1
|
Binder U, Skerra A. Strategies for extending the half-life of biotherapeutics: successes and complications. Expert Opin Biol Ther 2025; 25:93-118. [PMID: 39663567 DOI: 10.1080/14712598.2024.2436094] [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: 08/16/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION Engineering of the drug half-life in vivo has become an integral part of modern biopharmaceutical development due to the fact that many proteins/peptides with therapeutic potential are quickly cleared by kidney filtration after injection and, thus, circulate only a few hours in humans (or just minutes in mice). AREAS COVERED Looking at the growing list of clinically approved biologics that have been modified for prolonged activity, and also the plethora of such drugs under preclinical and clinical development, it is evident that not one solution fits all needs, owing to the vastly different structural features and functional properties of the pharmacologically active entities. This article provides an overview of established half-life extension strategies, as well as of emerging novel concepts for extending the in vivo stability of biologicals, and their pros and cons. EXPERT OPINION Beyond the classical and still dominating technologies for improving drug pharmacokinetics and bioavailability, Fc fusion and PEGylation, various innovative approaches that offer advantages in different respects have entered the clinical stage. While the Fc fusion partner may be gradually superseded by engineered albumin-binding domains, chemical PEGylation may be replaced by biodegradable recombinant amino-acid polymers like PASylation, thus also offering a purely biotechnological manufacturing route.
Collapse
Affiliation(s)
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Freising, Germany
| |
Collapse
|
2
|
Radhakrishnan H, Newmyer SL, Javitz HS, Bhatnagar P. Engineered CD4 T cells for in vivo delivery of therapeutic proteins. Proc Natl Acad Sci U S A 2024; 121:e2318687121. [PMID: 39312667 PMCID: PMC11459198 DOI: 10.1073/pnas.2318687121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
The CD4 T cell, when engineered with a chimeric antigen receptor (CAR) containing specific intracellular domains, has been transformed into a zero-order drug-delivery platform. This introduces the capability of prolonged, disease-specific engineered protein biologics production, at the disease site. Experimental findings demonstrate that CD4 T cells offer a solution when modified with a CAR that includes 4-1BB but excludes CD28 intracellular domain. In this configuration, they achieve ~3X transduction efficiency of CD8 T cells, ~2X expansion rates, generating ~5X more biologic, and exhibit minimal cytolytic activity. Cumulatively, this addresses two main hurdles in the translation of cell-based drug delivery: scaling the production of engineered T cell ex vivo and generating sufficient biologics in vivo. When programmed to induce IFNβ upon engaging the target antigen, the CD4 T cells outperforms CD8 T cells, effectively suppressing cancer cell growth in vitro and in vivo. In summary, this platform enables precise targeting of disease sites with engineered protein-based therapeutics while minimizing healthy tissue exposure. Leveraging CD4 T cells' persistence could enhance disease management by reducing drug administration frequency, addressing critical challenges in cell-based therapy.
Collapse
|
3
|
Tripathy RK, Anakha J, Pande AH. Towards development of biobetter: L-asparaginase a case study. Biochim Biophys Acta Gen Subj 2024; 1868:130499. [PMID: 37914146 DOI: 10.1016/j.bbagen.2023.130499] [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: 10/03/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND L-asparaginase (ASNase) has played a key role in the management of acute lymphoblastic leukaemia (ALL). As an amidohydrolase, it catalyzes the hydrolysis of L-asparagine, a crucial step in the treatment of ALL. Various ASNase variants have evolved from diverse sources since it was first used in paediatric patients in the 1960s. This review describes the available ASNase and approaches being used to develop ASNase as a biobetter candidate. SCOPE OF REVIEW The review discusses the Glycosylation and PEGylation techniques, which are frequently used to develop biobetter versions of the majority of the therapeutic proteins. Further, it explores current ASNase biobetters in therapeutic use and discusses the protein engineering and chemical modification approaches that were employed to reduce immunogenicity, extend protein half-life, and enhance protease stability of ASNase. Emerging strategies like immobilization and encapsulation are also highlighted as potential pathways for improving ASNase properties. MAJOR CONCLUSIONS The purpose of the development of ASNase biobetter is to achieve a novel therapeutic candidate that could improve catalytic efficiency, in vivo stability with minimum glutaminase (GLNase) activity and toxicity. Modification of ASNase by immobilization and encapsulation or by fusion technologies like Albumin fusion, Fc fusion, ELP fusion, XTEN fusion, etc. can be exploited to develop a novel biobetter candidate suitable for therapeutic approaches. GENERAL SIGNIFICANCE This review emphasizes the importance of biobetter development for therapeutic proteins like ASNase. Improved ASNase molecules have the potential to significantly advance the treatment of ALL and have broader implications in the pharmaceutical industry.
Collapse
Affiliation(s)
- Rajan K Tripathy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - J Anakha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
4
|
Maejima A, Suzuki S, Makabe K, Kumagai I, Asano R. Incorporation of a repeated polypeptide sequence in therapeutic antibodies as a universal masking procedure: A case study of T cell-engaging bispecific antibodies. N Biotechnol 2023; 77:80-89. [PMID: 37467927 DOI: 10.1016/j.nbt.2023.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/06/2023] [Accepted: 07/16/2023] [Indexed: 07/21/2023]
Abstract
Prodrug design is a promising approach for reducing the off-target effects of therapeutic antibodies, particularly bispecific antibodies (bsAbs) that recruit T cells for activation; this design uses masking sequences that inhibit antibody binding until they reach the tumor microenvironment, where they are removed. In this study, we propose PAS, a polypeptide sequence composed of repeated Pro, Ala, and Ser residues, as a universal masking sequence. PAS has no specificity, but can inhibit antibody binding through steric hindrance caused by its large fluid dynamic radius and disordered structure; additionally, its length can be adjusted. We fused PAS to the N-terminus of an anti-CD3 single-chain variable fragment (scFv) and a bsAb, that targets both the epidermal growth factor receptor and CD3, via a recognition sequence cleaved by cancer-related proteases. PAS integration inhibited anti-CD3 scFv binding with higher efficacy than the epitope sequence, and the extent of inhibition was proportional to the length of the PAS sequence. For masked bsAbs, T cell-binding ability, cancer growth inhibition effects, and T cell activation effects were also reduced depending on the length of PAS and were fully restored upon removing PAS sequences using protease. The masking procedure using PAS was successfully applied to another scFv. The provision to adjust the masking effects of PAS by tuning its length, makes PAS fusion a valuable tool for the universal design of prodrug antibodies.
Collapse
Affiliation(s)
- Atsushi Maejima
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Saori Suzuki
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Koki Makabe
- Graduate School of Science and Engineering, Faculty of Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa 992-8510, Japan
| | - Izumi Kumagai
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Ryutaro Asano
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan; Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1, Harumi-cho, Fuchu, Tokyo 183-8509, Japan.
| |
Collapse
|
5
|
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: 9] [Impact Index Per Article: 4.5] [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
|
6
|
Friedrich L, Kikuchi Y, Matsuda Y, Binder U, Skerra A. Efficient secretory production of proline/alanine/serine (PAS) biopolymers in Corynebacterium glutamicum yielding a monodisperse biological alternative to polyethylene glycol (PEG). Microb Cell Fact 2022; 21:227. [PMID: 36307781 PMCID: PMC9616612 DOI: 10.1186/s12934-022-01948-5] [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: 06/10/2022] [Accepted: 10/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background PAS biopolymers are recombinant polypeptides comprising the small uncharged l-amino acids Pro, Ala and/or Ser which resemble the widely used poly-ethylene glycol (PEG) in terms of pronounced hydrophilicity. Likewise, their random chain behaviour in physiological solution results in a strongly expanded hydrodynamic volume. Thus, apart from their use as fusion partner for biopharmaceuticals to achieve prolonged half-life in vivo, PAS biopolymers appear attractive as substitute for PEG—or other poorly degradable chemical polymers—in many areas. As a prerequisite for the wide application of PAS biopolymers at affordable cost, we have established their highly efficient biotechnological production in Corynebacterium glutamicum serving as a well characterized bacterial host organism. Results Using the CspA signal sequence, we have secreted two representative PAS biopolymers as polypeptides with ~ 600 and ~ 1200 amino acid residues, respectively. Both PAS biopolymers were purified from the culture supernatant by means of a simple downstream process in a truly monodisperse state as evidenced by ESI–MS. Yields after purification were up to ≥ 4 g per liter culture, with potential for further increase by strain optimization as well as fermentation and bioprocess development. Beyond direct application as hydrocolloids or to exploit their rheological properties, such PAS biopolymers are suitable for site-specific chemical conjugation with pharmacologically active molecules via their unique terminal amino or carboxyl groups. To enable the specific activation of the carboxylate, without interference by the free amino group, we generated a blocked N-terminus for the PAS(1200) polypeptide simply by introducing an N-terminal Gln residue which, after processing of the signal peptide, was cyclised to a chemically inert pyroglutamyl group upon acid treatment. The fact that PAS biopolymers are genetically encoded offers further conjugation strategies via incorporation of amino acids with reactive side chains (e.g., Cys, Lys, Glu/Asp) at defined positions. Conclusions Our new PAS expression platform using Corynex® technology opens the way to applications of PASylation® technology in multiple areas such as the pharmaceutical industry, cosmetics and food technology.
