1
|
Gaballa SA, Shimizu T, Ando H, Takata H, Emam SE, Ramadan E, Naguib YW, Mady FM, Khaled KA, Ishida T. Treatment-induced and Pre-existing Anti-peg Antibodies: Prevalence, Clinical Implications, and Future Perspectives. J Pharm Sci 2024; 113:555-578. [PMID: 37931786 DOI: 10.1016/j.xphs.2023.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Polyethylene glycol (PEG) is a versatile polymer that is used in numerous pharmaceutical applications like the food industry, a wide range of disinfectants, cosmetics, and many commonly used household products. PEGylation is the term used to describe the covalent attachment of PEG molecules to nanocarriers, proteins and peptides, and it is used to prolong the circulation half-life of the PEGylated products. Consequently, PEGylation improves the efficacy of PEGylated therapeutics. However, after four decades of research and more than two decades of clinical applications, an unappealing side of PEGylation has emerged. PEG immunogenicity and antigenicity are remarkable challenges that confound the widespread clinical application of PEGylated therapeutics - even those under clinical trials - as anti-PEG antibodies (Abs) are commonly reported following the systemic administration of PEGylated therapeutics. Furthermore, pre-existing anti-PEG Abs have also been reported in healthy individuals who have never been treated with PEGylated therapeutics. The circulating anti-PEG Abs, both treatment-induced and pre-existing, selectively bind to PEG molecules of the administered PEGylated therapeutics inducing activation of the complement system, which results in remarkable clinical implications with varying severity. These include increased blood clearance of the administered PEGylated therapeutics through what is known as the accelerated blood clearance (ABC) phenomenon and initiation of serious adverse effects through complement activation-related pseudoallergic reactions (CARPA). Therefore, the US FDA industry guidelines have recommended the screening of anti-PEG Abs, in addition to Abs against PEGylated proteins, in the clinical trials of PEGylated protein therapeutics. In addition, strategies revoking the immunogenic response against PEGylated therapeutics without compromising their therapeutic efficacy are important for the further development of advanced PEGylated therapeutics and drug-delivery systems.
Collapse
Affiliation(s)
- Sherif A Gaballa
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Sherif E Emam
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519 Egypt
| | - Eslam Ramadan
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Youssef W Naguib
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Fatma M Mady
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Khaled A Khaled
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan.
| |
Collapse
|
2
|
Chen H, Zhang Q. Polypeptides as alternatives to PEGylation of therapeutic agents. Expert Opin Drug Deliv 2024; 21:1-12. [PMID: 38116624 DOI: 10.1080/17425247.2023.2297937] [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: 10/05/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION Due to the concerns raised by the extensive application of PEGylation, polypeptides have stood out as excellent candidates with adequate biocompatibility and biodegradability with tunable hydrophilicity. AREAS COVERED In this review, polypeptides with the potential to replace PEGylation have been summarized and their application has been reviewed, including XTEN, PASylation, polysarcosine, zwitterion polypeptides, ELPylation, etc. Besides their strengths, the remaining challenges have also been discussed and the future perspectives have been provided. EXPERT OPINION Polypeptides have been applied in the designing of peptide/protein drugs as well as nanomedicines, and some of the pharmaceutics have made it into the clinical trials and got approved. These polypeptides showed similar hydrophilic properties to PEGylation, which increased the hydrodynamic volumes of protein drugs, reduced kidney elimination, decreased protein-polymer interaction and potentially improved the drug delivery efficiency due to the extended circulation time in the system. Moreover, they demonstrated superior biodegradability and biocompatibility, compensating for the deficiencies for polymers such as PEG.
