1
|
Shetty S, Alvarado PC, Pettie D, Collier JH. Next-Generation Vaccine Development with Nanomaterials: Recent Advances, Possibilities, and Challenges. Annu Rev Biomed Eng 2024; 26:273-306. [PMID: 38959389 DOI: 10.1146/annurev-bioeng-110122-124359] [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] [Indexed: 07/05/2024]
Abstract
Nanomaterials are becoming important tools for vaccine development owing to their tunable and adaptable nature. Unique properties of nanomaterials afford opportunities to modulate trafficking through various tissues, complement or augment adjuvant activities, and specify antigen valency and display. This versatility has enabled recent work designing nanomaterial vaccines for a broad range of diseases, including cancer, inflammatory diseases, and various infectious diseases. Recent successes of nanoparticle vaccines during the coronavirus disease 2019 (COVID-19) pandemic have fueled enthusiasm further. In this review, the most recent developments in nanovaccines for infectious disease, cancer, inflammatory diseases, allergic diseases, and nanoadjuvants are summarized. Additionally, challenges and opportunities for clinical translation of this unique class of materials are discussed.
Collapse
Affiliation(s)
- Shamitha Shetty
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Pablo Cordero Alvarado
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Deleah Pettie
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| |
Collapse
|
2
|
Cohen JR, Brych SR, Prabhu S, Bi V, Elbaradei A, Tokuda JM, Xiang C, Hokom M, Cui X, Ly C, Amos N, Sun J, Calamba D, Herskovitz J, Capili A, Nourbakhsh K, Merlo A, Carreon J, Wypych J, Narhi LO, Jawa V, Joubert MK. A High Threshold of Biotherapeutic Aggregate Numbers is Needed to Induce an Immunogenic Response In Vitro, In Vivo, and in the Clinic. Pharm Res 2024; 41:651-672. [PMID: 38519817 DOI: 10.1007/s11095-024-03678-2] [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: 09/29/2023] [Accepted: 02/15/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND AND PURPOSE There is concern that subvisible aggregates in biotherapeutic drug products pose a risk to patient safety. We investigated the threshold of biotherapeutic aggregates needed to induce immunogenic responses. METHODS AND RESULTS Highly aggregated samples were tested in cell-based assays and induced cellular responses in a manner that depended on the number of particles. The threshold of immune activation varied by disease state (cancer, rheumatoid arthritis, allergy), concomitant therapies, and particle number. Compared to healthy donors, disease state patients showed an equal or lower response at the late phase (7 days), suggesting they may not have a higher risk of responding to aggregates. Xeno-het mice were used to assess the threshold of immune activation in vivo. Although highly aggregated samples (~ 1,600,000 particles/mL) induced a weak and transient immunogenic response in mice, a 100-fold dilution of this sample (~ 16,000 particles/mL) did not induce immunogenicity. To confirm this result, subvisible particles (up to ~ 18,000 particles/mL, containing aggregates and silicone oil droplets) produced under representative administration practices (created upon infusion of a drug product through an IV catheter) did not induce a response in cell-based assays or appear to increase the rate of adverse events or immunogenicity during phase 3 clinical trials. CONCLUSION The ability of biotherapeutic aggregates to elicit an immune response in vitro, in vivo, and in the clinic depends on high numbers of particles. This suggests that there is a high threshold for aggregates to induce an immunogenic response which is well beyond that seen in standard biotherapeutic drug products.
Collapse
Affiliation(s)
- Joseph R Cohen
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA.
| | - Stephen R Brych
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Siddharth Prabhu
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Vivian Bi
- The Department of Biosimilars, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Ahmed Elbaradei
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Joshua M Tokuda
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Cathie Xiang
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Martha Hokom
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of BioAnalytical Sciences, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Xiaohong Cui
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Claudia Ly
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Nathan Amos
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Jilin Sun
- Translational Safety and Bioanalytical Sciences, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Dominador Calamba
- Translational Safety and Bioanalytical Sciences, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Jonathan Herskovitz
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Allyson Capili
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Kimya Nourbakhsh
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Anthony Merlo
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Julia Carreon
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Jette Wypych
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Linda O Narhi
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Vibha Jawa
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | - Marisa K Joubert
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA.
| |
Collapse
|
3
|
Li JX, Shang RY, Xie DD, Luo XC, Hu TY, Cheng BH, Lin HW, Jiao WH. Arenarialins A-F, Anti-inflammatory Meroterpenoids with Rearranged Skeletons from the Marine Sponge Dysidea arenaria. JOURNAL OF NATURAL PRODUCTS 2024; 87:396-403. [PMID: 38330072 DOI: 10.1021/acs.jnatprod.3c01239] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Six new sesquiterpene quinone/hydroquinone meroterpenoids, arenarialins A-F (1-6), were isolated from the marine sponge Dysidea arenaria collected from the South China Sea. Their chemical structures and absolute configurations were determined by HRMS and NMR data analyses coupled with DP4+ and ECD calculations. Arenarialin A (1) features an unprecedented tetracyclic 6/6/5/6 carbon skeleton, whereas arenarialins B-D (2-4) possess two rare secomeroterpene scaffolds. Arenarialins A-F showed inhibitory activity on the production of inflammatory cytokines TNF-α and IL-6 in LPS-induced RAW264.7 macrophages with arenarialin D regulating the NF-κB/MAPK signaling pathway.
Collapse
Affiliation(s)
- Jia-Xin Li
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Ru-Yi Shang
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Dong-Dong Xie
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiang-Chao Luo
- Department of Pharmaceutical Engineering, College of Chemical Engineering, Northwest University, Xi'an 710127, China
| | - Tian-Yong Hu
- Department of Otolaryngology, Shenzhen Longgang Otolaryngology Hospital & Shenzhen Otolaryngology Research Institute, Shenzhen 518172, China
| | - Bao-Hui Cheng
- Department of Otolaryngology, Shenzhen Longgang Otolaryngology Hospital & Shenzhen Otolaryngology Research Institute, Shenzhen 518172, China
| | - Hou-Wen Lin
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei-Hua Jiao
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
4
|
Wang Z, Huang AS, Tang L, Wang J, Wang G. Microfluidic-assisted single-cell RNA sequencing facilitates the development of neutralizing monoclonal antibodies against SARS-CoV-2. LAB ON A CHIP 2024; 24:642-657. [PMID: 38165771 DOI: 10.1039/d3lc00749a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
As a class of antibodies that specifically bind to a virus and block its entry, neutralizing monoclonal antibodies (neutralizing mAbs) have been recognized as a top choice for combating COVID-19 due to their high specificity and efficacy in treating serious infections. Although conventional approaches for neutralizing mAb development have been optimized for decades, there is an urgent need for workflows with higher efficiency due to time-sensitive concerns, including the high mutation rate of SARS-CoV-2. One promising approach is the identification of neutralizing mAb candidates via single-cell RNA sequencing (RNA-seq), as each B cell has a unique transcript sequence corresponding to its secreted antibody. The state-of-the-art high-throughput single-cell sequencing technologies, which have been greatly facilitated by advances in microfluidics, have greatly accelerated the process of neutralizing mAb development. Here, we provide an overview of the general procedures for high-throughput single-cell RNA-seq enabled by breakthroughs in droplet microfluidics, introduce revolutionary approaches that combine single-cell RNA-seq to facilitate the development of neutralizing mAbs against SARS-CoV-2, and outline future steps that need to be taken to further improve development strategies for effective treatments against infectious diseases.