Collapse
Affiliation(s)
- L Friedrich
- XL-protein GmbH, Lise-Meitner-Strasse 30, 85354, Freising, Germany
| | - Y Kikuchi
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki, 210-8681, Japan
| | - Y Matsuda
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki, 210-8681, Japan
| | - U Binder
- XL-protein GmbH, Lise-Meitner-Strasse 30, 85354, Freising, Germany
| | - A Skerra
- XL-protein GmbH, Lise-Meitner-Strasse 30, 85354, Freising, Germany. .,Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354, Freising, Germany.
| |
Collapse
|
7
|
Abstract
The lack of a human immunodeficiency virus (HIV) cure has heightened interest in immunotherapy. As such, type I interferons (IFNs), in particular, IFN alpha (IFN-α), have gained renewed attention. However, HIV pathogenesis is driven by sustained IFN-mediated immune activation, and the use of IFNs is rather controversial. The following questions therein remain: (i) which IFN-α subtype to use, (ii) at which regimen, and (iii) at what time point in HIV infection it might be beneficial. Here, we used IFN-α14 modified by PASylation for its long half-life in vivo to eventually treat HIV infection. We defined the IFN dosing regimen based on the maximum increase in interferon-stimulated gene (ISG) expression 6 h after its administration and a return to baseline of ubiquitin-specific protease 18 (USP18) prior to the next dose. Notably, USP18 is the major negative regulator of type I IFN signaling. HIV infection resulted in increased ISG expression levels in humanized mice. Intriguingly, high baseline ISG levels correlated with lower HIV load. No effect was observed on HIV replication when PASylated IFN-α14 was administered in the chronic phase. However, combined antiretroviral therapy (cART) restored responsiveness to IFN, and PASylated IFN-α14 administered during analytical cART interruption resulted in a transiently lower HIV burden than in the mock-treated mice. In conclusion, cART-mediated HIV suppression restored transient IFN responsiveness and provided a potential window for immunoenhancing therapies in the context of analytical cART interruption. IMPORTANCE cART is highly efficient in suppressing HIV replication in HIV-infected patients and has resulted in a dramatic reduction in morbidity and mortality in HIV-infected people, yet it does not cure HIV infection. In addition, cART has several disadvantages. Thus, the HIV research community is exploring novel ways to control HIV infection for longer periods without cART. Here, we explored novel, long-acting IFN-α14 for its efficacy to control HIV replication in HIV-infected humanized mice. We found that IFN-α14 had no effect on chronic HIV infection. However, when mice were treated first with cART, we observed a transiently restored responsiveness to INF and a transiently lower HIV burden after stopping cART. These data emphasize (i) the value of cART-mediated HIV suppression and immune reconstitution in creating a window of opportunity for exploring novel immunotherapies, (ii) the potential of IFNs for constraining HIV, and (iii) the value of humanized mice for exploring novel immunotherapies.
Collapse
|
8
|
Sumi T, Harada K. Immune response to SARS-CoV-2 in severe disease and long COVID-19. iScience 2022; 25:104723. [PMID: 35813874 PMCID: PMC9251893 DOI: 10.1016/j.isci.2022.104723] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/23/2022] [Accepted: 06/29/2022] [Indexed: 01/10/2023] Open
Abstract
COVID-19 is mild to moderate in otherwise healthy individuals but may nonetheless cause life-threatening disease and/or a wide range of persistent symptoms. The general determinant of disease severity is age mainly because the immune response declines in aging patients. Here, we developed a mathematical model of the immune response to SARS-CoV-2 and revealed that typical age-related risk factors such as only a several 10% decrease in innate immune cell activity and inhibition of type-I interferon signaling by autoantibodies drastically increased the viral load. It was reported that the numbers of certain dendritic cell subsets remained less than half those in healthy donors even seven months after infection. Hence, the inflammatory response was ongoing. Our model predicted the persistent DC reduction and showed that certain patients with severe and even mild symptoms could not effectively eliminate the virus and could potentially develop long COVID.
Collapse
Affiliation(s)
- Tomonari Sumi
- Research Institute for Interdisciplinary Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
- Department of Chemistry, Faculty of Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Kouji Harada
- Department of Computer Science and Engineering, Toyohashi University of Technology, Tempaku-cho, Toyohashi 441-8580, Japan
- Center for IT-Based Education, Toyohashi University of Technology, Tempaku-cho, Toyohashi, Aichi 441-8580, Japan
| |
Collapse
|
9
|
Khorramdelazad H, Kazemi MH, Azimi M, Aghamajidi A, Mehrabadi AZ, Shahba F, Aghamohammadi N, Falak R, Faraji F, Jafari R. Type-I interferons in the immunopathogenesis and treatment of Coronavirus disease 2019. Eur J Pharmacol 2022; 927:175051. [PMID: 35618037 PMCID: PMC9124632 DOI: 10.1016/j.ejphar.2022.175051] [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: 01/14/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 12/16/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), is currently the major global health problem. Still, it continues to infect people globally and up to the end of February 2022, over 436 million confirmed cases of COVID-19, including 5.95 million deaths, were reported to the world health organization (WHO). No specific treatment is currently available for COVID-19, and the discovery of effective therapeutics requires understanding the effective immunologic and immunopathologic mechanisms behind this infection. Type-I interferons (IFN-Is), as the critical elements of the immediate immune response against viral infections, can inhibit the replication and spread of the viruses. However, the available evidence shows that the antiviral IFN-I response is impaired in patients with the severe form of COVID-19. Moreover, the administration of exogenous IFN-I in different phases of the disease can lead to various outcomes. Therefore, understanding the role of IFN-I molecules in COVID-19 development and its severity can provide valuable information for better management of this disease. This review summarizes the role of IFN-Is in the pathogenesis of COIVD-19 and discusses the importance of autoantibodies against this cytokine in the spreading of SARS-CoV-2 and control of the subsequent excessive inflammation.
Collapse
Affiliation(s)
- Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Kazemi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Azimi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Azin Aghamajidi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Zarezadeh Mehrabadi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Faezeh Shahba
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Aghamohammadi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Faraji
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran,Corresponding author. Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Floor 3, Building No. 3, Hazrat-e Rasool General Hospital, Niyayesh St, Sattar Khan St, 1445613131, Tehran, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran,Corresponding author. Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Shafa St., Ershad Blvd, Imam Khomeini Hospital Complex, 113857147, Urmia, Iran
| |
Collapse
|
10
|
Yin S, Zhang B, Lin J, Liu Y, Su Z, Bi J. Development of purification process for dual-function recombinant human heavy-chain ferritin by the investigation of genetic modification impact on conformation. Eng Life Sci 2021; 21:630-642. [PMID: 34690634 PMCID: PMC8518560 DOI: 10.1002/elsc.202000105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/08/2021] [Accepted: 05/21/2021] [Indexed: 12/02/2022] Open
Abstract
Ferritin is a promising drug delivery platform and has been functionalized through genetic modifications. This work has designed and expressed a dual-functional engineered human heavy-chain ferritin (HFn) with the inserted functional peptide PAS and RGDK to extend half-life and improve tumor targeted drug delivery. A facile and cost-effective two-step purification pathway for recombinant HFn was developed. The genetic modification was found to affect HFn conformation, and therefore varied the purification performance. Heat-acid precipitation followed by butyl fast flow hydrophobic interaction chromatography (HIC) has been developed to purify HFn and modified HFns. Nucleic acid removal reached above 99.8% for HFn and modified HFns. However, HFn purity reached above 95% and recovery yield (overall) above 90%, compared with modified HFns purity above 82% and recovery yield (overall) above 58%. It is interesting to find that the inserted functional peptides significantly changed the molecule conformation, where a putative turnover of the E-helix with the inserted functional peptides formed a "flop" conformation, in contrast with the "flip" conformation of HFn. It could be the cause of fragile stability of modified HFns, and therefore less tolerant to heat and acid condition, observed by the lower recovery yield in heat-acid precipitation.
Collapse
Affiliation(s)
- Shuang Yin
- School of Chemical Engineering & Advanced MaterialsFaculty of Engineering, Computer and Mathematical SciencesUniversity of AdelaideAdelaideAustralia
| | - Bingyang Zhang
- School of Chemical Engineering & Advanced MaterialsFaculty of Engineering, Computer and Mathematical SciencesUniversity of AdelaideAdelaideAustralia
| | - Jianying Lin
- College of Biomedical EngineeringTaiyuan University of TechnologyTaiyuanP. R. China
| | - Yongdong Liu
- State Key Laboratory of Biochemistry EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
| | - Zhiguo Su
- State Key Laboratory of Biochemistry EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijingP. R. China
| | - Jingxiu Bi
- School of Chemical Engineering & Advanced MaterialsFaculty of Engineering, Computer and Mathematical SciencesUniversity of AdelaideAdelaideAustralia
| |
Collapse
|
11
|
Zhao J, Roberts A, Wang Z, Savage J, Ji RR. Emerging Role of PD-1 in the Central Nervous System and Brain Diseases. Neurosci Bull 2021; 37:1188-1202. [PMID: 33877518 PMCID: PMC8353059 DOI: 10.1007/s12264-021-00683-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/19/2020] [Indexed: 12/13/2022] Open
Abstract
Programmed cell death protein 1 (PD-1) is an immune checkpoint modulator and a major target of immunotherapy as anti-PD-1 monoclonal antibodies have demonstrated remarkable efficacy in cancer treatment. Accumulating evidence suggests an important role of PD-1 in the central nervous system (CNS). PD-1 has been implicated in CNS disorders such as brain tumors, Alzheimer's disease, ischemic stroke, spinal cord injury, multiple sclerosis, cognitive function, and pain. PD-1 signaling suppresses the CNS immune response via resident microglia and infiltrating peripheral immune cells. Notably, PD-1 is also widely expressed in neurons and suppresses neuronal activity via downstream Src homology 2 domain-containing protein tyrosine phosphatase 1 and modulation of ion channel function. An improved understanding of PD-1 signaling in the cross-talk between glial cells, neurons, and peripheral immune cells in the CNS will shed light on immunomodulation, neuromodulation, and novel strategies for treating brain diseases.