Collapse
Affiliation(s)
- Huali Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Qianyu Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Ji Y, Liu D, Zhu H, Bao L, Chang R, Gao X, Yin J. Unstructured Polypeptides as a Versatile Drug Delivery Technology. Acta Biomater 2023; 164:74-93. [PMID: 37075961 DOI: 10.1016/j.actbio.2023.04.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/23/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Although polyethylene glycol (PEG), or "PEGylation" has become a widely applied approach for improving the efficiency of drug delivery, the immunogenicity and non-biodegradability of this synthetic polymer have prompted an evident need for alternatives. To overcome these caveats and to mimic PEG -or other natural or synthetic polymers- for the purpose of drug half-life extension, unstructured polypeptides are designed. Due to their tunable length, biodegradability, low immunogenicity and easy production, unstructured polypeptides have the potential to replace PEG as the preferred technology for therapeutic protein/peptide delivery. This review provides an overview of the evolution of unstructured polypeptides, starting from natural polypeptides to engineered polypeptides and discusses their characteristics. Then, it is described that unstructured polypeptides have been successfully applied to numerous drugs, including peptides, proteins, antibody fragments, and nanocarriers, for half-life extension. Innovative applications of unstructured peptides as releasable masks, multimolecular adaptors and intracellular delivery carriers are also discussed. Finally, challenges and future perspectives of this promising field are briefly presented. STATEMENT OF SIGNIFICANCE: : Polypeptide fusion technology simulating PEGylation has become an important topic for the development of long-circulating peptide or protein drugs without reduced activity, complex processes, and kidney injury caused by PEG modification. Here we provide a detailed and in-depth review of the recent advances in unstructured polypeptides. In addition to the application of enhanced pharmacokinetic performance, emphasis is placed on polypeptides as scaffolders for the delivery of multiple drugs, and on the preparation of reasonably designed polypeptides to manipulate the performance of proteins and peptides. This review will provide insight into future application of polypeptides in peptide or protein drug development and the design of novel functional polypeptides.
Collapse
Affiliation(s)
- Yue Ji
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Dingkang Liu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Haichao Zhu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Lichen Bao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 210009, China
| | - Ruilong Chang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Jun Yin
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
4
|
PASylation improves pharmacokinetic of liposomes and attenuates anti-PEG IgM production: An alternative to PEGylation. NANOMEDICINE: NANOTECHNOLOGY, BIOLOGY AND MEDICINE 2023; 47:102622. [DOI: 10.1016/j.nano.2022.102622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/22/2022] [Accepted: 10/15/2022] [Indexed: 12/12/2022]
|
5
|
Zheng Q, Duan D, Xu J, Wang X, Ge Y, Xiong L, Yang J, Wulayin S, Luo X. Comparative safety of multiple doses of erythropoietin for the treatment of traumatic brain injury: A systematic review and network meta-analysis. Front Neurol 2022; 13:998320. [PMID: 36582613 PMCID: PMC9793776 DOI: 10.3389/fneur.2022.998320] [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: 07/19/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Over the past few decades, advances in traumatic brain injury (TBI) pathology research have dynamically enriched our knowledge. Therefore, we aimed to systematically elucidate the safety and efficacy of erythropoietin (EPO) dosing regimens in patients with TBI. Methods Data search included PubMed, the Cochrane Library, Embase, Web of Science, and ClinicalTrials.gov for related research published before July 2022. The network meta-analysis was conducted using ADDIS 1.16.8, and the CINeMA tool was used to assess the quality level of evidence. Results A total of six RCTs involving 981 patients were included in the network meta-analysis. EPO did not significantly reduce mortality in patients with TBI, but its risk of death decreased with increasing dosage (odds ratio (OR) of 12,000u vs. placebo = 0.98, 95% CI: 0.03-40.34; OR of group 30,000u vs. placebo = 0.56, 95% CI: 0.06-5.88; OR of 40,000u vs. placebo = 0.35, 95% CI: 0.01-9.43; OR of 70,000u vs. placebo = 0.29, 95% CI: 0.01-9.26; OR of group 80,000u vs. placebo = 0.22, 95% CI: 0.00-7.45). A total of three studies involving 739 patients showed that EPO did not increase the incidence of deep vein thrombosis in patients with TBI. However, the risk tended to rise as the dosage increased. Another two studies demonstrated that EPO did not increase the incidence of pulmonary embolism. The quality of evidence for all outcomes was low to moderate. Conclusion Although the efficacy of EPO was not statistically demonstrated, we found a trend toward an association between EPO dosage and reduced mortality and increased embolic events in patients with TBI. More high-quality original studies should be conducted to obtain strong evidence on the optimal dosage of EPO. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=272500. The study protocol was registered with PROSPERO (CRD42021272500).