Collapse
Affiliation(s)
- Ziwei Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Amelia Siqi Huang
- Dalton Academy, The Affiliated High School of Peking University, Beijing, 100190, China
| | - Lingfang Tang
- Dalton Academy, The Affiliated High School of Peking University, Beijing, 100190, China
| | - Jianbin Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Guanbo Wang
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
5
|
Habtemariam S. Anti-Inflammatory Therapeutic Mechanisms of Natural Products: Insight from Rosemary Diterpenes, Carnosic Acid and Carnosol. Biomedicines 2023; 11:biomedicines11020545. [PMID: 36831081 PMCID: PMC9953345 DOI: 10.3390/biomedicines11020545] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Carnosic acid (CA) and carnosol (CAR) are two major diterpenes of the rosemary plant (Rosmarinus officinalis). They possess a phenolic structural moiety and are endowed with the power to remove cellular reactive oxygen species (ROS) either through direct scavenging reaction or indirectly through upregulation of antioxidant defences. Hand in hand with these activities are their multiple biological effects and therapeutic potential orchestrated through modulating various signalling pathways of inflammation, including the NF-κB, MAPK, Nrf2, SIRT1, STAT3 and NLRP3 inflammasomes, among others. Consequently, they ameliorate the expression of pro-inflammatory cytokines (e.g., TNF-α, IL-1 and IL-6), adhesion molecules, chemokines and prostaglandins. These anti-inflammatory mechanisms of action as a therapeutic link to various effects of these compounds, as in many other natural products, are scrutinised.
Collapse
Affiliation(s)
- Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| |
Collapse
|
6
|
Roszkowski L, Jaszczyk B, Plebańczyk M, Ciechomska M. S100A8 and S100A12 Proteins as Biomarkers of High Disease Activity in Patients with Rheumatoid Arthritis That Can Be Regulated by Epigenetic Drugs. Int J Mol Sci 2022; 24:ijms24010710. [PMID: 36614150 PMCID: PMC9820830 DOI: 10.3390/ijms24010710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune chronic inflammatory disease that is still not well understood in terms of its pathogenesis and presents diagnostic and therapeutic challenges. Monocytes are key players in initiating and maintaining inflammation through the production of pro-inflammatory cytokines and S100 proteins in RA. This study aimed to test a specific DNA methylation inhibitor (RG108) and activator (budesonide) in the regulation of pro-inflammatory mediators-especially the S100 proteins. We also searched for new biomarkers of high disease activity in RA patients. RNA sequencing analysis of healthy controls (HCs) and RA monocytes was performed. Genes such as the S100 family, TNF, and IL-8 were validated by qRT-PCR following DNA-methylation-targeted drug treatment in a monocytic THP-1 cell line. The concentrations of the S100A8, S100A11, and S100A12 proteins in the sera and synovial fluids of RA patients were tested and correlated with clinical parameters. We demonstrated that RA monocytes had significantly increased levels of S100A8, S100A9, S100A11, S100A12, MYD88, JAK3, and IQGAP1 and decreased levels of IL10RA and TGIF1 transcripts. In addition, stimulation of THP-1 cells with budesonide statistically reduced the expression of the S100 family, IL-8, and TNF genes. In contrast, THP-1 cells treated with RG108 had increased levels of the S100 family and TNF genes. We also revealed a significant upregulation of S100A8, S100A11, and S100A12 in RA patients, especially in early RA compared to HC sera. In addition, protein levels of S100A8, S100A11, and S100A12 in RA synovial fluids compared to HC sera were significantly increased. Overall, our data suggest that the S100A8 and S100A12 proteins are strongly elevated during ongoing inflammation, so they could be used as a better biomarker of disease activity than CRP. Interestingly, epigenetic drugs can regulate these S100 proteins, suggesting their potential use in targeting RA inflammation.
Collapse
Affiliation(s)
- Leszek Roszkowski
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology and Rehabilitation (NIGRiR), 02-637 Warsaw, Poland
| | - Bożena Jaszczyk
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology and Rehabilitation (NIGRiR), 02-637 Warsaw, Poland
| | - Magdalena Plebańczyk
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation (NIGRiR), 02-637 Warsaw, Poland
| | - Marzena Ciechomska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation (NIGRiR), 02-637 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-670-95-63
| |
Collapse
|
7
|
Dammen-Brower K, Epler P, Zhu S, Bernstein ZJ, Stabach PR, Braddock DT, Spangler JB, Yarema KJ. Strategies for Glycoengineering Therapeutic Proteins. Front Chem 2022; 10:863118. [PMID: 35494652 PMCID: PMC9043614 DOI: 10.3389/fchem.2022.863118] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/25/2022] [Indexed: 12/14/2022] Open
Abstract
Almost all therapeutic proteins are glycosylated, with the carbohydrate component playing a long-established, substantial role in the safety and pharmacokinetic properties of this dominant category of drugs. In the past few years and moving forward, glycosylation is increasingly being implicated in the pharmacodynamics and therapeutic efficacy of therapeutic proteins. This article provides illustrative examples of drugs that have already been improved through glycoengineering including cytokines exemplified by erythropoietin (EPO), enzymes (ectonucleotide pyrophosphatase 1, ENPP1), and IgG antibodies (e.g., afucosylated Gazyva®, Poteligeo®, Fasenra™, and Uplizna®). In the future, the deliberate modification of therapeutic protein glycosylation will become more prevalent as glycoengineering strategies, including sophisticated computer-aided tools for "building in" glycans sites, acceptance of a broad range of production systems with various glycosylation capabilities, and supplementation methods for introducing non-natural metabolites into glycosylation pathways further develop and become more accessible.
Collapse
Affiliation(s)
- Kris Dammen-Brower
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paige Epler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Stanley Zhu
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Zachary J. Bernstein
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paul R. Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Demetrios T. Braddock
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Jamie B. Spangler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kevin J. Yarema
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
8
|
Reichardt SD, Amouret A, Muzzi C, Vettorazzi S, Tuckermann JP, Lühder F, Reichardt HM. The Role of Glucocorticoids in Inflammatory Diseases. Cells 2021; 10:cells10112921. [PMID: 34831143 PMCID: PMC8616489 DOI: 10.3390/cells10112921] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
For more than 70 years, glucocorticoids (GCs) have been a powerful and affordable treatment option for inflammatory diseases. However, their benefits do not come without a cost, since GCs also cause side effects. Therefore, strong efforts are being made to improve their therapeutic index. In this review, we illustrate the mechanisms and target cells of GCs in the pathogenesis and treatment of some of the most frequent inflammatory disorders affecting the central nervous system, the gastrointestinal tract, the lung, and the joints, as well as graft-versus-host disease, which often develops after hematopoietic stem cell transplantation. In addition, an overview is provided of novel approaches aimed at improving GC therapy based on chemical modifications or GC delivery using nanoformulations. GCs remain a topic of highly active scientific research despite being one of the oldest class of drugs in medical use.
Collapse
Affiliation(s)
- Sybille D. Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; (S.D.R.); (A.A.); (C.M.)
| | - Agathe Amouret
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; (S.D.R.); (A.A.); (C.M.)
| | - Chiara Muzzi
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; (S.D.R.); (A.A.); (C.M.)
| | - Sabine Vettorazzi
- Institute of Comparative Molecular Endocrinology, Ulm University, 89081 Ulm, Germany; (S.V.); (J.P.T.)
| | - Jan P. Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, 89081 Ulm, Germany; (S.V.); (J.P.T.)
| | - Fred Lühder
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - Holger M. Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; (S.D.R.); (A.A.); (C.M.)