Collapse
Affiliation(s)
- Junli Zhao
- Department of Anesthesiology, Duke University Medical Center, Durham, 27710, USA.
| | - Alexus Roberts
- Department of Anesthesiology, Duke University Medical Center, Durham, 27710, USA
- Department of Biology, Duke University Medical Center, Durham, 27710, USA
| | - Zilong Wang
- Department of Anesthesiology, Duke University Medical Center, Durham, 27710, USA
| | - Justin Savage
- Department of Neurobiology, Duke University Medical Center, Durham, 27710, USA
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, Durham, 27710, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, 27710, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, 27710, USA.
| |
Collapse
|
12
|
Molecular recognition of structurally disordered Pro/Ala-rich sequences (PAS) by antibodies involves an Ala residue at the hot spot of the epitope. J Mol Biol 2021; 433:167113. [PMID: 34161780 DOI: 10.1016/j.jmb.2021.167113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 11/20/2022]
Abstract
Pro/Ala-rich sequences (PAS) are polypeptides that were developed as a biological alternative to poly-ethylene glycol (PEG) to generate biopharmaceuticals with extended plasma half-life. Like PEG, PAS polypeptides are conformationally disordered and show high solubility in water. Devoid of any charged or prominent hydrophobic side chains, these biosynthetic polymers represent an extreme case of intrinsically disordered proteins. Despite lack of immunogenicity of PAS tags in numerous animal studies we now succeeded in generating monoclonal antibodies (MAbs) against three different PAS versions. To this end, mice were immunized with a PAS#1, P/A#1 or APSA 40mer peptide conjugated to keyhole limpet hemocyanin as highly immunogenic carrier protein. In each case, one MAb with high binding activity and specificity towards a particular PAS motif was obtained. The apparent affinity was strongly dependent on the avidity effect and most pronounced for the bivalent MAb when interacting with a long PAS repeat. X-ray structural analysis of four representative anti-PAS Fab fragments in complex with their cognate PAS epitope peptides revealed interactions dominated by hydrogen bond networks involving the peptide backbone as well as multiple Van der Waals contacts arising from intimate shape complementarity. Surprisingly, Ala, the L-amino acid with the smallest side chain, emerged as a crucial feature for epitope recognition, contributing specific contacts at the center of the paratope in several anti-PAS complexes. Apart from these insights into how antibodies can recognize feature-less peptides without secondary structure, the MAbs characterized in this study offer valuable reagents for the preclinical and clinical development of PASylated biologics.
Collapse
|
13
|
Rasouli J, Casella G, Ishikawa LLW, Thome R, Boehm A, Ertel A, Melo-Silva CR, Mari ER, Porazzi P, Zhang W, Xiao D, Sigal LJ, Fortina P, Zhang GX, Rostami A, Ciric B. IFN-β Acts on Monocytes to Ameliorate CNS Autoimmunity by Inhibiting Proinflammatory Cross-Talk Between Monocytes and Th Cells. Front Immunol 2021; 12:679498. [PMID: 34149716 PMCID: PMC8213026 DOI: 10.3389/fimmu.2021.679498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/12/2021] [Indexed: 01/18/2023] Open
Abstract
IFN-β has been the treatment for multiple sclerosis (MS) for almost three decades, but understanding the mechanisms underlying its beneficial effects remains incomplete. We have shown that MS patients have increased numbers of GM-CSF+ Th cells in circulation, and that IFN-β therapy reduces their numbers. GM-CSF expression by myelin-specific Th cells is essential for the development of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. These findings suggested that IFN-β therapy may function via suppression of GM-CSF production by Th cells. In the current study, we elucidated a feedback loop between monocytes and Th cells that amplifies autoimmune neuroinflammation, and found that IFN-β therapy ameliorates central nervous system (CNS) autoimmunity by inhibiting this proinflammatory loop. IFN-β suppressed GM-CSF production in Th cells indirectly by acting on monocytes, and IFN-β signaling in monocytes was required for EAE suppression. IFN-β increased IL-10 expression by monocytes, and IL-10 was required for the suppressive effects of IFN-β. IFN-β treatment suppressed IL-1β expression by monocytes in the CNS of mice with EAE. GM-CSF from Th cells induced IL-1β production by monocytes, and, in a positive feedback loop, IL-1β augmented GM-CSF production by Th cells. In addition to GM-CSF, TNF and FASL expression by Th cells was also necessary for IL-1β production by monocyte. IFN-β inhibited GM-CSF, TNF, and FASL expression by Th cells to suppress IL-1β secretion by monocytes. Overall, our study describes a positive feedback loop involving several Th cell- and monocyte-derived molecules, and IFN-β actions on monocytes disrupting this proinflammatory loop.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Autoimmunity/drug effects
- Cell Communication/genetics
- Cell Communication/immunology
- Cytokines/metabolism
- Disease Models, Animal
- Disease Susceptibility/immunology
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis
- Interferon-beta/metabolism
- Interferon-beta/pharmacology
- Mice
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/metabolism
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
- Javad Rasouli
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Giacomo Casella
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | | | - Rodolfo Thome
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexandra Boehm
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Adam Ertel
- Sidney Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Carolina R. Melo-Silva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Elisabeth R. Mari
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patrizia Porazzi
- Sidney Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Weifeng Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Dan Xiao
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Luis J. Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Paolo Fortina
- Sidney Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Translation and Precision Medicine, Sapienza University, Rome, Italy
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
14
|
Johnson BM, Uchimura T, Gallovic MD, Thamilarasan M, Chou WC, Gibson SA, Deng M, Tam JW, Batty CJ, Williams J, Matsushima GK, Bachelder EM, Ainslie KM, Markovic-Plese S, Ting JPY. STING Agonist Mitigates Experimental Autoimmune Encephalomyelitis by Stimulating Type I IFN-Dependent and -Independent Immune-Regulatory Pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2015-2028. [PMID: 33820855 PMCID: PMC8406342 DOI: 10.4049/jimmunol.2001317] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022]
Abstract
The cGAS-cyclic GMP-AMP (cGAMP)-stimulator of IFN genes (STING) pathway induces a powerful type I IFN (IFN-I) response and is a prime candidate for augmenting immunity in cancer immunotherapy and vaccines. IFN-I also has immune-regulatory functions manifested in several autoimmune diseases and is a first-line therapy for relapsing-remitting multiple sclerosis. However, it is only moderately effective and can induce adverse effects and neutralizing Abs in recipients. Targeting cGAMP in autoimmunity is unexplored and represents a challenge because of the intracellular location of its receptor, STING. We used microparticle (MP)-encapsulated cGAMP to increase cellular delivery, achieve dose sparing, and reduce potential toxicity. In the C57BL/6 experimental allergic encephalomyelitis (EAE) model, cGAMP encapsulated in MPs (cGAMP MPs) administered therapeutically protected mice from EAE in a STING-dependent fashion, whereas soluble cGAMP was ineffective. Protection was also observed in a relapsing-remitting model. Importantly, cGAMP MPs protected against EAE at the peak of disease and were more effective than rIFN-β. Mechanistically, cGAMP MPs showed both IFN-I-dependent and -independent immunosuppressive effects. Furthermore, it induced the immunosuppressive cytokine IL-27 without requiring IFN-I. This augmented IL-10 expression through activated ERK and CREB. IL-27 and subsequent IL-10 were the most important cytokines to mitigate autoreactivity. Critically, cGAMP MPs promoted IFN-I as well as the immunoregulatory cytokines IL-27 and IL-10 in PBMCs from relapsing-remitting multiple sclerosis patients. Collectively, this study reveals a previously unappreciated immune-regulatory effect of cGAMP that can be harnessed to restrain T cell autoreactivity.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell-Derived Microparticles/immunology
- Cell-Derived Microparticles/metabolism
- Cells, Cultured
- Cytokines/immunology
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Female
- Humans
- Interferon Type I/immunology
- Interferon Type I/metabolism
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Membrane Proteins/agonists
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Mice, Knockout
- Nucleotides, Cyclic/administration & dosage
- Nucleotides, Cyclic/immunology
- Nucleotides, Cyclic/metabolism
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Mice
Collapse
Affiliation(s)
- Brandon M Johnson
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Toru Uchimura
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew D Gallovic
- Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Madhan Thamilarasan
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Wei-Chun Chou
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sara A Gibson
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Meng Deng
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Oral and Craniofacial Biomedicine Program, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jason W Tam
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Cole J Batty
- Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jonathan Williams
- Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Glenn K Matsushima
- Neuroscience Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Eric M Bachelder
- Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kristy M Ainslie
- Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Silva Markovic-Plese
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jenny P-Y Ting
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC;
- Neuroscience Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Center for Translational Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC; and
- Institute for Inflammatory Diseases, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
15
|
Yin S, Wang Y, Zhang B, Qu Y, Liu Y, Dai S, Zhang Y, Wang Y, Bi J. Engineered Human Heavy-Chain Ferritin with Half-Life Extension and Tumor Targeting by PAS and RGDK Peptide Functionalization. Pharmaceutics 2021; 13:pharmaceutics13040521. [PMID: 33918853 PMCID: PMC8070472 DOI: 10.3390/pharmaceutics13040521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/24/2021] [Accepted: 04/06/2021] [Indexed: 01/05/2023] Open
Abstract
Ferritin, one of the most investigated protein nanocages, is considered as a promising drug carrier because of its advantageous stability and safety. However, its short half-life and undesirable tumor targeting ability has limited its usage in tumor treatment. In this work, two types of functional peptides, half-life extension peptide PAS, and tumor targeting peptide RGDK (Arg-Gly-Asp-Lys), are inserted to human heavy-chain ferritin (HFn) at C-terminal through flexible linkers with two distinct enzyme cleavable sites. Structural characterizations show both HFn and engineered HFns can assemble into nanoparticles but with different apparent hydrodynamic volumes and molecular weights. RGDK peptide enhanced the internalization efficiency of HFn and showed a significant increase of growth inhibition against 4T1 cell line in vitro. Pharmacokinetic study in vivo demonstrates PAS peptides extended ferritin half-life about 4.9 times in Sprague Dawley rats. RGDK peptides greatly enhanced drug accumulation in the tumor site rather than in other organs in biodistribution analysis. Drug loaded PAS-RGDK functionalized HFns curbed tumor growth with significantly greater efficacies in comparison with drug loaded HFn.