Collapse
Affiliation(s)
- Qingyong Zheng
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China,Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou, Gansu, China,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Dan Duan
- Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou, Gansu, China
| | - Jianguo Xu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Xing Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yonggui Ge
- Department of Rehabilitation, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lu Xiong
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Jingjing Yang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Saimire Wulayin
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaofeng Luo
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China,*Correspondence: Xiaofeng Luo
| |
Collapse
|
6
|
Yang B, Dong Y, Xu Z, Li X, Wang F, Zhang Y. Improved stability and pharmacokinetics of wogonin through loading into PASylated ferritin. Colloids Surf B Biointerfaces 2022; 216:112515. [PMID: 35512464 DOI: 10.1016/j.colsurfb.2022.112515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/16/2022] [Accepted: 04/20/2022] [Indexed: 11/25/2022]
Abstract
Wogonin (Wog) plays an important role in human diseases, especially cancer and inflammatory diseases, but its poor solubility, unstable metabolism and low bioavailability greatly limit its application in biomedical fields. Therefore, we developed a temperature-dependent method to encapsulate wogonin into a novel ferritin-based nanocarrier. To improve the loading capacity and stability, the human H chain ferritin (HFtn) was functionalized with a repetitive polypeptide sequence composed of proline (Pro), alanine (Ala), and serine (Ser) in different residues lengths (PAS10 and PAS30). Wogonin loading and release studies demonstrated that the encapsulation efficiency and stability of the PASylated nanocarriers were significantly higher than those of the wild type. PAS-HFtn-Wog exhibited enhanced cytotoxicity to MCF-7 breast cancer cells and HepG2 liver cancer cells. Notably, the PASylated HFtn, especially PAS30-HFtn greatly prolonged the pharmacokinetics of wogonin in the mice bloodstream. Therefore, wogonin-loaded PAS-HFtn may be a promising drug candidate for cancer therapy.
Collapse
Affiliation(s)
- Bingyan Yang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-Forest Biomass, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Yixin Dong
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-Forest Biomass, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Zicheng Xu
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, PR China
| | - Xun Li
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-Forest Biomass, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Fei Wang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-Forest Biomass, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Yu Zhang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-Forest Biomass, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China.
| |
Collapse
|
7
|
Microbial Transglutaminase-Mediated Formation of Erythropoietin-Polyester Conjugates. J Biotechnol 2022; 346:1-10. [PMID: 35038459 DOI: 10.1016/j.jbiotec.2022.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 01/22/2023]
Abstract
Erythropoietin (EPO) is a glycoprotein hormone that has been used to treat anemia in patients with chronic kidney disease and in cancer patients who are receiving chemotherapy. Here, we investigated the accessibility of the glutamine (Gln, Q) residues of recombinant human erythropoietin (rHuEPO) towards a thermoresistant variant microbial transglutaminase (mTGase), TG16 with the aim of developing novel rHuEPO conjugates that may potentially enhance its biological efficacy. As a model bioconjugation, we studied the reactivity of rHuEPO towards TG16 with a low molar mass amine group containing substrate, monodansyl cadaverine (MDC). The reactions were carried out at a Tm of 54.3 °C, the transition temperature of rHuEPO. Characterization by SDS-PAGE and mass spectrometry confirmed the conjugates formation. Then, we examined the conjugation of rHuEPO with a biodegradable and biocompatible polyester, poly(D-sorbitol adipate) (PDSA). To achieve this, PDSA was enzymatically synthesized using lipase B from Candida antartica (CAL-B), chemically modified with side chains having free primary amine (NH2) groups that can be acyl acceptor substrate of TG16, thoroughly characterized by 1H NMR spectroscopy, and then applied for the TG16-mediated conjugation reaction with rHuEPO. rHuEPO conjugates generated by this approach were identified by SDS-PAGE proving that the amine-grafted PDSA is accepted as a substrate for TG16. The successful conjugation was further verified by the detection of high molar mass fluorescent bands after labelling of amine-grafted PDSA with rhodamine B-isothiocyanate. Overall, this enzymatic procedure is considered as an effective approach to prepare biodegradable rHuEPO-polymer conjugates even in the presence of N- and O-glycans.