- Correspondence: ; Tel.: +49-551-3963365
| |
Collapse
|
9
|
Van Lent J, Breukers J, Ven K, Ampofo L, Horta S, Pollet F, Imbrechts M, Geukens N, Vanhoorelbeke K, Declerck P, Lammertyn J. Miniaturized single-cell technologies for monoclonal antibody discovery. LAB ON A CHIP 2021; 21:3627-3654. [PMID: 34505611 DOI: 10.1039/d1lc00243k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Antibodies (Abs) are among the most important class of biologicals, showcasing a high therapeutic and diagnostic value. In the global therapeutic Ab market, fully-human monoclonal Abs (FH-mAbs) are flourishing thanks to their low immunogenicity and high specificity. The rapidly emerging field of single-cell technologies has paved the way to efficiently discover mAbs by facilitating a fast screening of the antigen (Ag)-specificity and functionality of Abs expressed by B cells. This review summarizes the principles and challenges of the four key concepts to discover mAbs using these technologies, being confinement of single cells using either droplet microfluidics or microstructure arrays, identification of the cells of interest, retrieval of those cells and single-cell sequence determination required for mAb production. This review reveals the enormous potential for mix-and-matching of the above-mentioned strategies, which is illustrated by the plethora of established, highly integrated devices. Lastly, an outlook is given on the many opportunities and challenges that still lie ahead to fully exploit miniaturized single-cell technologies for mAb discovery.
Collapse
Affiliation(s)
- Julie Van Lent
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| | - Jolien Breukers
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| | - Karen Ven
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| | - Louanne Ampofo
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, Leuven 3000, Belgium
| | - Sara Horta
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk 8500, Belgium
| | - Francesca Pollet
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| | - Maya Imbrechts
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, Leuven 3000, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Nick Geukens
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk 8500, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, Leuven 3000, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Jeroen Lammertyn
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| |
Collapse
|
10
|
Roszkowski L, Ciechomska M. Tuning Monocytes and Macrophages for Personalized Therapy and Diagnostic Challenge in Rheumatoid Arthritis. Cells 2021; 10:cells10081860. [PMID: 34440629 PMCID: PMC8392289 DOI: 10.3390/cells10081860] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/21/2022] Open
Abstract
Monocytes/macrophages play a central role in chronic inflammatory disorders, including rheumatoid arthritis (RA). Activation of these cells results in the production of various mediators responsible for inflammation and RA pathogenesis. On the other hand, the depletion of macrophages using specific antibodies or chemical agents can prevent their synovial tissue infiltration and subsequently attenuates inflammation. Their plasticity is a major feature that helps the switch from a pro-inflammatory phenotype (M1) to an anti-inflammatory state (M2). Therefore, understanding the precise strategy targeting pro-inflammatory monocytes/macrophages should be a powerful way of inhibiting chronic inflammation and bone erosion. In this review, we demonstrate potential consequences of different epigenetic regulations on inflammatory cytokines production by monocytes. In addition, we present unique profiles of monocytes/macrophages contributing to identification of new biomarkers of disease activity or predicting treatment response in RA. We also outline novel approaches of tuning monocytes/macrophages by biologic drugs, small molecules or by other therapeutic modalities to reduce arthritis. Finally, the importance of cellular heterogeneity of monocytes/macrophages is highlighted by single-cell technologies, which leads to the design of cell-specific therapeutic protocols for personalized medicine in RA in the future.
Collapse
|
11
|
Yang A, Kantor B, Chiba-Falek O. APOE: The New Frontier in the Development of a Therapeutic Target towards Precision Medicine in Late-Onset Alzheimer's. Int J Mol Sci 2021; 22:1244. [PMID: 33513969 PMCID: PMC7865856 DOI: 10.3390/ijms22031244] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) has a critical unmet medical need. The consensus around the amyloid cascade hypothesis has been guiding pre-clinical and clinical research to focus mainly on targeting beta-amyloid for treating AD. Nevertheless, the vast majority of the clinical trials have repeatedly failed, prompting the urgent need to refocus on other targets and shifting the paradigm of AD drug development towards precision medicine. One such emerging target is apolipoprotein E (APOE), identified nearly 30 years ago as one of the strongest and most reproduceable genetic risk factor for late-onset Alzheimer's disease (LOAD). An exploration of APOE as a new therapeutic culprit has produced some very encouraging results, proving that the protein holds promise in the context of LOAD therapies. Here, we review the strategies to target APOE based on state-of-the-art technologies such as antisense oligonucleotides, monoclonal antibodies, and gene/base editing. We discuss the potential of these initiatives in advancing the development of novel precision medicine therapies to LOAD.
Collapse
Affiliation(s)
- Anna Yang
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Boris Kantor
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA;
- Viral Vector Core, Duke University Medical Center, Durham, NC 27710, USA
- Duke Center for Advanced Genomic Technologies, Durham, NC 27708, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA;
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
12
|
Lee YH, Kim K, Lee JH, Kim HJ. Protection of Alcohol Dehydrogenase against Freeze-Thaw Stress by Ice-Binding Proteins Is Proportional to Their Ice Recrystallization Inhibition Property. Mar Drugs 2020; 18:md18120638. [PMID: 33322085 PMCID: PMC7764648 DOI: 10.3390/md18120638] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 11/16/2022] Open
Abstract
Ice-binding proteins (IBPs) have ice recrystallization inhibition (IRI) activity. IRI property has been extensively utilized for the cryopreservation of different types of cells and tissues. Recent reports demonstrated that IRI can also play a significant role in protecting proteins from freezing damage during freeze-thaw cycles. In this study, we hypothesized that the protective capability of IBPs on proteins against freeze-thaw damage is proportional to their IRI activity. Hence we used two IBPs: one with higher IRI activity (LeIBP) and the other with lower activity (FfIBP). Yeast alcohol dehydrogenase (ADH) was used as a freeze-labile model protein. IBPs and ADH were mixed, frozen at -20 °C, and thawed repeatedly. The structure of ADH was assessed using fluorescence emission spectra probed by 1-anilinonaphthalene-8-sulfonate over the repeated freeze-thaw cycles. The activity was monitored at 340 nm spectrophotometrically. Fluorescence data and activity clearly indicated that ADH without IBP was freeze-labile. However, ADH maintained about 70% residual activity after five repeated cycles at a minimal concentration of 0.1 mg mL-1 of high IRI-active LeIBP, but only 50% activity at 4 mg mL-1 of low active FfIBP. These results showed that the protection of proteins from freeze-thaw stress by IBPs is proportional to their IRI activity.
Collapse
Affiliation(s)
- Young Hoon Lee
- Department of Chemistry, Pukyong National University, Busan 48513, Korea;
| | - Kitae Kim
- Research Unit of Cryogenic Novel Material, Korea Polar Research Institute, Incheon 21990, Korea; (K.K.); (J.H.L.)
- Department of Polar Sciences, University of Science and Technology, Incheon 21990, Korea
| | - Jun Hyuck Lee
- Research Unit of Cryogenic Novel Material, Korea Polar Research Institute, Incheon 21990, Korea; (K.K.); (J.H.L.)
