immunogenicity is crucial to improving their therapeutic efficacy and safety and developing the most effective strategies to determine and mitigate their immunogenic risk. This review highlights recent advances in immunogenicity prediction strategies, with a focus on protein engineering strategies used throughout development to reduce immunogenicity.
Collapse
Affiliation(s)
- Chantal T Harris
- Department of BioAnalytical Sciences, Genentech Inc., South San Francisco, CA, 94080-4990, USA
| | - Sivan Cohen
- Department of BioAnalytical Sciences, Genentech Inc., South San Francisco, CA, 94080-4990, USA.
| |
Collapse
|
28
|
Lee G, Kim A, Kang HR, Hwang JH, Park JH, Lee MJ, Kim B, Kim SM. Porcine interferon-α linked to the porcine IgG-Fc induces prolonged and broad-spectrum antiviral effects against foot-and-mouth disease virus. Antiviral Res 2024; 223:105836. [PMID: 38360296 DOI: 10.1016/j.antiviral.2024.105836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/22/2024] [Accepted: 02/13/2024] [Indexed: 02/17/2024]
Abstract
Foot-and-mouth disease (FMD) is an economically important disease, and the FMD virus (FMDV) can spread rapidly in susceptible animals. FMD is usually controlled through vaccination. However, commercial FMD vaccines are only effective 4-7 days after vaccination. Furthermore, FMDV comprises seven serotypes and various topotypes, and these aspects should be considered when selecting a vaccine. Antiviral agents could provide rapid and broad protection against FMDV. Therefore, this study aimed to develop a fusion protein of consensus porcine interferon-α and Fc portion of porcine antibody IgG (poIFN-α-Fc) using a baculovirus expression system to develop a novel antiviral agent against FMDV. We measured the antiviral effects of the poIFN-α-Fc protein against FMDV and the enhanced duration in vitro and in vivo. The broad-spectrum antiviral effects were tested against seven FMDV serotypes, porcine reproductive and respiratory syndrome virus (PRRSV), and bovine enterovirus (BEV). Furthermore, the early protective effects and neutralizing antibody levels were tested by co-injecting poIFN-α-Fc and an FMD-inactivated vaccine into mice or pigs. Sustained antiviral effects in pig sera and mice were observed, and pigs injected with a combination of the poIFN-α-Fc and an inactivated FMD vaccine were protected against FMDV in a dose-dependent manner at 2- and 4-days post-vaccination. In addition, combined with the inactivated FMD vaccine, poIFN-α-Fc increased the neutralizing antibody levels in mice. Therefore, poIFN-α-Fc is a potential broad-spectrum antiviral and adjuvant candidate that can be used with inactivated FMD vaccines to protect pigs against FMDV.
Collapse
Affiliation(s)
- Gyeongmin Lee
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-City, Gyeongsangbuk-do, Republic of Korea
| | - Aro Kim
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-City, Gyeongsangbuk-do, Republic of Korea
| | - Hyo Rin Kang
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-City, Gyeongsangbuk-do, Republic of Korea
| | - Ji-Hyeon Hwang
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-City, Gyeongsangbuk-do, Republic of Korea
| | - Jong-Hyeon Park
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-City, Gyeongsangbuk-do, Republic of Korea
| | - Min Ja Lee
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-City, Gyeongsangbuk-do, Republic of Korea
| | - Byounghan Kim
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-City, Gyeongsangbuk-do, Republic of Korea
| | - Su-Mi Kim
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-City, Gyeongsangbuk-do, Republic of Korea.
| |
Collapse
|
29
|
Lteif M, Pallardy M, Turbica I. Antibodies internalization mechanisms by dendritic cells and their role in therapeutic antibody immunogenicity. Eur J Immunol 2024; 54:e2250340. [PMID: 37985174 DOI: 10.1002/eji.202250340] [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: 05/18/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Internalization and processing by antigen-presenting cells such as dendritic cells (DCs) are critical steps for initiating a T-cell response to therapeutic antibodies. Consequences are the production of neutralizing antidrug antibodies altering the clinical response, the presence of immune complexes, and, in some rare cases, hypersensitivity reactions. In recent years, significant progress has been made in the knowledge of cellular uptake mechanisms of antibodies in DCs. The uptake of antibodies could be directly related to their immunogenicity by regulating the quantity of materials entering the DCs in relation to antibody structure. Here, we summarize the latest insights into cellular uptake mechanisms and pathways in DCs. We highlight the approaches to study endocytosis, the impact of endocytosis routes on T-cell response, and discuss the link between how DCs internalize therapeutic antibodies and the potential mechanisms that could give rise to immunogenicity. Understanding these processes could help in developing assays to evaluate the immunogenicity potential of biotherapeutics.
Collapse
Affiliation(s)
- Maria Lteif
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Marc Pallardy
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Isabelle Turbica
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| |
Collapse
|
30
|
Zheng M, Kim MH, Park SG, Kim WS, Oh SH, Sung JH. CXCL12 Neutralizing Antibody Promotes Hair Growth in Androgenic Alopecia and Alopecia Areata. Int J Mol Sci 2024; 25:1705. [PMID: 38338982 PMCID: PMC10855715 DOI: 10.3390/ijms25031705] [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: 12/21/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
We had previously investigated the expression and functional role of C-X-C Motif Chemokine Ligand 12 (CXCL12) during the hair cycle progression. CXCL12 was highly expressed in stromal cells such as dermal fibroblasts (DFs) and inhibition of CXCL12 increased hair growth. Therefore, we further investigated whether a CXCL12 neutralizing antibody (αCXCL12) is effective for androgenic alopecia (AGA) and alopecia areata (AA) and studied the underlying molecular mechanism for treating these diseases. In the AGA model, CXCL12 is highly expressed in DFs. Subcutaneous (s.c.) injection of αCXCL12 significantly induced hair growth in AGA mice, and treatment with αCXCL12 attenuated the androgen-induced hair damage in hair organ culture. Androgens increased the secretion of CXCL12 from DFs through the androgen receptor (AR). Secreted CXCL12 from DFs increased the expression of the AR and C-X-C Motif Chemokine Receptor 4 (CXCR4) in dermal papilla cells (DPCs), which induced hair loss in AGA. Likewise, CXCL12 expression is increased in AA mice, while s.c. injection of αCXCL12 significantly inhibited hair loss in AA mice and reduced the number of CD8+, MHC-I+, and MHC-II+ cells in the skin. In addition, injection of αCXCL12 also prevented the onset of AA and reduced the number of CD8+ cells. Interferon-γ (IFNγ) treatment increased the secretion of CXCL12 from DFs through the signal transducer and activator of transcription 3 (STAT3) pathway, and αCXCL12 treatment protected the hair follicle from IFNγ in hair organ culture. Collectively, these results indicate that CXCL12 is involved in the progression of AGA and AA and antibody therapy for CXCL12 is promising for hair loss treatment.
