1
|
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
|
2
|
Mehrizi TZ, Mirzaei M, Ardestani MS. Pegylation, a Successful Strategy to Address the Storage and Instability Problems of Blood Products: Review 2011-2021. Curr Pharm Biotechnol 2024; 25:247-267. [PMID: 37218184 DOI: 10.2174/1389201024666230522091958] [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: 08/02/2022] [Revised: 01/24/2023] [Accepted: 02/06/2023] [Indexed: 05/24/2023]
Abstract
Conjugation of polyethylene glycol (PEGylation) to blood proteins and cells has emerged as a successful approach to address some of the issues attributed to the storage of blood products, including their short half-life and instability. In this regard, this review study aims to compare the influence of different PEGylation strategies on the quality of several blood products like red blood cells (RBCs), platelets, plasma proteins, i.e., albumin, coagulation factor VIII, and antibodies. The results indicated that conjugating succinimidyl carbonate methoxyPEG (SCmPEG) to platelets could improve blood transfusion safety by preventing these cells from being attached to low-load hidden bacteria in blood products. Moreover, coating of 20 kD succinimidyl valerate (SVA)-mPEG to RBCs was able to extend the half-life and stability of these cells during storage, as well as immune camouflage their surface antigens to prevent alloimmunisation. As regards albumin products, PEGylation improved the albumin stability, especially during sterilization, and there was a relationship between the molecular weight (MW) of PEG molecules and the biological half-life of the conjugate. Although coating antibodies with short-chain PEG molecules could enhance their stabilities, these modified proteins were cleared from the blood faster. Also, branched PEG molecules enhanced the retention and shielding of the fragmented and bispecific antibodies. Overall, the results of this literature review indicate that PEGylation can be considered a useful tool for enhancing the stability and storage of blood components.
Collapse
Affiliation(s)
| | - Mehdi Mirzaei
- Iran Ministry of Health and Medical Education, Deputy Ministry for Education, Tehran, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Zadeh Mehrizi T, Mousavi Hosseini K. An overview on the investigation of nanomaterials' effect on plasma components: immunoglobulins and coagulation factor VIII, 2010-2020 review. NANOSCALE ADVANCES 2021; 3:3730-3745. [PMID: 36133015 PMCID: PMC9419877 DOI: 10.1039/d1na00119a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/16/2021] [Indexed: 05/04/2023]
Abstract
FVIII and immunoglobulins (Igs) are the most prominent plasma proteins, which play a vital role in plasma hemostasis. These proteins have been implemented frequently in protein therapy. Therefore, their maintenance, durability, and stability are highly essential. Herein, various approaches to improve protein functions have been investigated, such as using recombinant protein replacement. In comparison, advances in nanotechnology have provided adequate context to boost biomaterial utilization. In this regard, the applications of various nanoparticles such as polymeric nanomaterials (PEG and PLGA), metal nanoparticles, dendrimers, and lipid based nanomaterials (liposomes and lipid nanoparticles) in stability and the functional improvement of antibodies and coagulation factor VIII (FVIII) have been reviewed from 2010 to 2020. Reviewing related articles has shown that not only can nanomaterials adequately protect the structure of proteins, but have also improved proteins' functions in some cases. For example, the high rate of FVIII instability has been successfully enhanced by bio-PEGylation. Also, utilizing PEGylated liposomes, using the PEG-lip technique for coating nanostructures, leads to FIIIV half-life prolongation. Hence, PEGylation had most impact on the stability of FVIII. Likewise, PEG-coated liposome nano-carriers also presented such a good effect on stability improvements for FVIII due to their ability to tune the immune system by reducing FVIII immunogenicity. Similarly, Ig PEGylation and conjugation to magnetic nanoparticles resulted in increased half-life and better purification of Igs, respectively, without any loss in structural or functional features. Consequently, metal-organic frameworks and recent hybrid systems have been introduced as promising nanomaterials in biomedical applications. As far as we know, this is the first study in this field, which considers the applications of nanoparticles for improving the storage and stability of antibodies and coagulation FVIII.