- Department of Polar Sciences, University of Science and Technology, Incheon 21990, Korea
| | - Hak Jun Kim
- Department of Chemistry, Pukyong National University, Busan 48513, Korea;
- Correspondence: ; Tel.: +82-51-629-5587
| |
Collapse
|
13
|
Li R, Ng TS, Garlin MA, Weissleder R, Miller MA. Understanding the in vivo Fate of Advanced Materials by Imaging. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910369. [PMID: 38545084 PMCID: PMC10972611 DOI: 10.1002/adfm.201910369] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/09/2020] [Indexed: 11/13/2024]
Abstract
Engineered materials are ubiquitous in biomedical applications ranging from systemic drug delivery systems to orthopedic implants, and their actions unfold across multiple time- and length-scales. The efficacy and safety of biologics, nanomaterials, and macroscopic implants are all dictated by the same general principles of pharmacology as apply to small molecule drugs, comprising how the body affects materials (pharmacokinetics, PK) and conversely how materials affect the body (pharmacodynamics, PD). Imaging technologies play an increasingly insightful role in monitoring both of these processes, often simultaneously: translational macroscopic imaging modalities such as MRI and PET/CT offer whole-body quantitation of biodistribution and structural or molecular response, while ex vivo approaches and optical imaging via in vivo (intravital) microscopy reveal behaviors at subcellular resolution. In this review, the authors survey developments in imaging the in situ behavior of systemically and locally administered materials, with a particular focus on using microscopy to understand transport, target engagement, and downstream host responses at a single-cell level. The themes of microenvironmental influence, controlled drug release, on-target molecular action, and immune response, especially as mediated by macrophages and other myeloid cells are examined. Finally, the future directions of how new imaging technologies may propel efficient clinical translation of next-generation therapeutics and medical devices are proposed.
Collapse
Affiliation(s)
- Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Michelle A. Garlin
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
- Department of Systems Biology, Harvard Medical School
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
14
|
Zheng S, Shen F, Jones B, Fink D, Geist B, Nnane I, Zhou Z, Hall J, Malaviya R, Ort T, Wang W. Characterization of concurrent target suppression by JNJ-61178104, a bispecific antibody against human tumor necrosis factor and interleukin-17A. MAbs 2020; 12:1770018. [PMID: 32544369 PMCID: PMC7531573 DOI: 10.1080/19420862.2020.1770018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tumor necrosis factor (TNF) and interleukin (IL)-17A are pleiotropic cytokines implicated in the pathogenesis of several autoimmune diseases including rheumatoid arthritis (RA) and psoriatic arthritis (PsA). JNJ-61178104 is a novel human anti-TNF and anti-IL-17A monovalent, bispecific antibody that binds to both human TNF and human IL-17A with high affinities and blocks the binding of TNF and IL-17A to their receptors in vitro. JNJ-61178104 also potently neutralizes TNF and IL-17A-mediated downstream effects in multiple cell-based assays. In vivo, treatment with JNJ-61178104 resulted in dose-dependent inhibition of cellular influx in a human IL-17A/TNF-induced murine lung neutrophilia model and the inhibitory effects of JNJ-61178104 were more potent than the treatment with bivalent parental anti-TNF or anti-IL-17A antibodies. JNJ-61178104 was shown to engage its targets, TNF and IL-17A, in systemic circulation measured as drug/target complex formation in normal cynomolgus monkeys (cyno). Surprisingly, quantitative target engagement assessment suggested lower apparent in vivo target-binding affinities for JNJ-61178104 compared to its bivalent parental antibodies, despite their similar in vitro target-binding affinities. The target engagement profiles of JNJ-61178104 in humans were in general agreement with the predicted profiles based on cyno data, suggesting similar differences in the apparent in vivo target-binding affinities. These findings show that in vivo target engagement of monovalent bispecific antibody does not necessarily recapitulate that of the molar-equivalent dose of its bivalent parental antibody. Our results also offer valuable insights into the understanding of the pharmacokinetics/pharmacodynamics and target engagement of other bispecific biologics against dimeric and/or trimeric soluble targets in vivo.
Collapse
Affiliation(s)
- Songmao Zheng
- Biologics Development Sciences, Janssen Biotherapeutics, Janssen R&D , Spring House, PA, USA
| | - Fang Shen
- Immunology Discovery, Clinical Pharmacology and Pharmacometrics, Janssen R&D , Spring House, PA, USA
| | - Brian Jones
- Immunology Discovery, Clinical Pharmacology and Pharmacometrics, Janssen R&D , Spring House, PA, USA
| | - Damien Fink
- Biologics Development Sciences, Janssen Biotherapeutics, Janssen R&D , Spring House, PA, USA
| | - Brian Geist
- Biologics Development Sciences, Janssen Biotherapeutics, Janssen R&D , Spring House, PA, USA
| | - Ivo Nnane
- Clinical Pharmacology and Pharmacometrics, Janssen R&D , Spring House, PA, USA
| | - Zhao Zhou
- Immunology Discovery, Clinical Pharmacology and Pharmacometrics, Janssen R&D , Spring House, PA, USA
| | - Jeff Hall
- Immunology Discovery, Clinical Pharmacology and Pharmacometrics, Janssen R&D , Spring House, PA, USA
| | - Ravi Malaviya
- Immunology Discovery, Clinical Pharmacology and Pharmacometrics, Janssen R&D , Spring House, PA, USA
| | - Tatiana Ort
- Immunology Discovery, Clinical Pharmacology and Pharmacometrics, Janssen R&D , Spring House, PA, USA
| | - Weirong Wang
- Clinical Pharmacology and Pharmacometrics, Janssen R&D , Spring House, PA, USA
| |
Collapse
|
15
|
Grubb A. Shrunken pore syndrome - a common kidney disorder with high mortality. Diagnosis, prevalence, pathophysiology and treatment options. Clin Biochem 2020; 83:12-20. [PMID: 32544475 DOI: 10.1016/j.clinbiochem.2020.06.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
Invasive studies show that the glomerular sieving coefficients for 5-30 kDa plasma proteins in the human kidney may be selectively reduced compared to those for small molecules < 0.9 kDa, commonly used to measure glomerular filtration rate (GFR). Identification of this pathophysiological state, called shrunken pore syndrome (SPS), can easily and non-invasively be done by comparing estimations of GFR using cystatin C (13.3 kDa) and creatinine (0.113 kDa). SPS is present if the estimate of GFR using cystatin C is lower than 60 or 70% of the estimate using creatinine in the absence of non-renal influences on cystatin C or creatinine. All studies of SPS show that the 3- or 5-year mortality is strongly increased and high hazard ratios for mortality associated with SPS have been observed for many different patient cohorts, including cohorts with normal measured GFR, no albuminuria and no diagnosis. The prevalence of SPS in the cohorts so far investigated is between 0.2 and 36%. Proteome studies of SPS demonstrate that the high mortality associated with the syndrome might be caused by the accumulation of 10-30 kDa signalling proteins promoting development of atherosclerosis and thus suggesting use of monoclonal antibodies to reduce the levels of the most detrimental signalling proteins as a treatment option. The KDIGO recommendations for classification of chronic kidney disease (CKD) comprise determination, or estimation, of GFR and analysis of albuminuria and therefore cannot identify a large fraction of the patients with SPS. The high prevalence and mortality of SPS and the possible treatment options strongly suggest that the KDIGO recommendations should be expanded to include determination of cystatin C to be able to identify all patients with SPS.