Collapse
Affiliation(s)
- Shuang Yin
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide SA5005, Australia; (S.Y.); (B.Z.); (Y.Q.)
| | - Yan Wang
- School of Chinese Medicine and Food Engineering, Shanxi University of Traditional Chinese Medicine, Jinzhong 030619, China;
| | - Bingyang Zhang
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide SA5005, Australia; (S.Y.); (B.Z.); (Y.Q.)
| | - Yiran Qu
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide SA5005, Australia; (S.Y.); (B.Z.); (Y.Q.)
| | - Yongdong Liu
- State Key Laboratory of Biochemistry Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; (Y.L.); (Y.Z.)
| | - Sheng Dai
- Department of Chemical Engineering, Brunel University London, Uxbridge UB8 3PH, UK;
| | - Yao Zhang
- State Key Laboratory of Biochemistry Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; (Y.L.); (Y.Z.)
| | - Yingli Wang
- School of Chinese Medicine and Food Engineering, Shanxi University of Traditional Chinese Medicine, Jinzhong 030619, China;
- Correspondence: (Y.W.); (J.B.)
| | - Jingxiu Bi
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide SA5005, Australia; (S.Y.); (B.Z.); (Y.Q.)
- Correspondence: (Y.W.); (J.B.)
| |
Collapse
|
16
|
Donnelly CR, Jiang C, Andriessen AS, Wang K, Wang Z, Ding H, Zhao J, Luo X, Lee MS, Lei YL, Maixner W, Ko MC, Ji RR. STING controls nociception via type I interferon signalling in sensory neurons. Nature 2021; 591:275-280. [PMID: 33442058 PMCID: PMC7977781 DOI: 10.1038/s41586-020-03151-1] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 12/01/2020] [Indexed: 01/29/2023]
Abstract
The innate immune regulator STING is a critical sensor of self- and pathogen-derived DNA. DNA sensing by STING leads to the induction of type-I interferons (IFN-I) and other cytokines, which promote immune-cell-mediated eradication of pathogens and neoplastic cells1,2. STING is also a robust driver of antitumour immunity, which has led to the development of STING activators and small-molecule agonists as adjuvants for cancer immunotherapy3. Pain, transmitted by peripheral nociceptive sensory neurons (nociceptors), also aids in host defence by alerting organisms to the presence of potentially damaging stimuli, including pathogens and cancer cells4,5. Here we demonstrate that STING is a critical regulator of nociception through IFN-I signalling in peripheral nociceptors. We show that mice lacking STING or IFN-I signalling exhibit hypersensitivity to nociceptive stimuli and heightened nociceptor excitability. Conversely, intrathecal activation of STING produces robust antinociception in mice and non-human primates. STING-mediated antinociception is governed by IFN-Is, which rapidly suppress excitability of mouse, monkey and human nociceptors. Our findings establish the STING-IFN-I signalling axis as a critical regulator of physiological nociception and a promising new target for treating chronic pain.
Collapse
Affiliation(s)
- Christopher R Donnelly
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Amanda S Andriessen
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Kaiyuan Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Zilong Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Huiping Ding
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Junli Zhao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Xin Luo
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Michael S Lee
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Yu L Lei
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - William Maixner
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
- W.G. Hefner Veterans Affairs Medical Center, Salisbury, NC, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
17
|
Wittling MC, Cahalan SR, Levenson EA, Rabin RL. Shared and Unique Features of Human Interferon-Beta and Interferon-Alpha Subtypes. Front Immunol 2021; 11:605673. [PMID: 33542718 PMCID: PMC7850986 DOI: 10.3389/fimmu.2020.605673] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022] Open
Abstract
Type I interferons (IFN-I) were first discovered as an antiviral factor by Isaacs and Lindenmann in 1957, but they are now known to also modulate innate and adaptive immunity and suppress proliferation of cancer cells. While much has been revealed about IFN-I, it remains a mystery as to why there are 16 different IFN-I gene products, including IFNβ, IFNω, and 12 subtypes of IFNα. Here, we discuss shared and unique aspects of these IFN-I in the context of their evolution, expression patterns, and signaling through their shared heterodimeric receptor. We propose that rather than investigating responses to individual IFN-I, these contexts can serve as an alternative approach toward investigating roles for IFNα subtypes. Finally, we review uses of IFNα and IFNβ as therapeutic agents to suppress chronic viral infections or to treat multiple sclerosis.
Collapse
Affiliation(s)
| | | | | | - Ronald L. Rabin
- Division of Bacterial, Parasitic, and Allergenic Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
18
|
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: 19] [Impact Index Per Article: 4.8] [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
|
19
|
Schreiber G. The Role of Type I Interferons in the Pathogenesis and Treatment of COVID-19. Front Immunol 2020; 11:595739. [PMID: 33117408 PMCID: PMC7561359 DOI: 10.3389/fimmu.2020.595739] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Type I interferons (IFN-I) were first discovered over 60 years ago in a classical experiment by Isaacs and Lindenman, who showed that IFN-Is possess antiviral activity. Later, it became one of the first approved protein drugs using heterologous protein expression systems, which allowed its large-scale production. It has been approved, and widely used in a pleiotropy of diseases, including multiple-sclerosis, hepatitis B and C, and some forms of cancer. Preliminary clinical data has supported its effectiveness against potential pandemic pathogens such as Ebola and SARS. Still, more efficient and specific drugs have taken its place in treating such diseases. The COVID-19 global pandemic has again lifted the status of IFN-Is to become one of the more promising drug candidates, with initial clinical trials showing promising results in reducing the severity and duration of the disease. Although SARS-CoV-2 inhibits the production of IFNβ and thus obstructs the innate immune response to this virus, it is sensitive to the antiviral activity of externally administrated IFN-Is. In this review I discuss the diverse modes of biological actions of IFN-Is and how these are related to biophysical parameters of IFN-I-receptor interaction and cell-type specificity in light of the large variety of binding affinities of the different IFN-I subtypes towards the common interferon receptor. Furthermore, I discuss how these may guide the optimized use IFN-Is in combatting COVID-19.