Collapse
Affiliation(s)
- Mei Zheng
- Epi Biotech Co., Ltd., Incheon 21983, Republic of Korea; (M.Z.); (M.-H.K.)
| | - Min-Ho Kim
- Epi Biotech Co., Ltd., Incheon 21983, Republic of Korea; (M.Z.); (M.-H.K.)
| | - Sang-Gyu Park
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea;
| | - Won-Serk Kim
- Department of Dermatology, School of Medicine, Sungkyunkwan University, Kangbuk Samsung Hospital, Seoul 03181, Republic of Korea;
| | - Sang-Ho Oh
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Jong-Hyuk Sung
- Epi Biotech Co., Ltd., Incheon 21983, Republic of Korea; (M.Z.); (M.-H.K.)
| |
Collapse
|
31
|
Zheng Y, Dou G, Liu S, Meng Z, Tsao EI, Yu G, Zhu X, Gu R, Wu Z, Sun Y, Han P, Gan H. Preclinical Pharmacokinetics and Biodistribution of LR004, a Novel Antiepidermal Growth Factor Receptor Monoclonal Antibody. Molecules 2024; 29:545. [PMID: 38276624 PMCID: PMC10821095 DOI: 10.3390/molecules29020545] [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: 12/08/2023] [Revised: 01/12/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024] Open
Abstract
LR004 is a novel chimeric (human/mouse) monoclonal antibody developed for the treatment of advanced colorectal carcinoma with detectable epidermal growth factor receptor (EGFR) expression. We aimed to investigate the preclinical pharmacokinetics (PK) and in vivo biodistribution of LR004. The PK profiles of LR004 were initially established in rhesus monkeys. Subsequently, 125I radionuclide-labeled LR004 was developed and the biodistribution, autoradiography, and NanoSPECT/CT of 125I-LR004 in xenograft mice bearing A431 tumors were examined. The PK data revealed a prolonged half-life and nonlinear PK characteristics of LR004 within the dose range of 6-54 mg/kg. The radiochemical purity of 125I-LR004 was approximately 98.54%, and iodination of LR004 did not affect its specific binding activity to the EGFR antigen. In a classical biodistribution study, 125I-LR004 exhibited higher uptake in highly perfused organs than in poorly perfused organs. Prolonged retention properties of 125I-LR004 in tumors were observed at 4 and 10 days. Autoradiography and NanoSPECT/CT confirmed the sustained retention of 125I-LR004 at the tumor site in xenograft mice. These findings demonstrated the adequate tumor targeting capabilities of 125I-LR004 in EGFR-positive tumors, which may improve dosing strategies and future drug development.
Collapse
Affiliation(s)
- Ying Zheng
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Guifang Dou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
| | - Shuchen Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
| | - Zhiyun Meng
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
| | - Eric I. Tsao
- Synermore Biologics Co., Ltd., Suzhou 215000, China;
| | - Gang Yu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China;
| | - Xiaoxia Zhu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
| | - Ruolan Gu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
| | - Zhuona Wu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
| | - Yunbo Sun
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
| | - Peng Han
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
| | - Hui Gan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Y.Z.); (G.D.); (S.L.); (Z.M.); (X.Z.); (R.G.); (Z.W.); (Y.S.); (P.H.)
| |
Collapse
|
32
|
Devanaboyina SC, Li P, LaGory EL, Poon-Andersen C, Cook KD, Soto M, Wang Z, Dang K, Uyeda C, Case RB, Thomas VA, Primack R, Ponce M, Di M, Ouyang B, Kaner J, Lam SK, Mostafavi M. Rapid depletion of "catch-and-release" anti-ASGR1 antibody in vivo. MAbs 2024; 16:2383013. [PMID: 39051531 PMCID: PMC11275528 DOI: 10.1080/19420862.2024.2383013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
Targeting antigens with antibodies exhibiting pH/Ca2+-dependent binding against an antigen is an attractive strategy to mitigate target-mediated disposition and antigen buffering. Studies have reported improved serum exposure of antibodies exhibiting pH/Ca2+-binding against membrane-bound receptors. Asialoglycoprotein receptor 1 (ASGR1) is a membrane-bound receptor primarily localized in hepatocytes. With a high expression level of approximately one million receptors per cell, high turnover, and rapid recycling, targeting this receptor with a conventional antibody is a challenge. In this study, we identified an antibody exhibiting pH/Ca2+-dependent binding to ASGR1 and generated antibody variants with increased binding to neonatal crystallizable fragment receptor (FcRn). Serum exposures of the generated anti-ASGR1 antibodies were analyzed in transgenic mice expressing human FcRn. Contrary to published reports of increased serum exposure of pH/Ca2+-dependent antibodies, the pH/Ca2+-dependent anti-ASGR1 antibody had rapid serum clearance in comparison to a conventional anti-ASGR1 antibody. We conducted sub-cellular trafficking studies of the anti-ASGR1 antibodies along with receptor quantification analysis for mechanistic understanding of the rapid serum clearance of pH/Ca2+-dependent anti-ASGR1 antibody. The findings from our study provide valuable insights in identifying the antigens, especially membrane bound, that may benefit from targeting with pH/Ca2+-dependent antibodies to obtain increased serum exposure.