Collapse
Affiliation(s)
- Anders Grubb
- Department of Clinical Chemistry and Pharmacology, Institute of Laboratory Medicine, Lund University, S-22185 Lund, Sweden.
| |
Collapse
|
16
|
Understanding Molecules that Mediate Leukocyte Extravasation. CURRENT PATHOBIOLOGY REPORTS 2020. [DOI: 10.1007/s40139-020-00207-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
17
|
Bispecific Antibodies for Autoimmune and Inflammatory Diseases: Clinical Progress to Date. BioDrugs 2020; 34:111-119. [DOI: 10.1007/s40259-019-00400-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
18
|
Hua G, Zein N, Daubeuf F, Chambon P. Glucocorticoid receptor modulators CpdX and CpdX-D3 exhibit the same in vivo antiinflammatory activities as synthetic glucocorticoids. Proc Natl Acad Sci U S A 2019; 116:14191-14199. [PMID: 31227605 PMCID: PMC6628818 DOI: 10.1073/pnas.1908258116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We previously reported that the nonsteroidal compound CpdX, which was initially characterized 20 y ago as a possible gestagen and, shortly afterward, as a possible drug for treatments of inflammatory diseases, selectively triggers the NFκB/AP1-mediated tethered indirect transrepression function of the glucocorticoid receptor (GR), and could therefore be a selective glucocorticoid receptor agonistic modulator (SEGRAM). We now demonstrate that, upon administration to the mouse, CpdX and one of its deuterated derivatives, CpdX-D3, repress as efficiently as a synthetic glucocorticoid (e.g., Dexamethasone) an induced skin atopic dermatitis, an induced psoriasis-like inflammation, a house dust mite (HDM)-induced asthma-like allergic lung inflammation, a collagen-induced arthritis, an induced ulcerative colitis, and an ovalbumin-induced allergic conjunctivitis. Interestingly, in the cases of an HDM-induced asthma-like allergic lung inflammation and of a collagen-induced arthritis, the CpdX antiinflammatory activity was selectively exerted by one of the two CpdX enantiomers, namely, CpdX(eA) or CpdX-D3(eA).
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/chemistry
- Anti-Inflammatory Agents/pharmacology
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/genetics
- Arthritis, Experimental/pathology
- Asthma/drug therapy
- Asthma/genetics
- Asthma/pathology
- Conjunctivitis, Allergic/drug therapy
- Conjunctivitis, Allergic/genetics
- Conjunctivitis, Allergic/pathology
- Dermatitis, Atopic/chemically induced
- Dermatitis, Atopic/drug therapy
- Dermatitis, Atopic/genetics
- Dermatitis, Atopic/pathology
- Dexamethasone/pharmacology
- Disease Models, Animal
- Glucocorticoids/genetics
- Glucocorticoids/pharmacology
- Humans
- Inflammation/drug therapy
- Inflammation/genetics
- Inflammation/pathology
- Mice
- NF-kappa B/genetics
- Ovalbumin/toxicity
- Progestins/chemistry
- Progestins/pharmacology
- Receptors, Glucocorticoid/agonists
- Receptors, Glucocorticoid/chemistry
- Receptors, Glucocorticoid/genetics
- Skin/drug effects
- Skin/pathology
- Transcriptional Activation/drug effects
Collapse
Affiliation(s)
- Guoqiang Hua
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, 67404 Illkirch, France
| | - Naimah Zein
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, 67404 Illkirch, France
| | - François Daubeuf
- Laboratoire d'Innovation Thérapeutique, Unité Mixte de Recherche 7200, Faculté de Pharmacie, Centre National de la Recherche Scientifique-Université de Strasbourg, F-67400 Illkirch, France
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, 67404 Illkirch, France;
- University of Strasbourg Institute for Advanced Study, 67404 Illkirch, France
- Collège de France, 67404 Illkirch, France
| |
Collapse
|
19
|
Siouti E, Andreakos E. The many facets of macrophages in rheumatoid arthritis. Biochem Pharmacol 2019; 165:152-169. [PMID: 30910693 DOI: 10.1016/j.bcp.2019.03.029] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/19/2019] [Indexed: 12/24/2022]
Abstract
Macrophages are central to the pathophysiology of rheumatoid arthritis (RA). They constitute the main source of pro-inflammatory cytokines and chemokines such as TNF and IL-1β, they activate a wide range of immune and non-immune cells, and they secrete diverse tissue degrading enzymes driving chronic pro-inflammatory, tissue destructive and pain responses in RA. However, they can also produce anti-inflammatory cytokines such as IL-10, secrete inhibitors of tissue degrading enzymes and promote immunoregulatory and protective responses, suggesting the existence of macrophages with distinct and diverse functional activities. Although the underlying basis of this phenomenon has remained obscure for years, emerging evidence has now provided insight into the mechanisms and molecular processes involved. Here, we review current knowledge on the biology of macrophages in RA, and highlight recent literature on the heterogeneity, origins and ontogeny of macrophages as part of the mononuclear phagocyte system. We also discuss their plasticity in the context of the M1/M2 paradigm, and the emerging theme of metabolic rewiring as a major mechanism for programming macrophage functions and pro-inflammatory activities. This sheds light into the many facets of macrophages in RA, their molecular regulation and their translational potential for developing novel protective and therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Eleni Siouti
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; Airway Disease Infection Section, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London W2 1NY, United Kingdom.
| |
Collapse
|
20
|
Akpalu DE, Frederick B, Nnane IP, Yao Z, Shen F, Ort T, Fink D, Dogmanits S, Raible D, Sharma A, Xu Z. Pharmacokinetics, Pharmacodynamics, Immunogenicity, Safety, and Tolerability of JNJ-61178104, a Novel Tumor Necrosis Factor-Alpha and Interleukin-17A Bispecific Antibody, in Healthy Subjects. J Clin Pharmacol 2019; 59:968-978. [PMID: 30776134 DOI: 10.1002/jcph.1393] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/28/2019] [Indexed: 12/19/2022]
Abstract
The safety, tolerability, pharmacokinetics, pharmacodynamics, and immunogenicity of JNJ-61178104, a novel anti-tumor necrosis factor-alpha (TNFα) and anti-interleukin-17A (IL-17A) bispecific antibody, were investigated in a placebo-controlled, first-in-human study. Healthy subjects (n = 54) received a single dose of JNJ-61178104 by either intravenous infusion (0.1, 0.3, 1, 3, and 10 mg/kg) or subcutaneous injection (1 mg/kg). Blood samples for measurement of serum JNJ-61178104 concentrations, total IL-17A, total TNFα, and detection of antidrug antibodies were collected for up to 16 weeks after dosing and assessed using electrochemiluminescence immunoassays. PK parameters were calculated by noncompartmental analysis and estimated by nonlinear mixed-effects modeling. JNJ-61178104 was generally well tolerated in healthy subjects. For the intravenous cohorts, mean maximum concentration, and area under the concentration-time curve values increased in a dose-proportional manner. Mean clearance ranged from 6.73 to 9.99 mL/day/kg, mean volume of distribution at terminal phase after intravenous administration ranged from 51.0 to 91.9 mL/kg, and mean half-life ranged from 4.3 to 9.7 days following intravenous administration. After a single subcutaneous dose of 1 mg/kg, median time to maximum concentration was 4.0 days, mean bioavailability was 52.0% and mean half-life was 5.3 days. A linear 2-compartment population model with first-order elimination adequately characterized the pharmacokinetics with parameters consistent with noncompartmental analysis estimates. Body weight and antidrug antibodies were significant covariates on JNJ-61178104 clearance. The time to reach mean maximum serum total TNFα and total IL-17A concentrations appeared to be dose dependent across the 0.1 mg/kg to 10 mg/kg IV dose groups. All subjects who received active treatment were antidrug antibody positive after dosing with JNJ-61178104.