Collapse
|
8
|
Khodabakhsh F, Salimian M, Hedayati MH, Ahangari Cohan R, Norouzian D. Challenges and advancements in the pharmacokinetic enhancement of therapeutic proteins. Prep Biochem Biotechnol 2021; 51:519-529. [PMID: 33459157 DOI: 10.1080/10826068.2020.1839907] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nowadays, proteins are frequently administered as therapeutic agents in human diseases. However, the main challenge regarding the clinical application of therapeutic proteins is short circulating plasma half-life that leads to more frequent injections for maintaining therapeutic plasma levels, increased therapy costs, immunogenic reactions, and low patient compliance. So, the development of novel strategies to enhance the pharmacokinetic profile of therapeutic proteins has attracted great attention in pharmaceuticals. So far, several techniques, each with their pros and cons, have been developed including chemical bonding to polymers, hyper glycosylation, Fc fusion, human serum albumin fusion, and recombinant PEG mimetics. These techniques mainly classify into three strategies; (i) the endosomal recycling of neonatal Fc receptor which is observed for immunoglobulins and albumin, (ii) decrease in receptor-mediated clearance, and (iii) increase in hydrodynamic radius through chemical and genetic modifications. Recently, novel PEG mimetic peptides like proline/alanine/serine repeat sequences are designed to overcome pitfalls associated with the previous technologies. Biodegradability, lack of or low immunogenicity, product homogeneity, and a simple production process, currently make these polypeptides as the preferred technology for plasma half-life extension of therapeutic proteins. In this review, challenges and pitfalls in the pharmacokinetic enhancement of therapeutic proteins using PEG-mimetic peptides will be discussed in detail.
Collapse
Affiliation(s)
- Farnaz Khodabakhsh
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Morteza Salimian
- Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hossein Hedayati
- Department of Quality Control, Research and Production Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Dariush Norouzian
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
9
|
Bunker A, Róg T. Mechanistic Understanding From Molecular Dynamics Simulation in Pharmaceutical Research 1: Drug Delivery. Front Mol Biosci 2020; 7:604770. [PMID: 33330633 PMCID: PMC7732618 DOI: 10.3389/fmolb.2020.604770] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
In this review, we outline the growing role that molecular dynamics simulation is able to play as a design tool in drug delivery. We cover both the pharmaceutical and computational backgrounds, in a pedagogical fashion, as this review is designed to be equally accessible to pharmaceutical researchers interested in what this new computational tool is capable of and experts in molecular modeling who wish to pursue pharmaceutical applications as a context for their research. The field has become too broad for us to concisely describe all work that has been carried out; many comprehensive reviews on subtopics of this area are cited. We discuss the insight molecular dynamics modeling has provided in dissolution and solubility, however, the majority of the discussion is focused on nanomedicine: the development of nanoscale drug delivery vehicles. Here we focus on three areas where molecular dynamics modeling has had a particularly strong impact: (1) behavior in the bloodstream and protective polymer corona, (2) Drug loading and controlled release, and (3) Nanoparticle interaction with both model and biological membranes. We conclude with some thoughts on the role that molecular dynamics simulation can grow to play in the development of new drug delivery systems.
Collapse
Affiliation(s)
- Alex Bunker
- Division of Pharmaceutical Biosciences, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Tomasz Róg
- Department of Physics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
10
|
Najjari A, Rahimi H, Nojoumi SA, Omidinia E. Computational Approach for Rational Design of Fusion Uricase with PAS Sequences. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2020; 9:90-103. [PMID: 32832488 PMCID: PMC7422847 DOI: 10.22088/ijmcm.bums.9.1.90] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Tumor lysis syndrome is a life-threatening condition for humans due to the lack of urate oxidase. In this study, several variants of PASylated uricase from the Aspergillus flavus species were analyzed computationally to find the appropriate fusions to solve short half-life and stability concern. The Ab initio method was performed using Rosetta software to structurally characterize the PAS sequences. The 3D structures of fusions were predicted for fused C- or N-terminally PAS sequences in different length to the uricase. The refinement and energy minimization steps revealed that physicochemical and conformational properties of fusions improved while the structures possessed prolonged PAS sequences. Molecular docking results showed that the highest binding affinity to uric acid belonged to uricase-PAS1-100 by the formation of six hydrogen and four non-hydrogen bonds. Altogether, the results indicated that the PASylation process would be promising upon the production of urate oxidase with improved solubility and stability.