Collapse
Affiliation(s)
- Siva Charan Devanaboyina
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Peng Li
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Edward L. LaGory
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Carrie Poon-Andersen
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Kevin D. Cook
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Marcus Soto
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Zhe Wang
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Khue Dang
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Craig Uyeda
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Ryan B. Case
- Department of Lead Discovery and Characterization, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Veena A. Thomas
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Ronya Primack
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Manuel Ponce
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Mei Di
- Department of Cardiometabolic disorders, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Brian Ouyang
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Joelle Kaner
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Sheung Kwan Lam
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Mina Mostafavi
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| |
Collapse
|
33
|
Boje AS, Pekar L, Koep K, Lipinski B, Rabinovich B, Evers A, Gehlert CL, Krohn S, Xiao Y, Krah S, Zaynagetdinov R, Toleikis L, Poetzsch S, Peipp M, Zielonka S, Klausz K. Impact of antibody architecture and paratope valency on effector functions of bispecific NKp30 x EGFR natural killer cell engagers. MAbs 2024; 16:2315640. [PMID: 38372053 PMCID: PMC10877975 DOI: 10.1080/19420862.2024.2315640] [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: 06/09/2023] [Accepted: 02/02/2024] [Indexed: 02/20/2024] Open
Abstract
Natural killer (NK) cells emerged as a promising effector population that can be harnessed for anti-tumor therapy. In this work, we constructed NK cell engagers (NKCEs) based on NKp30-targeting single domain antibodies (sdAbs) that redirect the cytotoxic potential of NK cells toward epidermal growth factor receptor (EGFR)-expressing tumor cells. We investigated the impact of crucial parameters such as sdAb location, binding valencies, the targeted epitope on NKp30, and the overall antibody architecture on the redirection capacity. Our study exploited two NKp30-specific sdAbs, one of which binds a similar epitope on NKp30 as its natural ligand B7-H6, while the other sdAb addresses a non-competing epitope. For EGFR-positive tumor targeting, humanized antigen-binding domains of therapeutic antibody cetuximab were used. We demonstrate that NKCEs bivalently targeting EGFR and bivalently engaging NKp30 are superior to monovalent NKCEs in promoting NK cell-mediated tumor cell lysis and that the architecture of the NKCE can substantially influence killing capacities depending on the NKp30-targeting sdAb utilized. While having a pronounced impact on NK cell killing efficacy, the capabilities of triggering antibody-dependent cellular phagocytosis or complement-dependent cytotoxicity were not significantly affected comparing the bivalent IgG-like NKCEs with cetuximab. However, the fusion of sdAbs can have a slight impact on the NK cell release of immunomodulatory cytokines, as well as on the pharmacokinetic profile of the NKCE due to unfavorable spatial orientation within the molecule architecture. Ultimately, our findings reveal novel insights for the engineering of potent NKCEs triggering the NKp30 axis.
Collapse
Affiliation(s)
- Ammelie Svea Boje
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Lukas Pekar
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Katharina Koep
- Drug Metabolism and Pharmacokinetics, Merck Healthcare KGaA, Darmstadt, Germany
| | - Britta Lipinski
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Brian Rabinovich
- Department of Oncology and Immuno-Oncology, EMD Serono Research & Development Institute Inc, 45A Middlesex Turnpike, Billerica, MA, USA
| | - Andreas Evers
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Carina Lynn Gehlert
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Steffen Krohn
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Yanping Xiao
- Department of Oncology and Immuno-Oncology, EMD Serono Research & Development Institute Inc, 45A Middlesex Turnpike, Billerica, MA, USA
| | - Simon Krah
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Rinat Zaynagetdinov
- Department of Oncology and Immuno-Oncology, EMD Serono Research & Development Institute Inc, 45A Middlesex Turnpike, Billerica, MA, USA
| | - Lars Toleikis
- Early Protein Supply & Characterization, Merck Healthcare KGaA, Darmstadt, Germany
| | - Sven Poetzsch
- Strategic Innovation, Merck Healthcare KGaA, Darmstadt, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Stefan Zielonka
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| |
Collapse
|
34
|
Baumeister J, Meudt M, Ebert S, Rosenau F, Mizaikoff B, Blech M, Aertker KMJ, Higel F. Decoding the mannose receptor-mAb interaction: the importance of high-mannose N-glycans and glycan-pairing. MAbs 2024; 16:2400414. [PMID: 39245969 PMCID: PMC11385167 DOI: 10.1080/19420862.2024.2400414] [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: 05/27/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024] Open
Abstract
During the development process of therapeutic monoclonal antibodies (mAbs), it is crucial to control (critical) quality attributes such as N-glycosylation influencing pharmacokinetics (PK) and Fc effector functions. Previous reports have shown that mAbs containing high-mannose N-glycans are cleared faster from blood circulation, leading to reduced half-lives. The high-mannose N-glycan content of mAbs can be influenced during the cell culture process by factors such as cell lines, process conditions, and media. Furthermore, mAbs have either one high mannose N-glycan (asymmetrical high-mannose glyco-pair) or two high mannose N-glycans (symmetrical high-mannose glyco-pair). The hypothesis that the mannose receptor (MR, CD206) accelerates clearance by facilitating their internalization and subsequent lysosomal degradation is widespread. However, the interaction between MR and mAbs has not been explicitly demonstrated. This study aimed to investigate this interaction, providing the first systematic demonstration of MR binding to the Fc region of mAbs with high-mannose N-glycans. Two novel analytical methods, MR surface plasmon resonance and MR affinity chromatography, were developed and applied to investigate the MR-mAb interaction. The interaction is found to be dependent on high-mannose content, but is independent of the mAb format or sequence. However, different glyco-pairs exhibited varying binding affinities to the MR, with the symmetrical high-mannose glyco-pair showing the strongest binding properties. These findings strengthen the hypothesis for the MR-mediated mAb interaction and contribute to a deeper understanding of the MR-mAb interaction, which could affect the criticality of high-mannose containing mAbs development strategies of IgG-based molecules and improve their PK profiles.
Collapse
Affiliation(s)
- Julia Baumeister
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- Institute of Pharmaceutical Biotechnology, Ulm University, Ulm, Germany
| | - Maximilian Meudt
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - Sybille Ebert
- Institute of Applied Biotechnology, Biberach University of Applied Sciences, Biberach an der Riss, Germany
| | - Frank Rosenau
- Institute of Pharmaceutical Biotechnology, Ulm University, Ulm, Germany
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - Michaela Blech
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Kristina M J Aertker
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Fabian Higel
- Global CMC Experts NBE, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
35
|
Vu TT, Kim K, Manna M, Thomas J, Remaily BC, Montgomery EJ, Costa T, Granchie L, Xie Z, Guo Y, Chen M, Castillo AMM, Kulp SK, Mo X, Nimmagadda S, Gregorevic P, Owen DH, Ganesan LP, Mace TA, Coss CC, Phelps MA. Decoupling FcRn and tumor contributions to elevated immune checkpoint inhibitor clearance in cancer cachexia. Pharmacol Res 2024; 199:107048. [PMID: 38145833 PMCID: PMC10798214 DOI: 10.1016/j.phrs.2023.107048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 12/27/2023]
Abstract
High baseline clearance of immune checkpoint inhibitors (ICIs), independent of dose or systemic exposure, is associated with cachexia and poor outcomes in cancer patients. Mechanisms linking ICI clearance, cachexia and ICI therapy failure are unknown. Here, we evaluate in four murine models and across multiple antibodies whether altered baseline catabolic clearance of administered antibody requires a tumor and/or cachexia and whether medical reversal of cachexia phenotype can alleviate altered clearance. Key findings include mild cachexia phenotype and lack of elevated pembrolizumab clearance in the MC38 tumor-bearing model. We also observed severe cachexia and decreased, instead of increased, baseline pembrolizumab clearance in the tumor-free cisplatin-induced cachexia model. Liver Fcgrt expression correlated with altered baseline catabolic clearance, though elevated clearance was still observed with antibodies having no (human IgA) or reduced (human H310Q IgG1) FcRn binding. We conclude cachexia phenotype coincides with altered antibody clearance, though tumor presence is neither sufficient nor necessary for altered clearance in immunocompetent mice. Magnitude and direction of clearance alteration correlated with hepatic Fcgrt, suggesting changes in FcRn expression and/or recycling function may be partially responsible, though factors beyond FcRn also contribute to altered clearance in cachexia.