Collapse
Affiliation(s)
- Derrick E Akpalu
- Clinical Pharmacology & Pharmacometrics, Janssen R&D, Spring House, PA, USA
| | - Bart Frederick
- Immunology Translational Medicine, Janssen R&D, Spring House, PA, USA
| | - Ivo P Nnane
- Clinical Pharmacology & Pharmacometrics, Janssen R&D, Spring House, PA, USA
| | - Zhenling Yao
- Clinical Pharmacology & Pharmacometrics, Janssen R&D, Spring House, PA, USA
| | - Fang Shen
- Immunology Discovery, Janssen R&D, Spring House, PA, USA
| | - Tatiana Ort
- Immunology Discovery, Janssen R&D, Spring House, PA, USA
| | - Damien Fink
- Biologics Development Sciences, Janssen R&D, Spring House, PA, USA
| | | | - Donald Raible
- Immunology Translational Medicine, Janssen R&D, Spring House, PA, USA
| | - Amarnath Sharma
- Clinical Pharmacology & Pharmacometrics, Janssen R&D, Spring House, PA, USA
| | - Zhenhua Xu
- Clinical Pharmacology & Pharmacometrics, Janssen R&D, Spring House, PA, USA
| |
Collapse
|
21
|
Mitchell DE, Fayter AER, Deller RC, Hasan M, Gutierrez-Marcos J, Gibson MI. Ice-recrystallization inhibiting polymers protect proteins against freeze-stress and enable glycerol-free cryostorage. MATERIALS HORIZONS 2019; 6:364-368. [PMID: 30931129 PMCID: PMC6394881 DOI: 10.1039/c8mh00727f] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/19/2018] [Indexed: 05/19/2023]
Abstract
Proteins are ubiquitous in molecular biotechnology, biotechnology and as therapeutics, but there are significant challenges in their storage and distribution, with freezing often required. This is traditionally achieved by the addition of cryoprotective agents such as glycerol (or trehalose) or covalent modification of mutated proteins with cryoprotectants. Here, ice recrystallization inhibiting polymers, inspired by antifreeze proteins, are used synergistically with poly(ethylene glycol) as an alternative to glycerol. The primary mechanism of action appears to be preventing irreversible aggregation due to ice growth. The polymer formulation is successfully used to cryopreserve a range of important proteins including insulin, Taq DNA polymerase and an IgG antibody. The polymers do not require covalent conjugation, nor modification of the protein and are already used in a wide range of biomedical applications, which will facilitate translation to a range of biologics.
Collapse
Affiliation(s)
- Daniel E Mitchell
- Department of Chemistry , University of Warwick , Coventry , CV47AL , UK .
| | - Alice E R Fayter
- Department of Chemistry , University of Warwick , Coventry , CV47AL , UK .
| | - Robert C Deller
- Department of Chemistry , University of Warwick , Coventry , CV47AL , UK .
| | - Muhammad Hasan
- Department of Chemistry , University of Warwick , Coventry , CV47AL , UK .
| | | | - Matthew I Gibson
- Department of Chemistry , University of Warwick , Coventry , CV47AL , UK .
- Warwick Medical School , University of Warwick , CV47AL , UK
| |
Collapse
|
22
|
Hifumi E, Taguchi H, Toorisaka E, Uda T. New technologies to introduce a catalytic function into antibodies: A unique human catalytic antibody light chain showing degradation of β-amyloid molecule along with the peptidase activity. FASEB Bioadv 2019; 1:93-104. [PMID: 32123823 PMCID: PMC6996398 DOI: 10.1096/fba.1025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/04/2018] [Accepted: 10/12/2018] [Indexed: 12/20/2022] Open
Abstract
Since the discovery of a natural catalytic antibody in 1989, many catalytic antibodies targeting peptides, nucleotides, virus and bacterial proteins, and many molecules have been prepared. Although catalytic antibodies should have features superior to non-catalytic monoclonal antibodies, the reports on catalytic antibodies are far fewer than those on normal (non-catalytic) antibodies. Nowadays, we can obtain any monoclonal antibody we want, which is not the case for catalytic antibodies. To overcome this drawback of catalytic antibodies, much basic research must be done. As one way to attain this purpose, we have been making a protein bank of human antibody light chains, in which the light chains were expressed, purified, and stored for use in screening against many kinds of antigen, to then get clues to introducing a catalytic function in normal antibodies. As the number of stored light chains in the above protein bank has reached the hundreds, in this study, we screened them against amyloid-beta (Aβ), which is well-known as one of the molecules causing Alzheimer's disease. We found two interesting light chains, #7TR and #7GY. The former could degrade both a fluorescence resonance energy transfer-Aβ substrate and Aβ1-40 full peptide. In contrast, #7GY, whose sequence is identical to that of #7TR except for the amino acids at the 29th and 30th positions, did not degrade the FRET-Aβ substrate at all. By using a synthetic substrate, Arg-pNA, the difference between the chemical features of the two light chains was investigated in detail. In addition, we found that the presence of Zn(II) ion hugely influenced the catalytic activity of the #7TR light chain but not #7GY. Through these facts and the discussion, we propose one of the clues to how to put a catalytic function in a normal (non-catalytic) antibody.
Collapse
Affiliation(s)
- Emi Hifumi
- Research Promotion Institute, Oita UniversityOitaJapan
| | - Hiroaki Taguchi
- Faculty of Pharmaceutical SciencesSuzuka University of Medical ScienceSuzukaJapan
| | - Eiichi Toorisaka
- Faculty of Engineering, Department of Sustainable EngineeringYamaguchi UniversityYamaguchiJapan
| | - Taizo Uda
- Faculty of Engineering, Department of Applied ChemistryOita UniversityOitaJapan
- Nanotechnology LaboratoryInstitute of Systems, Information Technologies and Nanotechnologies (ISIT)FukuokaJapan
| |
Collapse
|
23
|
Richter F, Zettlitz KA, Seifert O, Herrmann A, Scheurich P, Pfizenmaier K, Kontermann RE. Monovalent TNF receptor 1-selective antibody with improved affinity and neutralizing activity. MAbs 2019; 11:166-177. [PMID: 30252601 PMCID: PMC6343807 DOI: 10.1080/19420862.2018.1524664] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/29/2018] [Accepted: 09/10/2018] [Indexed: 01/08/2023] Open
Abstract
Selective inhibition of tumor necrosis factor (TNF) signaling through the proinflammatory axis of TNF-receptor 1 (TNFR1) while leaving pro-survival and regeneration-promoting signals via TNFR2 unaffected is a promising strategy to circumvent limitations of complete inhibition of TNF action by the approved anti-TNF drugs. A previously developed humanized antagonistic TNFR1-specific antibody, ATROSAB, showed potent inhibition of TNFR1-mediated cellular responses. Because the parental mouse antibody H398 possesses even stronger inhibitory potential, we scrutinized the specific binding parameters of the two molecules and revealed a faster dissociation of ATROSAB compared to H398. Applying affinity maturation and re-engineering of humanized variable domains, we generated a monovalent Fab derivative (13.7) of ATROSAB that exhibited increased binding to TNFR1 and superior inhibition of TNF-mediated TNFR1 activation, while lacking any agonistic activity even in the presence of cross-linking antibodies. In order to improve its pharmacokinetic properties, several Fab13.7-derived molecules were generated, including a PEGylated Fab, a mouse serum albumin fusion protein, a half-IgG with a dimerization-deficient Fc, and a newly designed Fv-Fc format, employing the knobs-into-holes technology. Among these derivatives, the Fv13.7-Fc displayed the best combination of improved pharmacokinetic properties and antagonistic activity, thus representing a promising candidate for further clinical development.