Collapse
Affiliation(s)
- Abbas Najjari
- Enzyme Technology Laboratory, Department of Biochemistry,Genetic and Metabolism Research Group, Pasteur Institute of Iran,Tehran, Iran
| | - Hamzeh Rahimi
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Ali Nojoumi
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Eskandar Omidinia
- Enzyme Technology Laboratory, Department of Biochemistry,Genetic and Metabolism Research Group, Pasteur Institute of Iran,Tehran, Iran
| |
Collapse
|
11
|
Khodabakhsh F, Salimian M, Mehdizadeh A, Khosravy MS, Vafabakhsh A, Karami E, Cohan RA. New Proline, Alanine, Serine Repeat Sequence for Pharmacokinetic Enhancement of Anti-VEGF Single-Domain Antibody. J Pharmacol Exp Ther 2020; 375:69-75. [PMID: 32669367 DOI: 10.1124/jpet.120.000012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/10/2020] [Indexed: 01/14/2023] Open
Abstract
Therapeutic fragmented antibodies show a poor pharmacokinetic profile that leads to frequent high-dose administration. In the current study, for the first time, a novel proline, alanine, serine (PAS) repeat sequence called PAS#208 was designed to extend the plasma half-life of a nanosized anti-vascular endothelial growth factor-A single-domain antibody. Polyacrylamide gel electrophoresis, circular dichroism, dynamic light scattering, and electrophoretic light scattering were used to assess the physicochemical properties of the newly designed PAS sequence. The effect of PAS#208 on the biologic activity of a single-domain antibody was studied using an in vitro proliferation assay. The pharmacokinetic parameters, including terminal half-life, the volume of distribution, elimination rate constant, and clearance, were determined in mice model and compared with the native protein and PAS#1(200) sequence. The novel PAS repeat sequence showed comparable physicochemical, biologic, and pharmacokinetic features to the previously reported PAS#1(200) sequence. The PAS#208 increased the hydrodynamic radius and decreased significantly the electrophoretic mobility of the native protein without any change in zeta potential. Surprisingly, the fusion of PAS#208 to the single-domain antibody increased the binding potency. In addition, it did not alter the biologic activity and did not show any cytotoxicity on the normal cells. The PAS#208 sequence improved the terminal half-life (14-fold) as well as other pharmacokinetic parameters significantly. The simplicity as well as superior effects on half-life extension make PAS#208 sequence a novel sequence for in vivo pharmacokinetic enhancement of therapeutic fragmented antibodies. SIGNIFICANCE STATEMENT: In the current study, a new proline, alanine, serine (PAS) sequence was developed that showed comparable physicochemical, biological, and pharmacokinetic features to the previously reported PAS#1(200) sequence. The simplicity as well as superior effects on half-life extension make PAS#208 sequence a novel sequence for in vivo pharmacokinetic enhancement of recombinant small proteins.
Collapse
Affiliation(s)
- Farnaz Khodabakhsh
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran (F.K.); Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, Iran (M.S.); Department of Civil Engineering, Sharif University of Technology, Tehran, Iran (A.M.); Department of Rabies, Virology Research Group (S.K.) and Department of Nanobiotechnology, New Technologies Research Group (R.A.C.), Pasteur Institute of Iran, Tehran, Iran; and Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran (A.V., E.K.)
| | - Morteza Salimian
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran (F.K.); Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, Iran (M.S.); Department of Civil Engineering, Sharif University of Technology, Tehran, Iran (A.M.); Department of Rabies, Virology Research Group (S.K.) and Department of Nanobiotechnology, New Technologies Research Group (R.A.C.), Pasteur Institute of Iran, Tehran, Iran; and Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran (A.V., E.K.)
| | - Ardavan Mehdizadeh
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran (F.K.); Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, Iran (M.S.); Department of Civil Engineering, Sharif University of Technology, Tehran, Iran (A.M.); Department of Rabies, Virology Research Group (S.K.) and Department of Nanobiotechnology, New Technologies Research Group (R.A.C.), Pasteur Institute of Iran, Tehran, Iran; and Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran (A.V., E.K.)
| | - Mohammad Sadeq Khosravy
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran (F.K.); Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, Iran (M.S.); Department of Civil Engineering, Sharif University of Technology, Tehran, Iran (A.M.); Department of Rabies, Virology Research Group (S.K.) and Department of Nanobiotechnology, New Technologies Research Group (R.A.C.), Pasteur Institute of Iran, Tehran, Iran; and Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran (A.V., E.K.)
| | - Alireza Vafabakhsh
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran (F.K.); Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, Iran (M.S.); Department of Civil Engineering, Sharif University of Technology, Tehran, Iran (A.M.); Department of Rabies, Virology Research Group (S.K.) and Department of Nanobiotechnology, New Technologies Research Group (R.A.C.), Pasteur Institute of Iran, Tehran, Iran; and Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran (A.V., E.K.)