Collapse
Affiliation(s)
- Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
20
|
Lin Y, Liu J, Bai R, Shi J, Zhu X, Liu J, Guo J, Zhang W, Liu H, Liu Z. Mitochondria-Inspired Nanoparticles with Microenvironment-Adapting Capacities for On-Demand Drug Delivery after Ischemic Injury. ACS NANO 2020; 14:11846-11859. [PMID: 32880428 DOI: 10.1021/acsnano.0c04727] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Stimuli-responsive nanoparticles (NPs), so-called "smart" NPs, possess great potentials in drug delivery. Presently, the intelligence of smart NPs is mainly based on their chemical or physical changes to stimuli, which are usually "mechanical" and fundamentally different from biological intelligence. Inspired by mitochondria (MT), a biosmart nanoparticle with microenvironment targeting and self-adaptive capacity (MTSNP) was fabricated for ischemic tissue repair. The nanoparticles were designed as shell@circular DNA@shell@core. The double shells were like the two-layered membranes of MT, the melatonin-loaded cores corresponded to the MT matrix, and the circular DNA corresponded to MTDNA. In function, melatonin-loaded cores simulated the cell-protective mechanism of MT, which naturally synthesized melatonin to resist ischemia, while circular DNA was constructed to mimic the biological oxygen-sensing mechanism, synthesizing VEGF for vascularization according to oxygen level, like the ATP supply by MT according to microenvironment demand. At the acute stage of ischemia, melatonin was rapidly released from MTSNP to scavenge reactive oxygen species and activated melatonin receptor I on MT to prevent cytochrome c release, which would activate apoptosis. During the chronic stage, circular DNA could sense hypoxia and actively secrete VEGF for revascularization as a response. Importantly, circular DNA could also receive feedback of revascularization and shut down VEGF secretion as an adverse response. Then, the therapeutic potentials of the MTSNP were verified in myocardial ischemia by the multimodality of the methods. Such nanoparticles may represent a promising intelligent nanodrug system.
Collapse
Affiliation(s)
- Yanxia Lin
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
- Department of Cardiology, The Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Jianfeng Liu
- Department of Cardiology, The Second Medical Center of PLA General Hospital, Beijing 100853, China
| | - Rui Bai
- Department of Cardiology, The Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Jinmiao Shi
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Xiaoming Zhu
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Jian Liu
- Department of Nuclear Medicine, The First Medical Center of PLA General Hospital, Beijing 100853, China
| | - Jing Guo
- Department of Cardiology, The Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Wei Zhang
- Department of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Huiliang Liu
- Department of Cardiology, The Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Zhiqiang Liu
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| |
Collapse
|
21
|
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: 0.6] [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
|
22
|
Powers NE, Swartzwelter B, Marchetti C, de Graaf DM, Lerchner A, Schlapschy M, Datar R, Binder U, Edwards CK, Skerra A, Dinarello CA. PASylation of IL-1 receptor antagonist (IL-1Ra) retains IL-1 blockade and extends its duration in mouse urate crystal-induced peritonitis. J Biol Chem 2020; 295:868-882. [PMID: 31819009 DOI: 10.1074/jbc.ra119.010340] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/11/2019] [Indexed: 01/21/2023] Open
Abstract
Interleukin-1 (IL-1) is a key mediator of inflammation and immunity. Naturally-occurring IL-1 receptor antagonist (IL-1Ra) binds and blocks the IL-1 receptor-1 (IL-1R1), preventing signaling. Anakinra, a recombinant form of IL-1Ra, is used to treat a spectrum of inflammatory diseases. However, anakinra is rapidly cleared from the body and requires daily administration. To create a longer-lasting alternative, PASylated IL-1Ra (PAS-IL-1Ra) has been generated by in-frame fusion of a long, defined-length, N-terminal Pro/Ala/Ser (PAS) random-coil polypeptide with IL-1Ra. Here, we compared the efficacy of two PAS-IL-1Ra molecules, PAS600-IL-1Ra and PAS800-IL-1Ra (carrying 600 and 800 PAS residues, respectively), with that of anakinra in mice. PAS600-IL-1Ra displayed markedly extended blood plasma levels 3 days post-administration, whereas anakinra was undetectable after 24 h. We also studied PAS600-IL-1Ra and PAS800-IL-1Ra for efficacy in monosodium urate (MSU) crystal-induced peritonitis. 5 days post-administration, PAS800-IL-1Ra significantly reduced leukocyte influx and inflammatory markers in MSU-induced peritonitis, whereas equimolar anakinra administered 24 h before MSU challenge was ineffective. The 6-h pretreatment with equimolar anakinra or PAS800-IL-1Ra before MSU challenge similarly reduced inflammatory markers. In cultured A549 lung carcinoma cells, anakinra, PAS600-IL-1Ra, and PAS800-IL-Ra reduced IL-1α-induced IL-6 and IL-8 levels with comparable potency. In human peripheral blood mononuclear cells, these molecules suppressed Candida albicans-induced production of the cancer-promoting cytokine IL-22. Surface plasmon resonance analyses revealed significant binding between PAS-IL-1Ra and IL-1R1, although with a slightly lower affinity than anakinra. These results validate PAS-IL-1Ra as an active IL-1 antagonist with marked in vivo potency and a significantly extended half-life compared with anakinra.
Collapse
Affiliation(s)
- Nicholas E Powers
- Department of Medicine, University of Colorado, Aurora, Colorado 80045
| | | | - Carlo Marchetti
- Department of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Dennis M de Graaf
- Department of Medicine, University of Colorado, Aurora, Colorado 80045.,Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | | | - Martin Schlapschy
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
| | - Rajiv Datar
- DNX Biopharmaceuticals, Inc., San Diego, California 92121
| | - Uli Binder
- XL-protein GmbH, Lise-Meitner-Strasse 30, 85354 Freising, Germany
| | - Carl K Edwards
- DNX Biopharmaceuticals, Inc., San Diego, California 92121
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
| | - Charles A Dinarello
- Department of Medicine, University of Colorado, Aurora, Colorado 80045.,Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
23
|
Powers NE, Swartzwelter B, Marchetti C, de Graaf DM, Lerchner A, Schlapschy M, Datar R, Binder U, Edwards CK, Skerra A, Dinarello CA. PASylation of IL-1 receptor antagonist (IL-1Ra) retains IL-1 blockade and extends its duration in mouse urate crystal-induced peritonitis. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49941-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
24
|
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: 190] [Impact Index Per Article: 38.0] [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
|
25
|
Tan H, Su W, Zhang W, Wang P, Sattler M, Zou P. Recent Advances in Half-life Extension Strategies for Therapeutic Peptides and Proteins. Curr Pharm Des 2019; 24:4932-4946. [PMID: 30727869 DOI: 10.2174/1381612825666190206105232] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/26/2019] [Indexed: 12/16/2022]
Abstract
Peptides and proteins are two classes of molecules with attractive possibilities for therapeutic applications. However, the bottleneck for the therapeutic application of many peptides and proteins is their short halflives in vivo, typically just a few minutes to hours. Half-life extension strategies have been extensively studied and many of them have been proven to be effective in the generation of long-acting therapeutics with improved pharmacokinetic and pharmacodynamic properties. In this review, we summarize the recent advances in half-life extension strategies, illustrate their potential applications and give some examples, highlighting the strategies that have been used in approved drugs and for drugs in clinical trials. Meanwhile, several novel strategies that are still in the process of discovery or at a preclinical stage are also introduced. In these strategies, the two most frequently used half-life extension methods are the reduction in the rate of renal clearance or the exploitation of the recycling mechanism of FcRn by binding to the albumin or IgG-Fc. Here, we discuss half-life extension strategies of recombinant therapeutic protein via genetic fusion, rather than chemical conjugation such as PEGylation. With the rapid development of genetic engineering and protein engineering, novel strategies for half-life extension have been emerged consistently. Some of these will be evaluated in clinical trials and may become viable alternatives to current strategies for making next-generation biodrugs.
Collapse
Affiliation(s)
- Huanbo Tan
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Wencheng Su
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Wenyu Zhang
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Pengju Wang
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Michael Sattler
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.,Institute of Structural Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Center for Integrated Protein Science Munich at Chair Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, Garching, Germany
| | - Peijian Zou
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.,Institute of Structural Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Center for Integrated Protein Science Munich at Chair Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, Garching, Germany
| |
Collapse
|
26
|
Iyengar ARS, Gupta S, Jawalekar S, Pande AH. Protein Chimerization: A New Frontier for Engineering Protein Therapeutics with Improved Pharmacokinetics. J Pharmacol Exp Ther 2019; 370:703-714. [PMID: 31010843 DOI: 10.1124/jpet.119.257063] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/16/2019] [Indexed: 03/08/2025] Open
Abstract
With the advancement of medicine, the utility of protein therapeutics is increasing exponentially. However, a significant number of protein therapeutics suffer from grave limitations, which include their subpar pharmacokinetics. In this study, we have reviewed the emerging field of protein chimerization for improving the short circulatory half-life of protein therapeutics. We have discussed various aspects of protein therapeutics aiming at their mechanism of clearance and various approaches used to increase their short circulatory half-life with principal focus on the concept of chimerization. Furthermore, we have comprehensively reviewed various components of chimera, such as half-life extension partners and linkers, their shortcomings, and prospective work to be undertaken for developing effective chimeric protein therapeutics.