Collapse
Affiliation(s)
- Fabian Richter
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Kirstin A. Zettlitz
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | | | - Peter Scheurich
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
24
|
Diagnoses and Management of Drug Hypersensitivity and Anaphylaxis in Cancer and Chronic Inflammatory Diseases: Reactions to Taxanes and Monoclonal Antibodies. Clin Rev Allergy Immunol 2018; 54:375-385. [PMID: 27277133 DOI: 10.1007/s12016-016-8556-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Due to the increase in utilization of chemotherapies and antibodies, drug hypersensitivity reactions have increased dramatically worldwide, preventing the use of first-line therapies and impacting patients' survival and quality of life. Some of the more frequently used medications in cancer include taxanes for ovarian, lung, breast, and prostate cancers. Monoclonal antibodies are used in the treatment of neoplastic, autoimmune, and inflammatory diseases, and their clinical applications are becoming broader. Monoclonal antibody targets include CD20, HER-2, EGFR, IL-6 receptor, TNF-α, CD30, VEGF-A, IgE, and more, and examples of immune-mediated and inflammatory diseases that respond to monoclonal antibodies include rheumatoid arthritis, Crohn's disease, ulcerative colitis, juvenile idiopathic arthritis, psoriasis and psoriatic arthritis, Wegener's granulomatosis, microscopic polyangiitis, ankylosing spondylitis, plaque psoriasis, and asthma. Neoplastic diseases include non-Hodgkin's lymphoma, chronic lymphocytic leukemia, and colorectal, breast, gastric, and lung cancer. The clinical presentation of drug hypersensitivity reactions ranges from mild cutaneous reactions to life-threatening symptoms including anaphylaxis. Rapid drug desensitization (RDD) has become a groundbreaking approach to the management of immediate drug hypersensitivity reactions IgE and non-IgE mediated. It is the only effective procedure that enables sensitized patients to receive the full treatment dose safely, thus representing an important advance in the patients' treatment and prognosis. The aim of this review is to provide an update on hypersensitivity reactions to commonly used monoclonal and taxanes, their clinical presentations, diagnosis, and the use of RDD for their management.
Collapse
|
25
|
Zhou W, Lin S, Chen R, Liu J, Li Y. Characterization of antibody-C1q interactions by Biolayer Interferometry. Anal Biochem 2018; 549:143-148. [DOI: 10.1016/j.ab.2018.03.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 02/20/2018] [Accepted: 03/21/2018] [Indexed: 12/17/2022]
|
26
|
Microfluidic single-cell technology in immunology and antibody screening. Mol Aspects Med 2018; 59:47-61. [DOI: 10.1016/j.mam.2017.09.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 11/20/2022]
|
27
|
Pacilio M, Lauri C, Prosperi D, Petitti A, Signore A. New SPECT and PET Radiopharmaceuticals for Imaging Inflammatory Diseases: A Meta-analysis of the Last 10 Years. Semin Nucl Med 2018; 48:261-276. [PMID: 29626943 DOI: 10.1053/j.semnuclmed.2017.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Modern molecular nuclear medicine is rapidly developing in the field of imaging of chronic inflammatory diseases, and many new radiopharmaceuticals have been recently described and tested in animals and man. These can detect early pathophysiological changes before the development of anatomical changes and, often, before clinical onset of symptoms. This field includes new radiopharmaceuticals for SPECT and PET use to define new strategies for imaging immune cells as well as tissue modifications induced by the inflammatory process. In this review, we present the results of a meta-analysis based on radiopharmaceuticals (for SPECT or PET) that are not commercially available and that have been used, at least once, in humans in the last 10 years.
Collapse
Affiliation(s)
- Marta Pacilio
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, Italy
| | - Chiara Lauri
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, Italy.; Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Daniela Prosperi
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, Italy
| | - Agnese Petitti
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, Italy
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, Italy.; Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, The Netherlands.
| |
Collapse
|
28
|
Patel AB, Tsilioni I, Weng Z, Theoharides TC. TNF stimulates IL-6, CXCL8 and VEGF secretion from human keratinocytes via activation of mTOR, inhibited by tetramethoxyluteolin. Exp Dermatol 2018; 27:135-143. [PMID: 29105195 DOI: 10.1111/exd.13461] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2017] [Indexed: 12/19/2022]
Abstract
Psoriasis is an autoimmune skin disease characterized by keratinocyte hyperproliferation and chronic inflammation. The pathogenesis of psoriasis involves proinflammatory cytokines, such as tumor necrosis factor (TNF), but the mechanism of keratinocyte activation is not well understood. Here, we show that TNF (10 or 50 ng/mL) stimulates a significant (P < .0001) gene expression and secretion of proinflammatory IL-6, CXCL8 and VEGF from both cultured human HaCaT and normal epidermal human keratinocytes (NHEKs). This effect occurs via activation of the mammalian target of rapamycin (mTOR) signalling complex as shown by Western blot analysis and phospho-ELISAs. Pretreatment with the novel natural flavonoid tetramethoxyluteolin (10-100 μmol L-1 ) significantly (P < .0001) inhibits gene expression and secretion (P < .0001) of all 3 mediators in a concentration-dependent manner. Moreover, tetramethoxyluteolin (50 μmol L-1 ) appears to be a potent inhibitor of the phosphorylated mTOR substrates (pmTORSer2448 , pp70S6KThr389 and p4EBP1Thr37/46 ) as compared to known mTOR inhibitors in keratinocytes. The present findings indicate that TNF stimulates skin inflammation via mTOR signalling. Inhibition by tetramethoxyluteolin may be used in the treatment for psoriasis.
Collapse
Affiliation(s)
- Arti B Patel
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA.,Graduate Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Irene Tsilioni
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - Zuyi Weng
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - Theoharis C Theoharides
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA.,Graduate Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA.,Department of Internal Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
29
|
Regan-Komito D, Valaris S, Kapellos TS, Recio C, Taylor L, Greaves DR, Iqbal AJ. Absence of the Non-Signalling Chemerin Receptor CCRL2 Exacerbates Acute Inflammatory Responses In Vivo. Front Immunol 2017; 8:1621. [PMID: 29209334 PMCID: PMC5702352 DOI: 10.3389/fimmu.2017.01621] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/08/2017] [Indexed: 01/20/2023] Open
Abstract
Chemerin is a chemotactic protein that induces migration of several immune cells including macrophages, immature dendritic cells, and NK cells. Chemerin binds to three G protein-coupled receptors (GPCRs), including CCRL2. The exact function of CCRL2 remains unclear. CCRL2 expression is rapidly upregulated during inflammation, but it lacks the intracellular DRYLAIV motif required for classical GPCR downstream signalling pathways, and it has not been reported to internalise chemerin upon binding. The aim of this study was to investigate what role if any CCRL2 plays during acute inflammation. Using the zymosan- and thioglycollate-induced murine models of acute inflammation, we report that mice deficient in the Ccrl2 gene display exaggerated local and systemic inflammatory responses, characterised by increased myeloid cell recruitment. This amplified myeloid cell recruitment was associated with increased chemerin and CXCL1 levels. Furthermore, we report that the inflammatory phenotype observed in these mice is dependent upon elevated levels of endogenous chemerin. Antibody neutralisation of chemerin activity in Ccrl2-/- mice abrogated the amplified inflammatory responses. Importantly, chemerin did not directly recruit myeloid cells but rather increased the production of other chemotactic proteins such as CXCL1. Administration of recombinant chemerin to wild-type mice before inflammatory challenge recapitulated the increased myeloid cell recruitment and inflammatory mediator production observed in Ccrl2-/- mice. We have demonstrated that the absence of CCRL2 results in increased levels of local and systemic chemerin levels and exacerbated inflammatory responses during acute inflammatory challenge. These results further highlight the importance of chemerin as a therapeutic target in inflammatory diseases.