Collapse
Affiliation(s)
- Trang T Vu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Kyeongmin Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Millennium Manna
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Justin Thomas
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Bryan C Remaily
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Emma J Montgomery
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Travis Costa
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Lauren Granchie
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Zhiliang Xie
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Yizhen Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Min Chen
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Alyssa Marie M Castillo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Samuel K Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Xiaokui Mo
- Center for Biostatistics, Ohio State University, Columbus, OH, USA; Pelotonia Institute for Immuno-Oncology, OSUCCC - James, The Ohio State University, Columbus, OH , USA
| | - Sridhar Nimmagadda
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul Gregorevic
- Department of Anatomy & Physiology and Centre for Muscle Research, The University of Melbourne, Parkville, VIC, Australia
| | - Dwight H Owen
- Pelotonia Institute for Immuno-Oncology, OSUCCC - James, The Ohio State University, Columbus, OH , USA; The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Latha P Ganesan
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Thomas A Mace
- Pelotonia Institute for Immuno-Oncology, OSUCCC - James, The Ohio State University, Columbus, OH , USA; The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Christopher C Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Mitch A Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA; Pelotonia Institute for Immuno-Oncology, OSUCCC - James, The Ohio State University, Columbus, OH , USA; The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
36
|
Coliat P, Erb S, Diemer H, Karouby D, Martin T, Banerjee M, Zhu C, Demarchi M, Cianférani S, Detappe A, Pivot X. Influence of pneumatic transportation on the stability of monoclonal antibodies. Sci Rep 2023; 13:21875. [PMID: 38072852 PMCID: PMC10710995 DOI: 10.1038/s41598-023-49235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
Pneumatic transportation systems (PTS) were recently proposed as a method to carry ready-for-injection diluted monoclonal antibodies (mAbs) from the pharmacy to the bedside of patients. This method reduces transportation time and improves the efficiency of drug distribution process. However, mAbs are highly sensitive molecules for which subtle alterations may lead to deleterious clinical effects. These alterations can be caused by various external factors such as temperature, pH, pressure, and mechanical forces that may occur during transportation. Hence, it is essential to ensure that the mAbs transported by PTS remain stable and active throughout the transportation process. This study aims to determine the safety profile of PTS to transport 11 routinely used mAbs in a clinical setting through assessment of critical quality attributes (CQA) and orthogonal analysis. Hence, we performed aggregation/degradation profiling, post-translational modifications identification using complementary mass spectrometry-based methods, along with visible and subvisible particle formation determination by light absorbance and light obscuration analysis. Altogether, these results highlight that PTS can be safely used for this purpose when air is removed from the bags during preparation.
Collapse
Affiliation(s)
- Pierre Coliat
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France.
| | - Stéphane Erb
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
- Institut du Médicament Strasbourg, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI-FR2048, Strasbourg, France
| | - Hélène Diemer
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
- Institut du Médicament Strasbourg, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI-FR2048, Strasbourg, France
| | - Dan Karouby
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Tristan Martin
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Mainak Banerjee
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Chen Zhu
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Martin Demarchi
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Sarah Cianférani
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
- Institut du Médicament Strasbourg, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI-FR2048, Strasbourg, France
| | - Alexandre Detappe
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
- Institut du Médicament Strasbourg, Strasbourg, France
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| |
Collapse
|
37
|
Kang JJ, Ohoka A, Sarkar CA. Designing Multivalent and Multispecific Biologics. Annu Rev Chem Biomol Eng 2023; 15:293-314. [PMID: 38064501 DOI: 10.1146/annurev-chembioeng-100722-112440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
In the era of precision medicine, multivalent and multispecific therapeutics present a promising approach for targeted disease intervention. These therapeutics are designed to interact with multiple targets simultaneously, promising enhanced efficacy, reduced side effects, and resilience against drug resistance. We dissect the principles guiding the design of multivalent biologics, highlighting challenges and strategies that must be considered to maximize therapeutic effect. Engineerable elements in multivalent and multispecific biologic design-domain affinities, valency, and spatial presentation-must be considered in the context of the molecular targets as well as the balance of important properties such as target avidity and specificity. We illuminate recent applications of these principles in designing protein and cell therapies and identify exciting future directions in this field, underscored by advances in biomolecular and cellular engineering and computational approaches. Expected final online publication date for the Annual Review of Chemical and Biomolecular Engineering , Volume 15 is June 2024. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Jennifer J Kang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA; , ,
| | - Ayako Ohoka
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA; , ,
- Present affiliation: AbbVie Inc., North Chicago, Illinois, USA
| | - Casim A Sarkar
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA; , ,
| |
Collapse
|
38
|
Larsen HA, Atkins WM, Nath A. The origins of nonideality exhibited by monoclonal antibodies and Fab fragments in human serum. Protein Sci 2023; 32:e4812. [PMID: 37861473 PMCID: PMC10659951 DOI: 10.1002/pro.4812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/21/2023]
Abstract
The development of therapeutic antibodies remains challenging, time-consuming, and expensive. A key contributing factor is a lack of understanding of how proteins are affected by complex biological environments such as serum and plasma. Nonideality due to attractive or repulsive interactions with cosolutes can alter the stability, aggregation propensity, and binding interactions of proteins in solution. Fluorescence correlation spectroscopy (FCS) can be used to measure apparent second virial coefficient (B2,app ) values for therapeutic and model monoclonal antibodies (mAbs) that capture the nature and strength of interactions with cosolutes directly in undiluted serum and similar complex biological media. Here, we use FCS-derived B2,app measurements to identify the components of human serum responsible for nonideal interactions with mAbs and Fab fragments. Most mAbs exhibit neutral or slightly attractive interactions with intact serum. Generally, mAbs display repulsive interactions with albumin and mildly attractive interactions with IgGs in the context of whole serum. Crucially, however, these attractive interactions are much stronger with pooled IgGs isolated from other serum components, indicating that the effects of serum nonideality can only be understood by studying the intact medium (rather than isolated components). Moreover, Fab fragments universally exhibited more attractive interactions than their parental mAbs, potentially rendering them more susceptible to nonideality-driven perturbations. FCS-based B2,app measurements have the potential to advance our understanding of how physiological environments impact protein-based therapeutics in general. Furthermore, incorporating such assays into preclinical biologics development may help de-risk molecules and make for a faster and more efficient development process.