Collapse
Affiliation(s)
- A R Satvik Iyengar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| | - Shreya Gupta
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| | - Snehal Jawalekar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| |
Collapse
|
27
|
Surface protein engineering increases the circulation time of a cell membrane-based nanotherapeutic. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 18:169-178. [DOI: 10.1016/j.nano.2019.02.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/31/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023]
|
28
|
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: 20] [Impact Index Per Article: 3.3] [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
|
29
|
Lee S, Son WS, Yang HB, Rajasekaran N, Kim SS, Hong S, Choi JS, Choi JY, Song K, Shin YK. A Glycoengineered Interferon-β Mutein (R27T) Generates Prolonged Signaling by an Altered Receptor-Binding Kinetics. Front Pharmacol 2019; 9:1568. [PMID: 30733680 PMCID: PMC6353837 DOI: 10.3389/fphar.2018.01568] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 12/24/2018] [Indexed: 12/15/2022] Open
Abstract
The glycoengineering approach is used to improve biophysical properties of protein-based drugs, but its direct impact on binding affinity and kinetic properties for the glycoengineered protein and its binding partner interaction is unclear. Type I interferon (IFN) receptors, composed of IFNAR1 and IFNAR2, have different binding strengths, and sequentially bind to IFN in the dominant direction, leading to activation of signals and induces a variety of biological effects. Here, we evaluated receptor-binding kinetics for each state of binary and ternary complex formation between recombinant human IFN-β-1a and the glycoengineered IFN-β mutein (R27T) using the heterodimeric Fc-fusion technology, and compared biological responses between them. Our results have provided evidence that the additional glycan of R27T, located at the binding interface of IFNAR2, destabilizes the interaction with IFNAR2 via steric hindrance, and simultaneously enhances the interaction with IFNAR1 by restricting the conformational freedom of R27T. Consequentially, altered receptor-binding kinetics of R27T in the ternary complex formation led to a substantial increase in strength and duration of biological responses such as prolonged signal activation and gene expression, contributing to enhanced anti-proliferative activity. In conclusion, our findings reveal N-glycan at residue 25 of R27T is a crucial regulator of receptor-binding kinetics that changes biological activities such as long-lasting activation. Thus, we believe that R27T may be clinically beneficial for patients with multiple sclerosis.
Collapse
Affiliation(s)
- Saehyung Lee
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Woo Sung Son
- Department of Pharmacy, College of Pharmacy, CHA University, Pocheon, South Korea
| | - Ho Bin Yang
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Nirmal Rajasekaran
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Sung-Su Kim
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul, South Korea
| | - Sungyoul Hong
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, South Korea
| | | | - Kyoung Song
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul, South Korea
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea.,Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Suwon, South Korea
| |
Collapse
|
30
|
Xia Y, Schlapschy M, Morath V, Roeder N, Vogt EI, Stadler D, Cheng X, Dittmer U, Sutter K, Heikenwalder M, Skerra A, Protzer U. PASylated interferon α efficiently suppresses hepatitis B virus and induces anti-HBs seroconversion in HBV-transgenic mice. Antiviral Res 2019; 161:134-143. [DOI: 10.1016/j.antiviral.2018.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/22/2018] [Accepted: 11/06/2018] [Indexed: 01/05/2023]
|
31
|
Gebauer M, Skerra A. Prospects of PASylation® for the design of protein and peptide therapeutics with extended half-life and enhanced action. Bioorg Med Chem 2018; 26:2882-2887. [DOI: 10.1016/j.bmc.2017.09.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 11/28/2022]
|
32
|
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: 5.3] [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
|
33
|
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: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
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: 1.8] [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
|
35
|
Abstract
Type I interferons (IFN-1) are cytokines that affect the expression of thousands of genes, resulting in profound cellular changes. IFN-1 activates the cell by dimerizing its two-receptor chains, IFNAR1 and IFNAR2, which are expressed on all nucleated cells. Despite a similar mode of binding, the different IFN-1s activate a spectrum of activities. The causes for differential activation may stem from differences in IFN-1-binding affinity, duration of binding, number of surface receptors, induction of feedbacks, and cell type-specific variations. All together these will alter the signal that is transmitted from the extracellular domain inward. The intracellular domain binds, directly or indirectly, different effector proteins that transmit signals. The composition of effector molecules deviates between different cell types and tissues, inserting an additional level of complexity to the system. Moreover, IFN-1s do not act on their own, and clearly there is much cross-talk between the activated effector molecules by IFN-1 and other cytokines. The outcome generated by all of these factors (processing step) is an observed phenotype, which can be the transformation of the cell to an antiviral state, differentiation of the cell to a specific immune cell, senescence, apoptosis, and many more. IFN-1 activities can be divided into robust and tunable. Antiviral activity, which is stimulated by minute amounts of IFN-1 and is common to all cells, is termed robust. The other activities, which we term tunable, are cell type-specific and often require more stringent modes of activation. In this review, I summarize the current knowledge on the mode of activation and processing that is initiated by IFN-1, in perspective of the resulting phenotypes.
Collapse
Affiliation(s)
- Gideon Schreiber
- From the Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
36
|
Control of Hepatitis B Virus by Cytokines. Viruses 2017; 9:v9010018. [PMID: 28117695 PMCID: PMC5294987 DOI: 10.3390/v9010018] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) infection remains a major public health problem worldwide with more than 240 million individuals chronically infected. Current treatments can control HBV replication to a large extent, but cannot eliminate HBV infection. Cytokines have been shown to control HBV replication and contribute to HBV cure in different models. Cytokines play an important role in limiting acute HBV infection in patients and mediate a non-cytolytic clearance of the virus. In this review, we summarize the effects of cytokines and cytokine-induced cellular signaling pathways on different steps of the HBV life cycle, and discuss possible strategies that may contribute to the eradication of HBV through innate immune activation.
Collapse
|
37
|
PASylation technology improves recombinant interferon-β1b solubility, stability, and biological activity. Appl Microbiol Biotechnol 2016; 101:1975-1987. [DOI: 10.1007/s00253-016-7944-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/25/2016] [Accepted: 10/13/2016] [Indexed: 10/20/2022]
|
38
|
Kuhn N, Schmidt CQ, Schlapschy M, Skerra A. PASylated Coversin, a C5-Specific Complement Inhibitor with Extended Pharmacokinetics, Shows Enhanced Anti-Hemolytic Activity in Vitro. Bioconjug Chem 2016; 27:2359-2371. [PMID: 27598771 DOI: 10.1021/acs.bioconjchem.6b00369] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Ornithodoros moubata Complement Inhibitor (OmCI) binds complement component 5 (C5) with high affinity and, thus, selectively prevents proteolytic activation of the terminal lytic complement pathway. A recombinant version of OmCI (also known as Coversin and rEV576) has proven efficacious in several animal models of complement-mediated diseases and successfully completed a phase Ia clinical trial. Coversin is a small 17 kDa lipocalin protein which has a very short plasma half-life if not bound to C5; therefore, the drug requires frequent dosing. We have improved the pharmacokinetics of Coversin by N-terminal translational conjugation with a 600 residue polypeptide composed of Pro, Ala, and Ser (PAS) residues. To this end, PAS-Coversin as well as the unmodified Coversin were functionally expressed in the cytoplasm of E. coli and purified to homogeneity. Both versions showed identical affinity to human C5, as determined by surface plasmon resonance measurements, and revealed similar complement inhibitory activity, as measured in ELISAs with human serum. In line with the PEG-like biophysical properties, PASylation dramatically prolonged the plasma half-life of uncomplexed Coversin by a factor ≥50 in mice. In a clinically relevant in vitro model of the complement-mediated disease paroxysmal nocturnal hemoglobinuria (PNH) both versions of Coversin effectively reduced erythrocyte lysis. Unexpectedly, while the IC50 values were comparable, PAS-Coversin reached a substantially lower plateau of residual lysis at saturating inhibitor concentrations. Taken together, our data demonstrate two clinically relevant improvements of PASylated Coversin: markedly increased plasma half-life and considerably reduced background hemolysis of erythrocytes with PNH-induced phenotype.
Collapse
Affiliation(s)
- Nadine Kuhn
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München , Emil-Erlenmeyer-Forum 5, 85354 Freising (Weihenstephan), Germany
| | - Christoph Q Schmidt
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University , Helmholtzstrasse 20, 89081 Ulm, Germany
| | - Martin Schlapschy
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München , Emil-Erlenmeyer-Forum 5, 85354 Freising (Weihenstephan), Germany.,XL-protein GmbH , Lise-Meitner-Strasse 30, 85354 Freising, Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München , Emil-Erlenmeyer-Forum 5, 85354 Freising (Weihenstephan), Germany.,XL-protein GmbH , Lise-Meitner-Strasse 30, 85354 Freising, Germany
| |
Collapse
|
39
|
Bolze F, Bast A, Mocek S, Morath V, Yuan D, Rink N, Schlapschy M, Zimmermann A, Heikenwalder M, Skerra A, Klingenspor M. Treatment of diet-induced lipodystrophic C57BL/6J mice with long-acting PASylated leptin normalises insulin sensitivity and hepatic steatosis by promoting lipid utilisation. Diabetologia 2016; 59:2005-12. [PMID: 27272237 DOI: 10.1007/s00125-016-4004-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/11/2016] [Indexed: 10/21/2022]
Abstract
AIMS/HYPOTHESIS Recombinant leptin offers a viable treatment for lipodystrophy (LD) syndromes. However, due to its short plasma half-life, leptin replacement therapy requires at least daily subcutaneous (s.c.) injections. Here, we optimised this treatment strategy in LD mice by using a novel leptin version with extended plasma half-life using PASylation technology. METHODS A long-acting leptin version was prepared by genetic fusion with a 600 residue polypeptide made of Pro, Ala and Ser (PASylation), which enlarges the hydrodynamic volume and, thus, retards renal filtration, allowing less frequent injection. LD was induced in C57BL/6J mice by feeding a diet supplemented with conjugated linoleic acid (CLA). Chronic and acute effects of leptin treatment were assessed by evaluating plasma insulin levels, insulin tolerance, histological liver sections, energy expenditure, energy intake and body composition. RESULTS In a cohort of female mice, 4 nmol PAS-leptin (applied via four s.c. injections every 3 days) successfully alleviated the CLA-induced LD phenotype, which was characterised by hyperinsulinaemia, insulin intolerance and hepatosteatosis. The same injection regimen had no measurable effect when unmodified recombinant leptin was administered at an equivalent dose. In a cohort of LD males, a single s.c. injection of PAS-leptin did not affect energy expenditure but inhibited food intake and promoted a shift in fuel selection towards preferential fat oxidation, which mechanistically substantiates the metabolic improvements. CONCLUSIONS/INTERPRETATION The excellent pharmacological properties render PASylated leptin an agent of choice for refining both animal studies and therapeutic strategies in the context of LD syndromes and beyond.