| | - Elmira Karami
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran (F.K.); Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, Iran (M.S.); Department of Civil Engineering, Sharif University of Technology, Tehran, Iran (A.M.); Department of Rabies, Virology Research Group (S.K.) and Department of Nanobiotechnology, New Technologies Research Group (R.A.C.), Pasteur Institute of Iran, Tehran, Iran; and Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran (A.V., E.K.)
| | - Reza Ahangari Cohan
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran (F.K.); Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, Iran (M.S.); Department of Civil Engineering, Sharif University of Technology, Tehran, Iran (A.M.); Department of Rabies, Virology Research Group (S.K.) and Department of Nanobiotechnology, New Technologies Research Group (R.A.C.), Pasteur Institute of Iran, Tehran, Iran; and Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran (A.V., E.K.)
| |
Collapse
|
12
|
Shamloo A, Rostami P, Mahmoudi A. PASylation Enhances the Stability, Potency, and Plasma Half-Life of Interferon α-2a: A Molecular Dynamics Simulation. Biotechnol J 2020; 15:e1900385. [PMID: 32277577 DOI: 10.1002/biot.201900385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 03/16/2020] [Indexed: 12/17/2022]
Abstract
In this study, the effectiveness of PASylation in enhancing the potency and plasma half-life of pharmaceutical proteins has been accredited as an alternative technique to the conventional methods such as PEGylation. Proline, alanine, and serine (PAS) chain has shown some advantages including biodegradability improvement and plasma half-life enhancement while lacking immunogenicity or toxicity. Although some experimental studies have been performed to find the mechanism behind PASylation, the detailed mechanism of PAS effects on the pharmaceutical proteins has remained obscure, especially at the molecular level. In this study, the interaction of interferon α-2a (IFN) and PAS chain is investigated using molecular dynamics simulation method. Several important parameters including secondary structure, root-mean-square distance, and solvent accessible surface area to investigate the stability, bioavailability, and bioactivity of the PASylated protein are studied. The results demonstrate that IFN conformation is not affected critically through PASylation while it results in improvement of the protein stability and bioactivity. Therefore, PASylation can be considered as a proper biological alternative technique to increase the plasma half-life of the biopharmaceutical proteins through enlarging apparent volume. The proposed simulation represents a computational approach that would provide a basis for the study of PASylated pharmaceutical proteins for different future applications.
Collapse
Affiliation(s)
- Amir Shamloo
- Department of mechanical engineering, Sharif University of Technology, Azadi Ave. 11155-9567, Tehran, Iran
| | - Peyman Rostami
- Department of mechanical engineering, Sharif University of Technology, Azadi Ave. 11155-9567, Tehran, Iran
| | - Ashkan Mahmoudi
- Department of Aerospace Engineering, Sharif University of Technology, Azadi Ave. 11365-11155, Tehran, Iran
| |
Collapse
|
13
|
Varanko A, Saha S, Chilkoti A. Recent trends in protein and peptide-based biomaterials for advanced drug delivery. Adv Drug Deliv Rev 2020; 156:133-187. [PMID: 32871201 PMCID: PMC7456198 DOI: 10.1016/j.addr.2020.08.008] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
Engineering protein and peptide-based materials for drug delivery applications has gained momentum due to their biochemical and biophysical properties over synthetic materials, including biocompatibility, ease of synthesis and purification, tunability, scalability, and lack of toxicity. These biomolecules have been used to develop a host of drug delivery platforms, such as peptide- and protein-drug conjugates, injectable particles, and drug depots to deliver small molecule drugs, therapeutic proteins, and nucleic acids. In this review, we discuss progress in engineering the architecture and biological functions of peptide-based biomaterials -naturally derived, chemically synthesized and recombinant- with a focus on the molecular features that modulate their structure-function relationships for drug delivery.
Collapse
Affiliation(s)
| | | | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
14
|
Roy U. Structure and Function of an Inflammatory Cytokine, Interleukin-2, Analyzed Using the Bioinformatic Approach. Protein J 2019; 38:525-536. [PMID: 31006082 DOI: 10.1007/s10930-019-09833-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The inflammatory cytokine, interleukin-2 (IL-2), is an important regulator of cellular functions. This relatively less studied member of the interleukin protein family is responsible for multiple immuno-modulatory and immuno-stimulatory tasks, like T cell activation, triggering of natural killer cells, inflammation, as well as proliferation and progression of autoimmune diseases and cancers. In this communication we report the temporally variant structural aspects of the IL-2 ligand and its receptor interfaces, based on the available crystal structures. The intended goal of this effort is to generate simulated results that could potentially aid the designs of novel structure based therapeutics.