Collapse
Affiliation(s)
- Hayli A. Larsen
- Department of Medicinal ChemistryUniversity of WashingtonSeattleWashingtonUSA
| | - William M. Atkins
- Department of Medicinal ChemistryUniversity of WashingtonSeattleWashingtonUSA
| | - Abhinav Nath
- Department of Medicinal ChemistryUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
39
|
Marsh MC, Owen SC. Therapeutic Fusion Proteins. AAPS J 2023; 26:3. [PMID: 38036919 DOI: 10.1208/s12248-023-00873-8] [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: 08/17/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
Therapeutic fusion proteins are a class of hybrid constructs that combine distinct biomolecules into a single platform with the additive effects of the components. The ability to fuse two unrelated proteins provides a means to localize mechanisms to better treat a range of diseases. Fusion proteins can be designed to impart diverse functions, including increasing half-life, providing targeting, and enabling sustained signaling. Of these, half-life extenders, which are fused to a therapeutic protein to increase exposure, are the most established group of fusion proteins, with many clinical successes. Rapid advances in antibody and antibody-derivative technology have enabled the fusion of targeting domains with therapeutic proteins. An emerging group of therapeutic fusion proteins has two separate active functions. Although most research for therapeutic fusion proteins focuses on cancer, prior successes provide a foundation for studies into other diseases as well. The exponential emergence of biopharmaceuticals gives precedence for increased research into therapeutic fusion proteins for a multitude of diseases.
Collapse
Affiliation(s)
- Morgan C Marsh
- Department of Molecular Pharmaceutics, University of Utah, 30 South 2000 East, Room 301, Salt Lake City, Utah, 84112, USA
| | - Shawn C Owen
- Department of Molecular Pharmaceutics, University of Utah, 30 South 2000 East, Room 301, Salt Lake City, Utah, 84112, USA.
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, 84112, USA.
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84112, USA.
| |
Collapse
|
40
|
Marin C, Noé G, Schlemmer D, Beaulieu Q, Robidou P, Mansour B, Hirtz C, Vialaret J, Antignac M, Moyon Q, Benameur N, Amoura Z, Zahr N. Determination of plasma concentration of Belimumab by LC-MS/MS: Method development, validation, and clinical application. J Pharm Biomed Anal 2023; 236:115730. [PMID: 37734255 DOI: 10.1016/j.jpba.2023.115730] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/23/2023] [Accepted: 09/16/2023] [Indexed: 09/23/2023]
Abstract
INTRODUCTION Belimumab is a monoclonal antibody against B cell activating factor (BLyS). This monoclonal antibody (mAb) has been shown to be effective in reducing disease activity in patients with systemic lupus erythematosus (SLE). Belimumab is available in two forms as a lyophilized powder for intravenous (IV) use, or single-dose syringe for subcutaneous (SC) use. The aim of this study was to develop and validate a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for quantitation of belimumab in human serum. MATERIAL AND METHODS All analyses relied on nano-surface and molecular-orientation limited (nSMOL) proteolysis coupled with LC-MS/MS. Quantifications was performed in multiple reactions monitoring (MRM) mode, and electrospray ionization was conducted in positive mode. RESULTS Belimumab was quantified with signature peptide QAPGQGLEWMGGIPFGTAK and normalized using P14R. The total run time per assay was 10 min. Linearity was measured from 5 to 800 μg/mL (r² > 0.995). Accuracy and precision based on three quality control levels range from 11.2 - 9.51 % and 1.24 - 13.12 % respectively. The carryover was less than 7 %. In all, 87 patient samples were processed (65, IV; 22, SC). Mean concentration of belimumab was significantly higher for SC (93.0 ± 74.0 µg/mL) than for IV (67.4 ± 38.9 µg/mL) administration. CONCLUSION We have developed the first method of belimumab quantification combining LC-MS/MS and nSMOL proteolysis. It can be used for future clinical pharmacokinetic studies of belimumab and for investigating the relationship between belimumab concentration, efficacy, and toxicity in SLE patients.
Collapse
Affiliation(s)
- Clémence Marin
- AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, Department of Pharmacology, CIC-1901, Pharmacokinetics and Therapeutic Drug Monitoring Unit, UMR-S 1166, F-75013 Paris, France
| | - Gaëlle Noé
- AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, Department of Pharmacology, CIC-1901, Pharmacokinetics and Therapeutic Drug Monitoring Unit, UMR-S 1166, F-75013 Paris, France
| | - Dimitri Schlemmer
- AP-HP. Sorbonne Université, Laboratoire de suivi thérapeutique pharmacologique spécialisé, F-75013 Paris, France
| | - Quentin Beaulieu
- AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, Department of Pharmacology, CIC-1901, Pharmacokinetics and Therapeutic Drug Monitoring Unit, UMR-S 1166, F-75013 Paris, France
| | - Pascal Robidou
- AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, Department of Pharmacology, CIC-1901, Pharmacokinetics and Therapeutic Drug Monitoring Unit, UMR-S 1166, F-75013 Paris, France
| | - Bochra Mansour
- AP-HP. Sorbonne Université, Laboratoire de suivi thérapeutique pharmacologique spécialisé, F-75013 Paris, France
| | - Christophe Hirtz
- IRMB-PPC, INM, Montpellier University Hospital, INSERM, CNRS, University of Montpellier, 34295 Montpellier, France
| | - Jérôme Vialaret
- IRMB-PPC, INM, Montpellier University Hospital, INSERM, CNRS, University of Montpellier, 34295 Montpellier, France
| | - Marie Antignac
- Department of Pharmacy, PITIE-SALPETRIERE Hospital, AP-HP Sorbonne université, F-75013 Paris, France
| | - Quentin Moyon
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des anti-phospholipides et autres maladies auto-immunes rares, Service de Médecine Interne 2, Paris, France
| | - Neila Benameur
- Department of Pharmacy, PITIE-SALPETRIERE Hospital, AP-HP Sorbonne université, F-75013 Paris, France
| | - Zahir Amoura
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des anti-phospholipides et autres maladies auto-immunes rares, Service de Médecine Interne 2, Paris, France
| | - Noël Zahr
- AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, Department of Pharmacology, CIC-1901, Pharmacokinetics and Therapeutic Drug Monitoring Unit, UMR-S 1166, F-75013 Paris, France; AP-HP. Sorbonne Université, Laboratoire de suivi thérapeutique pharmacologique spécialisé, F-75013 Paris, France.