Collapse
Affiliation(s)
- Tahereh Zadeh Mehrizi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine Tehran Iran +989338606292
| | - Kamran Mousavi Hosseini
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine Tehran Iran +989338606292
| |
Collapse
|
4
|
Hay CE, Ewing LE, Hambuchen MD, Zintner SM, Small JC, Bolden CT, Fantegrossi WE, Margaritis P, Owens SM, Peterson EC. The Development and Characterization of an scFv-Fc Fusion-Based Gene Therapy to Reduce the Psychostimulant Effects of Methamphetamine Abuse. J Pharmacol Exp Ther 2020; 374:16-23. [PMID: 32245884 DOI: 10.1124/jpet.119.261180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 03/31/2020] [Indexed: 12/17/2022] Open
Abstract
Methamphetamine (METH) continues to be among the most addictive and abused drugs in the United States. Unfortunately, there are currently no Food and Drug Administration-approved pharmacological treatments for METH-use disorder. We have previously explored the use of adeno-associated viral (AAV)-mediated gene transfer of an anti-METH monoclonal antibody. Here, we advance our approach by generating a novel anti-METH single-chain variable fragment (scFv)-Fc fusion construct (termed 7F9-Fc) packaged into AAV serotype 8 vector (called AAV-scFv-Fc) and tested in vivo and ex vivo. A range of doses [1 × 1010, 1 × 1011, and 1 × 1012 vector copies (vcs)/mouse] were administered to mice, eliciting a dose-dependent expression of 7F9-Fc in serum with peak circulating concentrations of 48, 1785, and 3831 µg/ml, respectively. Expressed 7F9-Fc exhibited high-affinity METH binding, IC50 = 17 nM. Between days 21 and 35 after vector administration, at both 1 × 1011 vc/mouse and 1 × 1012 vc/mouse doses, the AAV-7F9-Fc gene therapy significantly decreased the potency of METH in locomotor assays. On day 116 post-AAV administration, mice expressing 7F9-Fc sequestered over 2.5 times more METH in the serum than vehicle-treated mice, and METH concentrations in the brain were reduced by 1.2 times the value for vehicle mice. These data suggest that an AAV-delivered anti-METH Fc fusion antibody could be used to persistently reduce concentrations of METH in the central nervous system. SIGNIFICANCE STATEMENT: In this manuscript, we describe the testing of a novel antimethamphetamine (METH) single-chain variable fragment-Fc fusion protein delivered in mice using gene therapy. The results suggest that the gene therapy delivery system can lead to the production of significant antibody concentrations that mitigate METH's psychostimulant effects in mice over an extended time period.
Collapse
Affiliation(s)
- Charles E Hay
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| | - Laura E Ewing
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| | - Michael D Hambuchen
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| | - Shannon M Zintner
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| | - Juliana C Small
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| | - Chris T Bolden
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| | - William E Fantegrossi
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| | - Paris Margaritis
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| | - S Michael Owens
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| | - Eric C Peterson
- University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.E.H., L.E.E., M.D.H., C.T.B., W.E.F., S.M.O., E.C.P,); The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (S.M.Z., J.C.S., P.M.,); The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (P.M.); and Department of Pediatrics, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania (P.M.)
| |
Collapse
|
6
|
Pan H, Liu J, Deng W, Xing J, Li Q, Wang Z. Site-specific PEGylation of an anti-CEA/CD3 bispecific antibody improves its antitumor efficacy. Int J Nanomedicine 2018; 13:3189-3201. [PMID: 29881272 PMCID: PMC5985803 DOI: 10.2147/ijn.s164542] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Introduction Bispecific antibodies that engage immune cells to kill cancer cells are actively pursued in cancer immunotherapy. Different types of bispecific antibodies, including single-chain fragments, Fab fragments, nanobodies, and immunoglobulin Gs (IgGs), have been studied. However, the low molecular weight of bispecific antibodies with single-chain or Fab fragments generally leads to their rapid clearance in vivo, which limits the therapeutic potential of these bispecific antibodies. Materials and methods In this study, we used a site-specific PEGylation strategy to modify the bispecific single-domain antibody-linked Fab (S-Fab), which was designed by linking an anticarcinoembryonic antigen (anti-CEA) nanobody with an anti-CD3 Fab. Results The half-life (t1/2) of PEGylated S-Fab (polyethylene glycol-S-Fab) was increased 12-fold in vivo with a slightly decreased tumor cell cytotoxicity in vitro as well as more potent tumor growth inhibition in vivo compared to S-Fab. Conclusion This study demonstrated that PEGylation is an effective approach to enhance the antitumor efficacy of bispecific antibodies.
Collapse
Affiliation(s)
- Haitao Pan
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jiayu Liu
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Wentong Deng
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jieyu Xing
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Qing Li
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zhong Wang
- School of Pharmaceutical Sciences.,Centre for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|