Collapse
Affiliation(s)
- Urmi Roy
- Department of Chemistry & Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699-5820, USA.
| |
Collapse
|
15
|
Enhancing bioactivity, physicochemical, and pharmacokinetic properties of a nano-sized, anti-VEGFR2 Adnectin, through PASylation technology. Sci Rep 2019; 9:2978. [PMID: 30814652 PMCID: PMC6393559 DOI: 10.1038/s41598-019-39776-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
The crucial role of VEGF receptor 2 (VEGFR2) signaling in the angiogenesis and metastasis of solid tumors has prompted the development of inhibitors with minimal bystander effects. Recently, Adnectin C has attracted attention for cancer treatment. To overcome the problematic properties of Adnectin, a novel form of Adnectin C has been designed by its fusion to a biodegradable polymeric peptide containing Pro/Ala/Ser (PAS) repetitive residues. E. coli-expressed recombinant fused and unfused proteins were compared in terms of bioactivity, physicochemical, and pharmacokinetic properties using standard methods. Dynamic light scattering (DLS) analysis of PASylated adnectin C revealed an approximate 2-fold increase in particle size with a slight change in the net charge. Additionally, fusion of the PAS sequence improved its stability against the growth of thermo-induced aggregated forms. The high receptor-binding and improved binding kinetic parameters of PASylated Adnectin C was confirmed by ELISA and surface plasmon resonance assays, respectively. Pharmacokinetic studies showed a noticeable increase in the terminal half-life of Adnectin C-PAS#1(200) by a factor of 4.57 after single dose by intravenous injection into female BALB/c mice. The results suggest that PASylation could offer a superior delivery strategy for developing Adnectin-derived drugs with improved patient compliance.
Collapse
|
16
|
Rouhani M, Khodabakhsh F, Norouzian D, Cohan RA, Valizadeh V. Molecular dynamics simulation for rational protein engineering: Present and future prospectus. J Mol Graph Model 2018; 84:43-53. [DOI: 10.1016/j.jmgm.2018.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 12/19/2022]
|
17
|
Abstract
Nanoparticle delivery systems offer advantages over free drugs, in that they increase solubility and biocompatibility. Nanoparticles can deliver a high payload of therapeutic molecules while limiting off-target side effects. Therefore, delivery of an existing drug with a nanoparticle frequently results in an increased therapeutic index. Whether of synthetic or biologic origin, nanoparticle surface coatings are often required to reduce immune clearance and thereby increase circulation times allowing the carriers to reach their target site. To this end, polyethylene glycol (PEG) has long been used, with several PEGylated products reaching clinical use. Unfortunately, the growing use of PEG in consumer products has led to an increasing prevalence of PEG-specific antibodies in the human population, which in turn has fueled the search for alternative coating strategies. This review highlights alternative bioinspired nanoparticle shielding strategies, which may be more beneficial moving forward than PEG and other synthetic polymer coatings.
Collapse
Affiliation(s)
- Neetu M. Gulati
- Department of Pharmacology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Cleveland Center for Membrane and Structural Biology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
| | - Phoebe L. Stewart
- Department of Pharmacology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Cleveland Center for Membrane and Structural Biology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Department of Radiology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Department of Materials Science and Engineering, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Department of Macromolecular Science and Engineering, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
18
|
Breibeck J, Skerra A. The polypeptide biophysics of proline/alanine-rich sequences (PAS): Recombinant biopolymers with PEG-like properties. Biopolymers 2017; 109. [PMID: 29076532 PMCID: PMC5813227 DOI: 10.1002/bip.23069] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/21/2017] [Accepted: 09/23/2017] [Indexed: 12/14/2022]
Abstract
PAS polypeptides comprise long repetitive sequences of the small L‐amino acids proline, alanine and/or serine that were developed to expand the hydrodynamic volume of conjugated pharmaceuticals and prolong their plasma half‐life by retarding kidney filtration. Here, we have characterized the polymer properties both of the free polypeptides and in fusion with the biopharmaceutical IL‐1Ra. Data from size exclusion chromatography, dynamic light scattering, circular dichroism spectroscopy and quantification of hydrodynamic and polar properties demonstrate that the biosynthetic PAS polypeptides exhibit random coil behavior in aqueous solution astonishingly similar to the chemical polymer poly‐ethylene glycol (PEG). The solvent‐exposed PAS peptide groups, in the absence of secondary structure, account for strong hydrophilicity, with negligible contribution by the Ser side chains. Notably, PAS polypeptides exceed PEG of comparable molecular mass in hydrophilicity and hydrodynamic volume while exhibiting lower viscosity. Their uniform monodisperse composition as genetically encoded polymers and their biological nature, offering biodegradability, render PAS polypeptides a promising PEG mimetic for biopharmaceutical applications.