| |
Collapse
|
41
|
Xing P, Meng B, Hu X, Qu W, Wang S. Switching to Conbercept in Diabetic Macular Edema After Unsatisfactory Response to Previous Intravitreal Injection of Ranibizumab. Clin Ophthalmol 2023; 17:3491-3497. [PMID: 38026602 PMCID: PMC10661898 DOI: 10.2147/opth.s431145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Objective To assess the functional and anatomical effects of transitioning to conbercept intravitreal injection (IVC) treatment in patients with diabetic macular edema (DME) who had inadequate responses to prior anti-vascular endothelial growth factor (anti-VEGF) injections. Methods We retrospectively included eyes with persistent DME after at least 3 injections of intravitreal ranibizumab (IVR). The analysis included the assessment of best corrected visual acuity (BCVA) and central macular thickness (CMT) during 6 months after the switch. Results A total of 30 patients (30 eyes) were included. CMT dropped sharply from 437.8±40.67μm at baseline to 363.59±45.09,312.52 ± 39.15, 278.51 ± 37.92, and 292.59 ± 38.09 after 1, 2, 3 and 6 months of IVC, respectively (p <0.001). BCVA in log MAR units was significantly improved from 0.73±0.15 at baseline to 0.50±0.09,0.46±0.72, 0.40±0.06 and 0.48±0.04 after 1, 2, 3 and 6 months, respectively (p <0.001). Conclusion Switching to Conbercept effectively improved visual and anatomical structure in DME patients who had not responded satisfactorily to previous anti-VEGF injections.
Collapse
Affiliation(s)
- Peiyu Xing
- Department of Ophthalmology, China Medical University the Fourth People’s Hospital of Shenyang, Shenyang, People’s Republic of China
| | - Bo Meng
- Department of Ophthalmology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xiaojia Hu
- Department of Ophthalmology, China Medical University the Fourth People’s Hospital of Shenyang, Shenyang, People’s Republic of China
| | - Wei Qu
- Department of Ophthalmology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Shaowei Wang
- Department of Ophthalmology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
42
|
Zehnaker A, Vallet A, Gourdon J, Sarti C, Jugnarain V, Haj Hassan M, Mathias L, Gauthier C, Raynaud P, Boulo T, Beauclair L, Bigot Y, Casarini L, Crépieux P, Poupon A, Piégu B, Jean-Alphonse F, Bruneau G, Reiter É. Combined Multiplexed Phage Display, High-Throughput Sequencing, and Functional Assays as a Platform for Identifying Modulatory VHHs Targeting the FSHR. Int J Mol Sci 2023; 24:15961. [PMID: 37958944 PMCID: PMC10650796 DOI: 10.3390/ijms242115961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Developing modulatory antibodies against G protein-coupled receptors is challenging. In this study, we targeted the follicle-stimulating hormone receptor (FSHR), a significant regulator of reproduction, with variable domains of heavy chain-only antibodies (VHHs). We built two immune VHH libraries and submitted them to multiplexed phage display approaches. We used next-generation sequencing to identify 34 clusters of specifically enriched sequences that were functionally assessed in a primary screen based on a cAMP response element (CRE)-dependent reporter gene assay. In this assay, 23 VHHs displayed negative or positive modulation of FSH-induced responses, suggesting a high success rate of the multiplexed strategy. We then focused on the largest cluster identified (i.e., PRC1) that displayed positive modulation of FSH action. We demonstrated that PRC1 specifically binds to the human FSHR and human FSHR/FSH complex while potentiating FSH-induced cAMP production and Gs recruitment. We conclude that the improved selection strategy reported here is effective for rapidly identifying functionally active VHHs and could be adapted to target other challenging membrane receptors. This study also led to the identification of PRC1, the first potential positive modulator VHH reported for the human FSHR.
Collapse
Affiliation(s)
- Anielka Zehnaker
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Amandine Vallet
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Juliette Gourdon
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Caterina Sarti
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Vinesh Jugnarain
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Maya Haj Hassan
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Laetitia Mathias
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Camille Gauthier
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Pauline Raynaud
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Thomas Boulo
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Linda Beauclair
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Yves Bigot
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Livio Casarini
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Pascale Crépieux
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
| | - Anne Poupon
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
- MAbSilico, 1 Impasse du Palais, 37000 Tours, France
| | - Benoît Piégu
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Frédéric Jean-Alphonse
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
| | - Gilles Bruneau
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Éric Reiter
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
| |
Collapse
|
43
|
LaManna L, Chou CH, Lei H, Barton ER, Maliga P. Chloroplast transformation for bioencapsulation and oral delivery using the immunoglobulin G fragment crystallizable (Fc) domain. Sci Rep 2023; 13:18916. [PMID: 37919321 PMCID: PMC10622566 DOI: 10.1038/s41598-023-45698-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
Proinsulin Like Growth Factor I (prolGF-I) and myostatin (Mstn) regulate muscle regeneration and mass when intravenously delivered. We tested if chloroplast bioencapsulated forms of these proteins may serve as a non-invasive means of drug delivery through the digestive system. We created tobacco (Nicotiana tabacum) plants carrying GFP-Fc1, proIGF-I-Fc1, and Mstn-Fc1 fusion genes, in which fusion with the immunoglobulin G Fc domain improved both protein stability and absorption in the small intestine. No transplastomic plants were obtained with the Mstn-Fc1 gene, suggesting that the protein is toxic to plant cells. proIGF-I-Fc1 protein levels were too low to enable in vivo testing. However, GFP-Fc1 accumulated at a high level, enabling evaluation of chloroplast-made Fc fusion proteins for oral delivery. Tobacco leaves were lyophilized for testing in a mouse system. We report that the orally administered GFP-Fc1 fusion protein (5.45 µg/g GFP-Fc1) has been taken up by the intestinal epithelium cells, evidenced by confocal microscopy. GFP-Fc1 subsequently entered the circulation where it was detected by ELISA. Data reported here confirm that chloroplast expression and oral administration of lyophilized leaves is a potential delivery system of therapeutic proteins fused with Fc1, with the advantage that the proteins may be stored at room temperature.
Collapse
Affiliation(s)
- Lisa LaManna
- Waksman Institute of Microbiology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Chih-Hsuan Chou
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611, USA
| | - Hanqin Lei
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611, USA
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611, USA.
| | - Pal Maliga
- Waksman Institute of Microbiology, Rutgers University, Piscataway, NJ, 08854, USA.