Collapse
Affiliation(s)
- Florian Bolze
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Andrea Bast
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Sabine Mocek
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Volker Morath
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354, Freising (Weihenstephan), Germany
| | - Detian Yuan
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Rink
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Martin Schlapschy
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354, Freising (Weihenstephan), Germany
| | - Anika Zimmermann
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354, Freising (Weihenstephan), Germany.
- XL-protein GmbH, Freising, Germany.
| | - Martin Klingenspor
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany.
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany.
| |
Collapse
|
40
|
Zhang P, Sun F, Liu S, Jiang S. Anti-PEG antibodies in the clinic: Current issues and beyond PEGylation. J Control Release 2016; 244:184-193. [PMID: 27369864 DOI: 10.1016/j.jconrel.2016.06.040] [Citation(s) in RCA: 441] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/15/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022]
Abstract
The technique of attaching the polymer polyethylene glycol (PEG), or PEGylation, has brought more than ten protein drugs into market. The surface conjugation of PEG on proteins prolongs their blood circulation time and reduces immunogenicity by increasing their hydrodynamic size and masking surface epitopes. Despite this success, an emerging body of literature highlights the presence of antibodies produced by the immune system that specifically recognize and bind to PEG (anti-PEG Abs), including both pre-existing and treatment-induced Abs. More importantly, the existence of anti-PEG Abs has been correlated with loss of therapeutic efficacy and increase in adverse effects in several clinical reports examining different PEGylated therapeutics. To better understand the nature of anti-PEG immunity, we summarize a number of clinical reports and some critical animal studies regarding pre-existing and treatment-induced anti-PEG Abs. Various anti-PEG detection methods used in different studies were provided. Several protein modification technologies beyond PEGylation were also highlighted.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, United States
| | - Fang Sun
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, United States
| | - Sijun Liu
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
| | - Shaoyi Jiang
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, United States; Department of Bioengineering, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
41
|
Abstract
INTRODUCTION Many of the biotherapeutics approved or under development suffer from a short half-life necessitating frequent applications in order to maintain a therapeutic concentration over an extended period of time. The implementation of half-life extension strategies allows the generation of long-lasting therapeutics with improved pharmacokinetic and pharmacodynamic properties. AREAS COVERED This review gives an overview of the different half-life extension strategies developed over the past years and their application to generate next-generation biotherapeutics. It focuses on srategies already used in approved drugs and drugs that are in clinical development. These strategies include those aimed at increasing the hydrodynamic radius of the biotherapeutic and strategies which further implement recycling by the neonatal Fc receptor (FcRn). EXPERT OPINION Half-life extension strategies have become an integral part of development for many biotherapeutics. A diverse set of these strategies is available for the fine-tuning of half-life and adaption to the intended treatment modality and disease. Currently, half-life extension is dominated by strategies utilizing albumin binding or fusion, fusion to an immunoglobulin Fc region and PEGylation. However, a variety of alternative strategies, such as fusion of flexible polypeptide chains as PEG mimetic substitute, have reached advanced stages and offer further alternatives for half-life extension.
Collapse
Affiliation(s)
- Roland E Kontermann
- a Institute of Cell Biology and Immunology , University of Stuttgart , Stuttgart , Germany
| |
Collapse
|
42
|
Bolze F, Morath V, Bast A, Rink N, Schlapschy M, Mocek S, Skerra A, Klingenspor M. Long-Acting PASylated Leptin Ameliorates Obesity by Promoting Satiety and Preventing Hypometabolism in Leptin-Deficient Lep(ob/ob) Mice. Endocrinology 2016; 157:233-44. [PMID: 26492472 DOI: 10.1210/en.2015-1519] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Body weight loss of Lep(ob/ob) mice in response to leptin is larger than expected from the reduction in energy intake alone, suggesting a thermogenic action of unknown magnitude. We exploited the superior pharmacological properties of a novel long-acting leptin prepared via PASylation to study the contribution of its anorexigenic and thermogenic effects. PASylation, the genetic fusion of leptin with a conformationally disordered polypeptide comprising 600 Pro/Ala/Ser (PAS) residues, provides a superior way to increase the hydrodynamic volume of the fusion protein, thus retarding kidney filtration and extending plasma half-life. Here a single PAS(600)-leptin injection (300 pmol/g) resulted in a maximal weight reduction of 21% 6 days after application. The negative energy balance of 300 kJ/(4 d) was driven by a decrease in energy intake, whereas energy expenditure remained stable. Mice that were food restricted to the same extent showed an energy deficit of only 220 kJ/(4 d) owing to recurring torpor bouts. Therefore, the anorexigenic effect of PAS(600)-leptin contributes 75% to weight loss, whereas the thermogenic action accounts for 25% by preventing hypometabolism. In a second experiment, just four injections of PAS(600)-leptin (100 pmol/g) administered in 5- to 6-day intervals rectified the Lep(ob/ob) phenotype. In total, 16 nmol of PAS(600)-leptin per mouse triggered a weight loss of 43% within 20 days and normalized hypothermia and glucose homeostasis as well as hepatic steatosis. The beneficial properties of PAS(600)-leptin are substantiated by a comparison with previous studies in which approximately 400 nmol (∼25-fold) unmodified leptin was mandatory to achieve similar improvements.
Collapse
Affiliation(s)
- Florian Bolze
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Volker Morath
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Andrea Bast
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Nadine Rink
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Martin Schlapschy
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Sabine Mocek
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Arne Skerra
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Martin Klingenspor
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| |
Collapse
|
43
|
Falvo E, Tremante E, Arcovito A, Papi M, Elad N, Boffi A, Morea V, Conti G, Toffoli G, Fracasso G, Giacomini P, Ceci P. Improved Doxorubicin Encapsulation and Pharmacokinetics of Ferritin-Fusion Protein Nanocarriers Bearing Proline, Serine, and Alanine Elements. Biomacromolecules 2015; 17:514-22. [PMID: 26686226 DOI: 10.1021/acs.biomac.5b01446] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A novel human ferritin-based nanocarrier, composed of 24 modified monomers able to auto-assemble into a modified protein cage, was produced and used as selective carrier of anti-tumor payloads. Each modified monomer derives from the genetic fusion of two distinct modules, namely the heavy chain of human ferritin (HFt) and a stabilizing/protective PAS polypeptide sequence rich in proline (P), serine (S), and alanine (A) residues. Two genetically fused protein constructs containing PAS polymers with 40- and 75-residue lengths, respectively, were compared. They were produced and purified as recombinant proteins in Escherichia coli at high yields. Both preparations were highly soluble and stable in vitro as well as in mouse plasma. Size-exclusion chromatography, dynamic light scattering, and transmission electron microscopy results indicated that PASylated ferritins are fully assembled and highly monodispersed. In addition, yields and stability of encapsulated doxorubicin were significantly better for both HFt-PAS proteins than for wild-type HFt. Importantly, PAS sequences considerably prolonged the half-life of HFt in the mouse bloodstream. Finally, our doxorubicin-loaded nanocages preserved the pharmacological activity of the drug. Taken together, these results indicate that both of the developed HFt-PAS fusion proteins are promising nanocarriers for future applications in cancer therapy.