Collapse
Affiliation(s)
- Joscha Breibeck
- Lehrstuhl für Biologische Chemie, Technische Universität München, 85354, Freising (Weihenstephan), Germany
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, 85354, Freising (Weihenstephan), Germany.,XL-protein GmbH, Lise-Meitner-Str. 30, 85354, Freising, Germany
| |
Collapse
|
19
|
Binder U, Skerra A. PASylation®: A versatile technology to extend drug delivery. Curr Opin Colloid Interface Sci 2017. [DOI: 10.1016/j.cocis.2017.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
20
|
Khodabakhsh F, Norouzian D, Vaziri B, Ahangari Cohan R, Sardari S, Mahboudi F, Behdani M, Mansouri K, Mehdizadeh A. Development of a novel nano-sized anti-VEGFA nanobody with enhanced physicochemical and pharmacokinetic properties. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1402-1414. [PMID: 28841807 DOI: 10.1080/21691401.2017.1369426] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Since physiological and pathological processes occur at nano-environments, nanotechnology has considered as an efficient tool for designing of next generation specific biomolecules with enhanced pharmacodynamic and pharmacodynamic properties. In the current investigation, by control of the size and hydrodynamic volume at the nanoscale, for the first time, physicochemical and pharmacokinetic properties of an anti-VEGFA nanobody was remarkably improved by attachment of a Proline-Alanine-Serine (PAS) rich sequence. The results elucidated unexpected impressive effects of PAS sequence on physicochemical properties especially on size, hydrodynamics radius, and even solubility of nanobody. CD analysis revealed an increment in random coil structure of the PASylated protein in comparison to native one without any change in charge state or binding kinetic parameters of nanobody assessed by isoelectric focusing and surface plasmon resonance measurements, respectively. In vitro biological activities of nanobody were not affected by coupling of the PAS sequence. In contrast, the terminal half-life was significantly increased by a factor of 14 for the nanobody-PAS after single dose IV injection to the mice. Our study demonstrated that the control of size in the design of small therapeutic proteins has a promising effect on the stability and solubility, in addition to their physiochemical and pharmacokinetic properties. The designed new anti-VEGFA nanobody could promise a better therapeutic agent with a long administration intervals and lower dose, which in turn leads to a better patient compliance. Size adjustment of an anti-VEGF nanobody at the nanoscale by the attachment of a natural PAS polymer remarkably improves physicochemical properties, as well as a pharmacokinetic profile without any change in biological activity of the miniaturized antibody.
Collapse
Affiliation(s)
- Farnaz Khodabakhsh
- a Department of Nanobiotechnology , Advanced Technology Group, Pasteur Institute of Iran , Tehran , Iran.,b Biotechnology Research Center, Pasteur Institute of Iran , Tehran , Iran
| | - Dariush Norouzian
- a Department of Nanobiotechnology , Advanced Technology Group, Pasteur Institute of Iran , Tehran , Iran
| | - Behrouz Vaziri
- b Biotechnology Research Center, Pasteur Institute of Iran , Tehran , Iran
| | - Reza Ahangari Cohan
- a Department of Nanobiotechnology , Advanced Technology Group, Pasteur Institute of Iran , Tehran , Iran
| | - Soroush Sardari
- c Drug Design and Bioinformatics Unit, Department of Medical Biotechnology , Biotechnology Research Center, Pasteur Institute of Iran , Tehran , Iran
| | - Fereidoun Mahboudi
- b Biotechnology Research Center, Pasteur Institute of Iran , Tehran , Iran
| | - Mahdi Behdani
- d Venom & Biotherapeutics Molecules Laboratory , Biotechnology Research Center, Pasteur Institute of Iran , Tehran , Iran
| | - Kamran Mansouri
- e Medical Biology Research Center, Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Ardavan Mehdizadeh
- f Department of Civil Engineering , Sharif University of Technology , Tehran , Iran
| |
Collapse
|