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
44
|
Nguyen TTK, Pham KY, Yook S. Engineered therapeutic proteins for sustained-release drug delivery systems. Acta Biomater 2023; 171:131-154. [PMID: 37717712 DOI: 10.1016/j.actbio.2023.09.018] [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: 03/01/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
Proteins play a vital role in diverse biological processes in the human body, and protein therapeutics have been applied to treat different diseases such as cancers, genetic disorders, autoimmunity, and inflammation. Protein therapeutics have demonstrated their advantages, such as specific pharmaceutical effects, low toxicity, and strong solubility. However, several disadvantages arise in clinical applications, including short half-life, immunogenicity, and low permeation, leading to reduced drug effectiveness. The structure of protein therapeutics can be modified to increase molecular size, leading to prolonged stability and increased plasma half-life. Notably, the controlled-release delivery systems for the sustained release of protein drugs and preserving the stability of cargo proteins are envisioned as a potential approach to overcome these challenges. In this review, we summarize recent research progress related to structural modifications (PEGylation, glycosylation, poly amino acid modification, and molecular biology-based strategies) and promising long-term delivery systems, such as polymer-based systems (injectable gel/implants, microparticles, nanoparticles, micro/nanogels, functional polymers), lipid-based systems (liposomes, solid lipid nanoparticles, nanostructured lipid carriers), and inorganic nanoparticles exploited for protein therapeutics. STATEMENT OF SIGNIFICANCE: In this review, we highlight recent advances concerning modifying proteins directly to enhance their stability and functionality and discuss state-of-the-art methods for the delivery and controlled long-term release of active protein therapeutics to their target site. In terms of drug modifications, four widely used strategies, including PEGylation, poly amino acid modification, glycosylation, and genetic, are discussed. As for drug delivery systems, we emphasize recent progress relating to polymer-based systems, lipid-based systems developed, and inorganic nanoparticles for protein sustained-release delivery. This review points out the areas requiring focused research attention before the full potential of protein therapeutics for human health and disease can be realized.
Collapse
Affiliation(s)
- Thoa Thi Kim Nguyen
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea
| | - Khang-Yen Pham
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
45
|
Dadas O, Allen JD, Buchan SL, Kim J, Chan HTC, Mockridge CI, Duriez PJ, Rogel A, Crispin M, Al-Shamkhani A. Fcγ receptor binding is required for maximal immunostimulation by CD70-Fc. Front Immunol 2023; 14:1252274. [PMID: 37965342 PMCID: PMC10641686 DOI: 10.3389/fimmu.2023.1252274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction T cell expressed CD27 provides costimulation upon binding to inducible membrane expressed trimeric CD70 and is required for protective CD8 T cell responses. CD27 agonists could therefore be used to bolster cellular vaccines and anti-tumour immune responses. To date, clinical development of CD27 agonists has focussed on anti-CD27 antibodies with little attention given to alternative approaches. Methods Here, we describe the generation and activity of soluble variants of CD70 that form either trimeric (t) or dimer-of-trimer proteins and conduct side-by-side comparisons with an agonist anti-CD27 antibody. To generate a dimer-of-trimer protein (dt), we fused three extracellular domains of CD70 to the Fc domain of mouse IgG1 in a 'string of beads' configuration (dtCD70-Fc). Results Whereas tCD70 failed to costimulate CD8 T cells, both dtCD70-Fc and an agonist anti-CD27 antibody were capable of enhancing T cell proliferation in vitro. Initial studies demonstrated that dtCD70-Fc was less efficacious than anti-CD27 in boosting a CD8 T cell vaccine response in vivo, concomitant with rapid clearance of dtCD70-Fc from the circulation. The accelerated plasma clearance of dtCD70-Fc was not due to the lack of neonatal Fc receptor binding but was dependent on the large population of oligomannose type glycosylation. Enzymatic treatment to reduce the oligomannose-type glycans in dtCD70-Fc improved its half-life and significantly enhanced its T cell stimulatory activity in vivo surpassing that of anti-CD27 antibody. We also show that whereas the ability of the anti-CD27 to boost a vaccine response was abolished in Fc gamma receptor (FcγR)-deficient mice, dtCD70-Fc remained active. By comparing the activity of dtCD70-Fc with a variant (dtCD70-Fc(D265A)) that lacks binding to FcγRs, we unexpectedly found that FcγR binding to dtCD70-Fc was required for maximal boosting of a CD8 T cell response in vivo. Interestingly, both dtCD70-Fc and dtCD70-Fc(D265A) were effective in prolonging the survival of mice harbouring BCL1 B cell lymphoma, demonstrating that a substantial part of the stimulatory activity of dtCD70-Fc in this setting is retained in the absence of FcγR interaction. Discussion These data reveal that TNFRSF ligands can be generated with a tunable activity profile and suggest that this class of immune agonists could have broad applications in immunotherapy.
Collapse
Affiliation(s)
- Osman Dadas
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, European University of Lefke, Lefke, Cyprus
| | - Joel D. Allen
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Sarah L. Buchan
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jinny Kim
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - H. T. Claude Chan
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - C. Ian Mockridge
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Patrick J. Duriez
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Anne Rogel
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Max Crispin
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Aymen Al-Shamkhani
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
46
|
Yoshikawa M, Nakamura H, Oda-Ueda N, Ohkuri T. Analysis of thermostability for seven Phe to Ala and six Pro to Gly mutants in the Fab constant region of adalimumab. J Biochem 2023; 174:345-353. [PMID: 37390406 DOI: 10.1093/jb/mvad047] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023] Open
Abstract
To identify amino acids that play important roles in the structural stability of Fab, seven phenylalanine residues in the Fab constant region of the therapeutic antibody adalimumab were subjected to alanine mutagenesis. Six Fab mutants, H:F130A, H:F154A, H:F174A, L:F118A, L:F139A and L:F209A, showed decreased thermostability compared with wild-type Fab. In contrast, the Tm for the L:F116A mutant was 1.7°C higher than that of wild-type Fab, indicating that the F116 residue was unfavorable for Fab thermostability. Six proline mutants, H:P131G, H:P155G, H:P175G, L:P119G, L:P120G and L:P141G, were also prepared to investigate the effect of proline residues adjacent to mutated phenylalanine residues. The thermostability of the H:P155G and L:P141G mutants in particular was significantly reduced, with decreases in Tm of 5.0 and 3.0°C, respectively, compared with wild-type Fab. The H:P155 and L:P141 residues have a cis conformation, whereas the other mutated proline residues have a trans conformation. H:P155 and L:P141 had stacking interactions with the H:F154 and L:Y140, respectively, at the interface between the variable and constant regions. It is suggested that the interactions of the aromatic ring with a cis-form proline at the interface between the variable and constant regions is important for stability of Fab.