Collapse
Affiliation(s)
- Elisabetta Falvo
- Institute of Molecular Biology and Pathology CNR, National Research Council of Italy , 00185 Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", University "Sapienza" , 00185 Rome, Italy
| | - Elisa Tremante
- Regina Elena National Cancer Institute , 00144 Rome, Italy
| | - Alessandro Arcovito
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore , Largo F. Vito 1, 00168 Rome, Italy
| | - Massimiliano Papi
- Istituto di Fisica, Università Cattolica del Sacro Cuore , Largo F. Vito 1, 00168, Rome, Italy
| | - Nadav Elad
- Department of Chemical Research Support, Weizmann Institute of Science , Rehovot 7610001, Israel
| | - Alberto Boffi
- Institute of Molecular Biology and Pathology CNR, National Research Council of Italy , 00185 Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", University "Sapienza" , 00185 Rome, Italy.,Center for Life Nano Science at Sapienza, Istituto Italiano di Tecnologia (IIT) , 00161 Rome, Italy
| | - Veronica Morea
- Institute of Molecular Biology and Pathology CNR, National Research Council of Italy , 00185 Rome, Italy
| | - Giamaica Conti
- Department of Neurological, Biomedical and Movement Sciences, University of Verona , 37134 Verona, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute , 33081Aviano (Pordenone), Italy
| | - Giulio Fracasso
- Department of Medicine, University of Verona , 37134 Verona, Italy
| | | | - Pierpaolo Ceci
- Institute of Molecular Biology and Pathology CNR, National Research Council of Italy , 00185 Rome, Italy
| |
Collapse
|
44
|
Qi Y, Chilkoti A. Protein-polymer conjugation-moving beyond PEGylation. Curr Opin Chem Biol 2015; 28:181-93. [PMID: 26356631 DOI: 10.1016/j.cbpa.2015.08.009] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/05/2015] [Accepted: 08/18/2015] [Indexed: 01/09/2023]
Abstract
In this review, we summarize-from a materials science perspective-the current state of the field of polymer conjugates of peptide and protein drugs, with a focus on polymers that have been developed as alternatives to the current gold standard, poly(ethylene glycol) (PEG). PEGylation, or the covalent conjugation of PEG to biological therapeutics to improve their therapeutic efficacy by increasing their circulation half-lives and stability, has been the gold standard in the pharmaceutical industry for several decades. After years of research and development, the limitations of PEG, specifically its non-degradability and immunogenicity have become increasingly apparent. While PEG is still currently the best polymer available with the longest clinical track record, extensive research is underway to develop alternative materials in an effort to address these limitations of PEG. Many of these alternative materials have shown promise, though most of them are still in an early stage of development and their in vivo distribution, mechanism of degradation, route of elimination and immunogenicity have not been investigated to a similar extent as for PEG. Thus, further in-depth in vivo testing is essential to validate whether any of the alternative materials discussed in this review qualify as a replacement for PEG.
Collapse
Affiliation(s)
- Yizhi Qi
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Biologically Inspired Materials and Materials Systems, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
45
|
Abstract
The purpose of making a "biobetter" biologic is to improve on the salient characteristics of a known biologic for which there is, minimally, clinical proof of concept or, maximally, marketed product data. There already are several examples in which second-generation or biobetter biologics have been generated by improving the pharmacokinetic properties of an innovative drug, including Neulasta(®) [a PEGylated, longer-half-life version of Neupogen(®) (filgrastim)] and Aranesp(®) [a longer-half-life version of Epogen(®) (epoetin-α)]. This review describes the use of protein fusion technologies such as Fc fusion proteins, fusion to human serum albumin, fusion to carboxy-terminal peptide, and other polypeptide fusion approaches to make biobetter drugs with more desirable pharmacokinetic profiles.
Collapse
Affiliation(s)
- William R Strohl
- Janssen BioTherapeutics, Janssen Research and Development, LLC, Pharmaceutical Companies of Johnson & Johnson, SH31-21757, 1400 Welsh and McKean Roads, PO Box 776, Spring House, PA, 19477, USA,
| |
Collapse
|
46
|
Urin V, Levin D, Sharma N, Harari D, Schreiber G. Fine Tuning of a Type 1 Interferon Antagonist. PLoS One 2015; 10:e0130797. [PMID: 26158644 PMCID: PMC4497658 DOI: 10.1371/journal.pone.0130797] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/25/2015] [Indexed: 11/18/2022] Open
Abstract
Type I interferons are multi-potent cytokines that serve as first line of defense against viruses and other pathogens, posses immunomudolatory functions and elicit a growth inhibitory response. In recent years it has been shown that interferons are also detrimental, for example in lupus, AIDS, tuberculosis and cognitive decline, highlighted the need to develop interferon antagonists. We have previously developed the antagonist IFN-1ant, with much reduced binding to the IFNAR1 receptor and enhanced binding to IFNAR2. Here, we further tune the IFN-1ant by producing three additional antagonists based on IFN-1ant but with altered activity profiles. We show that in all three cases the antiproliferative activity of interferons is blocked and the induction of gene transcription of immunomudolatory and antiproliferative associated genes are substantially decreased. Conversely, each of the new antagonists elicits a different degree of antiviral response, STAT phosphorylation and related gene induction. Two of the new antagonists promote decreased activity in relation to the original IFN-1ant, while one of them promotes increased activity. As we do not know the exact causes of the detrimental effects of IFNs, the four antagonists that were produced and analyzed provide the opportunity to investigate the extent of antagonistic and agonistic activity optimal for a given condition.
Collapse
Affiliation(s)
- Victoria Urin
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Doron Levin
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Nanaocha Sharma
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Daniel Harari
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Gideon Schreiber
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, 76100, Israel
- * E-mail:
| |
Collapse
|
47
|
Morath V, Bolze F, Schlapschy M, Schneider S, Sedlmayer F, Seyfarth K, Klingenspor M, Skerra A. PASylation of Murine Leptin Leads to Extended Plasma Half-Life and Enhanced in Vivo Efficacy. Mol Pharm 2015; 12:1431-42. [PMID: 25811325 DOI: 10.1021/mp5007147] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Leptin plays a central role in the control of energy homeostasis and appetite and, thus, has attracted attention for therapeutic approaches in spite of its limited pharmacological activity owing to the very short circulation in the body. To improve drug delivery and prolong plasma half-life, we have fused murine leptin with Pro/Ala/Ser (PAS) polypeptides of up to 600 residues, which adopt random coil conformation with expanded hydrodynamic volume in solution and, consequently, retard kidney filtration in a similar manner as polyethylene glycol (PEG). Relative to unmodified leptin, size exclusion chromatography and dynamic light scattering revealed an approximately 21-fold increase in apparent size and a much larger molecular diameter of around 18 nm for PAS(600)-leptin. High receptor-binding activity for all PASylated leptin versions was confirmed in BIAcore measurements and cell-based dual-luciferase assays. Pharmacokinetic studies in mice revealed a much extended plasma half-life after ip injection, from 26 min for the unmodified leptin to 19.6 h for the PAS(600) fusion. In vivo activity was investigated after single ip injection of equimolar doses of each leptin version. Strongly increased and prolonged hypothalamic STAT3 phosphorylation was detected for PAS(600)-leptin. Also, a reduction in daily food intake by up to 60% as well as loss in body weight of >10% lasting for >5 days was observed, whereas unmodified leptin was merely effective for 1 day. Notably, application of a PASylated superactive mouse leptin antagonist (SMLA) led to the opposite effects. Thus, PASylated leptin not only provides a promising reagent to study its physiological role in vivo but also may offer a superior drug candidate for clinical therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Arne Skerra
- ∥XL-protein GmbH, Lise-Meitner-Strasse 30, 85354 Freising, Germany
| |
Collapse
|
48
|
Wang Y, Cao L, Xu LM, Cao FF, Peng B, Zhang X, Shen YF, Uzan G, Zhang DH. Celastrol Ameliorates EAE Induction by Suppressing Pathogenic T Cell Responses in the Peripheral and Central Nervous Systems. J Neuroimmune Pharmacol 2015; 10:506-16. [PMID: 25773257 DOI: 10.1007/s11481-015-9598-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/04/2015] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis (MS) is the prototypical inflammatory demyelinating disease of the central nervous system (CNS), and MS results in physical and cognitive impairments, such as fatigue, pain, depression and bladder dysfunction. Though many therapies for MS have been developed, the safety profile and effectiveness of these therapies still need to be defined. Thus, new therapies for MS must be explored. Celastrol, a quinonemethide triterpene, is a pharmacologically active compound present in Thunder God Vine root extracts used to treat inflammatory and autoimmune diseases. Molecular studies have identified several molecular targets, which are mostly centered on the inhibition of IKK-NF-κB signaling. The animal model of experimental autoimmune encephalomyelitis (EAE) has been widely used in MS studies; thus, we tried to explore the role of celastrol in EAE development in this study. We demonstrated that the intraperitoneal injection of celastrol significantly attenuated EAE disease. Th17 cell responses in the peripheral lymph nodes in EAE mice were also inhibited by celastrol. We determined that celastroldownregulated cytokine production in bone-marrow derived dendritic cells (BMDCs). Accordingly, T cells that were co-cultured with either BMDCs pre-treated with celastrolor splenic DCs and then collected on day 7 after EAE immunizationshowed that Th17 cell polarization is suppressed in the above two situations. Moreover, celastrol was required for tissue-infiltrating DCs to sustain Th17 responses in the central nervous system (CNS). Taken together, the results of our study demonstrate that celastrol ameliorates EAE development by suppressing pathogenic Th17 responses; this finding offers a better understanding of the role of celastrol in EAE development as well as new proposals for clinical interventions.
Collapse
Affiliation(s)
- Ying Wang
- Sino-French Cooperative Central Lab, Shanghai Gongli Hospital, Secondary Military Medical University, 219 Miao Pu Road, Pudong New District, Shanghai, 200135, China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Harari D, Orr I, Rotkopf R, Baranzini SE, Schreiber G. A robust type I interferon gene signature from blood RNA defines quantitative but not qualitative differences between three major IFN drugs in the treatment of multiple sclerosis. Hum Mol Genet 2015; 24:3192-205. [DOI: 10.1093/hmg/ddv071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/16/2015] [Indexed: 01/12/2023] Open
|
50
|
|