Collapse
Affiliation(s)
- Moeka Yoshikawa
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Hitomi Nakamura
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Naoko Oda-Ueda
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Takatoshi Ohkuri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| |
Collapse
|
47
|
Abdeldaim DT, Schindowski K. Fc-Engineered Therapeutic Antibodies: Recent Advances and Future Directions. Pharmaceutics 2023; 15:2402. [PMID: 37896162 PMCID: PMC10610324 DOI: 10.3390/pharmaceutics15102402] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Monoclonal therapeutic antibodies have revolutionized the treatment of cancer and other diseases. Fc engineering aims to enhance the effector functions or half-life of therapeutic antibodies by modifying their Fc regions. Recent advances in the Fc engineering of modern therapeutic antibodies can be considered the next generation of antibody therapy. Various strategies are employed, including altering glycosylation patterns via glycoengineering and introducing mutations to the Fc region, thereby enhancing Fc receptor or complement interactions. Further, Fc engineering strategies enable the generation of bispecific IgG-based heterodimeric antibodies. As Fc engineering techniques continue to evolve, an expanding portfolio of Fc-engineered antibodies is advancing through clinical development, with several already approved for medical use. Despite the plethora of Fc-based mutations that have been analyzed in in vitro and in vivo models, we focus here in this review on the relevant Fc engineering strategies of approved therapeutic antibodies to finetune effector functions, to modify half-life and to stabilize asymmetric bispecific IgGs.
Collapse
Affiliation(s)
- Dalia T. Abdeldaim
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
| |
Collapse
|
48
|
Thran M, Pönisch M, Danz H, Horscroft N, Ichtchenko K, Tzipori S, Shoemaker CB. Co-administration of an effector antibody enhances the half-life and therapeutic potential of RNA-encoded nanobodies. Sci Rep 2023; 13:14632. [PMID: 37670025 PMCID: PMC10480410 DOI: 10.1038/s41598-023-41092-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
The incidence of Clostridioides difficile infection (CDI) and associated mortality have increased rapidly worldwide in recent years. Therefore, it is critical to develop new therapies for CDI. Here we report on the development of mRNA-LNPs encoding camelid-derived VHH-based neutralizing agents (VNAs) targeting toxins A and/or B of C. difficile. In preclinical models, intravenous administration of the mRNA-LNPs provided serum VNA levels sufficient to confer protection of mice against severe disease progression following toxin challenge. Furthermore, we employed an mRNA-LNP encoded effector antibody, a molecular tool designed to specifically bind an epitopic tag linked to the VNAs, to prolong VNA serum half-life. Co-administration of VNA-encoding mRNA-LNPs and an effector antibody, either provided as recombinant protein or encoded by mRNA-LNP, increased serum VNA half-life in mice and in gnotobiotic piglets. Prolonged serum half-life was associated with higher concentrations of serum VNA and enhanced prophylactic protection of mice in challenge models.
Collapse
Affiliation(s)
| | | | - Hillary Danz
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA
| | | | - Konstantin Ichtchenko
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA
| | - Charles B Shoemaker
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA.
| |
Collapse
|
49
|
Lan Z, Lv Z, Zuo W, Xiao Y. From bench to bedside: The promise of sotatercept in hematologic disorders. Biomed Pharmacother 2023; 165:115239. [PMID: 37516019 DOI: 10.1016/j.biopha.2023.115239] [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: 05/30/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023] Open
Abstract
Sotatercept (ACE-011) is an activin receptor IIA-Fc (ActRIIA-Fc) fusion protein currently under investigation for its potential in the treatment of hematologic diseases. By impeding the activities of the overexpressed growth and differentiation factor 11 (GDF11), activin A, and other members of the transforming growth factor-β (TGF-β) superfamily, commonly found in hematologic disorders, sotatercept aims to restore the normal functioning of red blood cell maturation and osteoblast differentiation. This action is anticipated to enhance anemia management and hinder the progression of myeloma. Simultaneously, comprehensive research is ongoing to investigate sotatercept's pharmacokinetics and potential adverse reactions, thus laying a robust foundation for its prospective clinical use. In this review, we provide a detailed overview of TGF-β pathways in physiological and hematologic disorder contexts, outline the potential mechanism of sotatercept, and delve into its pharmacokinetics and clinical research advancements in various hematologic diseases. A particular emphasis is given to the relationship between sotatercept dosage and its efficacy or associated adverse reactions.
Collapse
Affiliation(s)
- Zehao Lan
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha 410011, China; Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Zhaohua Lv
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha 410011, China; Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Wanyun Zuo
- Department of Hematology, Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yichao Xiao
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha 410011, China.
| |
Collapse
|
50
|
Prabaharan CB, Giri S, Allen KJH, Bato KEM, Mercado TR, Malo ME, Carvalho JLC, Dadachova E, Uppalapati M. Comparative Molecular Characterization and Pharmacokinetics of IgG1-Fc and Engineered Fc Human Antibody Variants to Insulin-like Growth Factor 2 Receptor (IGF2R). Molecules 2023; 28:5839. [PMID: 37570809 PMCID: PMC10420659 DOI: 10.3390/molecules28155839] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Novel therapeutic approaches are much needed for the treatment of osteosarcoma. Targeted radionuclide therapy (TRT) and radioimmunotherapy (RIT) are promising approaches that deliver therapeutic radiation precisely to the tumor site. We have previously developed a fully human antibody, named IF3, that binds to insulin-like growth factor 2 receptor (IGF2R). IF3 was used in TRT to effectively inhibit tumor growth in osteosarcoma preclinical models. However, IF3's relatively short half-life in mice raised the need for improvement. We generated an Fc-engineered version of IF3, termed IF3δ, with amino acid substitutions known to enhance antibody half-life in human serum. In this study, we confirmed the specific binding of IF3δ to IGF2R with nanomolar affinity, similar to wild-type IF3. Additionally, IF3δ demonstrated binding to human and mouse neonatal Fc receptors (FcRn), indicating the potential for FcRn-mediated endocytosis and recycling. Biodistribution studies in mice showed a higher accumulation of IF3δ in the spleen and bone than wild-type IF3, likely attributed to abnormal spleen expression of IGF2R in mice. Therefore, the pharmacokinetics data from mouse xenograft models may not precisely reflect their behavior in canine and human patients. However, the findings suggest both IF3 and IF3δ as promising options for the RIT of osteosarcoma.
Collapse
Affiliation(s)
- Chandra B. Prabaharan
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada;
| | - Sabeena Giri
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.G.); (K.J.H.A.); (M.E.M.); (J.L.C.C.)
| | - Kevin J. H. Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.G.); (K.J.H.A.); (M.E.M.); (J.L.C.C.)
| | - Katrina E. M. Bato
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (K.E.M.B.); (T.R.M.)
| | - Therese R. Mercado
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (K.E.M.B.); (T.R.M.)
| | - Mackenzie E. Malo
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.G.); (K.J.H.A.); (M.E.M.); (J.L.C.C.)
| | - Jorge L. C. Carvalho
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.G.); (K.J.H.A.); (M.E.M.); (J.L.C.C.)
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.G.); (K.J.H.A.); (M.E.M.); (J.L.C.C.)
| | - Maruti Uppalapati
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada;
| |
Collapse
|