Collapse
Affiliation(s)
- Daniel Regan-Komito
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Sophia Valaris
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Theodore S. Kapellos
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Carlota Recio
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Lewis Taylor
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Asif J. Iqbal
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
- College of Medical and Dental Sciences, Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
30
|
Akinrinmade OA, Chetty S, Daramola AK, Islam MU, Thepen T, Barth S. CD64: An Attractive Immunotherapeutic Target for M1-type Macrophage Mediated Chronic Inflammatory Diseases. Biomedicines 2017; 5:biomedicines5030056. [PMID: 28895912 PMCID: PMC5618314 DOI: 10.3390/biomedicines5030056] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/12/2022] Open
Abstract
To date, no curative therapy is available for the treatment of most chronic inflammatory diseases such as atopic dermatitis, rheumatoid arthritis, or autoimmune disorders. Current treatments require a lifetime supply for patients to alleviate clinical symptoms and are unable to stop the course of disease. In contrast, a new series of immunotherapeutic agents targeting the Fc γ receptor I (CD64) have emerged and demonstrated significant clinical potential to actually resolving chronic inflammation driven by M1-type dysregulated macrophages. This subpopulation plays a key role in the initiation and maintenance of a series of chronic diseases. The novel recombinant M1-specific immunotherapeutics offer the prospect of highly effective treatment strategies as they have been shown to selectively eliminate the disease-causing macrophage subpopulations. In this review, we provide a detailed summary of the data generated, together with the advantages and the clinical potential of CD64-based targeted therapies for the treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Olusiji A Akinrinmade
- South African Research Chair in Cancer Biotechnology, Institute of Infectious Disease and Molecular Medicine (IDM), Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, 7925 Cape Town, South Africa.
| | - Shivan Chetty
- South African Research Chair in Cancer Biotechnology, Institute of Infectious Disease and Molecular Medicine (IDM), Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, 7925 Cape Town, South Africa.
| | - Adebukola K Daramola
- South African Research Chair in Cancer Biotechnology, Institute of Infectious Disease and Molecular Medicine (IDM), Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, 7925 Cape Town, South Africa.
| | - Mukit-Ul Islam
- South African Research Chair in Cancer Biotechnology, Institute of Infectious Disease and Molecular Medicine (IDM), Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, 7925 Cape Town, South Africa.
| | - Theo Thepen
- Institute for Transfusion Medicine and Immunohematology and Blood Bank. University Hospital Magdeburg A.ö.R, 39120 Magdeburg, Germany.
| | - Stefan Barth
- South African Research Chair in Cancer Biotechnology, Institute of Infectious Disease and Molecular Medicine (IDM), Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, 7925 Cape Town, South Africa.
| |
Collapse
|
31
|
Babb R, Pirofski LA. Help is on the way: Monoclonal antibody therapy for multi-drug resistant bacteria. Virulence 2017; 8:1055-1058. [PMID: 28306387 PMCID: PMC5711430 DOI: 10.1080/21505594.2017.1306620] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Rachelle Babb
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY, USA
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY, USA,CONTACT Liise-anne Pirofski Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
32
|
In vitro assays supporting the safety assessment of immunomodulatory monoclonal antibodies. Toxicol In Vitro 2017; 45:296-308. [PMID: 28263892 DOI: 10.1016/j.tiv.2017.02.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/08/2017] [Accepted: 02/28/2017] [Indexed: 01/11/2023]
Abstract
Many monoclonal antibodies (mAbs) licensed for human use or in clinical development for cancer and autoimmune disease directly interact with the immune system. These immunomodulatory mAbs have an inherent risk of adverse immune-mediated drug reactions, including infusion reactions, cytokine storms, immunosuppression and autoimmunity. A thorough understanding of the potential for immunotoxicity of a mAb is required to support administration to humans. This review will highlight the key role of in vitro assays in defining the immunopharmacology, immunotoxicity and immunogenicity of mAbs. A wide range of in vitro tests with multiple formats of different complexity can be utilized to characterize i) the antibody-binding domains of the mAb, such as on-target binding and downstream pharmacological effects (e.g. immunosuppression, immune activation, cytokine release) in both humans and animal species used for toxicology studies and off-target binding; ii) Fc-dependent effects such as Fc-mediated cellular activation (e.g. of leukocytes, platelets) and cytokine release, complement activation; and iii) product-related factors (sequence, physical-chemical properties and impurities) that can impact both pharmacological activity and immunogenicity potential of a mAb. These assays can be crucial to the selection of mAbs with an optimum balance of safety and efficacy, in defining whether a mAb is a high risk molecule, and together with animal data, can inform human safe starting doses and escalation schemes.
Collapse
|
33
|
Aptamers for CD Antigens: From Cell Profiling to Activity Modulation. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 6:29-44. [PMID: 28325295 PMCID: PMC5363458 DOI: 10.1016/j.omtn.2016.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/02/2016] [Accepted: 12/02/2016] [Indexed: 01/01/2023]
Abstract
Nucleic acid-based aptamers are considered to be a promising alternative to antibodies because of their strong and specific binding to diverse targets, fast and inexpensive chemical synthesis, and easy labeling with a fluorescent dye or therapeutic agent. Cluster of differentiation (CD) proteins are among the most popular antigens for aptamers on the cell surface. These anti-CD aptamers could be used in cell biology and biomedicine, from simple cell phenotyping by flow cytometry or fluorescent microscopy to diagnosis and treatment of HIV/AIDS to cancer and immune therapies. The unique feature of aptamers is that they can act simultaneously as an agonist and antagonist of CD receptors depending on a degree of aptamer oligomerization. Aptamers can also deliver small interfering RNA to silence vital genes in CD-positive cells. In this review, we summarize nucleic acid sequences of anti-CD aptamers and their use, which have been validated in multiple studies.
Collapse
|
34
|
Brennan FR, Kiessling A. Translational immunotoxicology of immunomodulatory monoclonal antibodies. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:85-93. [PMID: 27978992 DOI: 10.1016/j.ddtec.2016.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 12/28/2022]
Abstract
While immunomodulatory monoclonal antibodies (mAbs) have a wide therapeutic potential, exaggerated immunopharmacology may drive both acute and delayed immunotoxicity. The existing tools for immunotoxicity assessment do not accurately predict the full range of immunotoxicities observed in humans. New and optimized models, assays, endpoints and biomarkers in animals and humans are required to safeguard patients and allow them access to these often transformational therapies.
Collapse
|
35
|
Dharmaraj VL, Godfrin PD, Liu Y, Hudson SD. Rheology of clustering protein solutions. BIOMICROFLUIDICS 2016; 10:043509. [PMID: 27478524 PMCID: PMC4947037 DOI: 10.1063/1.4955162] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/22/2016] [Indexed: 05/26/2023]
Abstract
High viscosity is a major challenge with protein therapeutics at extremely high concentrations. To overcome this obstacle, it is essential to understand the relationship between the concentration of a protein solution and its viscosity as a function of shear rate and temperature. Here, lysozyme is a model charged globular protein having both short-ranged attraction (SA) and long-ranged repulsion (LR) that promote the formation of dynamic clusters at high concentrations. We report viscosity measurements from a micro-capillary rheometer (using only several microliters of solution) over a wide range of lysozyme solution concentrations, shear rates, and temperatures. Solution structural relaxation dynamics are also probed by dynamic light scattering (DLS). As a result of lysozyme's SALR interactions, the viscosity increased dramatically across all shear rates with increasing concentration and decreasing temperature. While most of the solutions exhibited Newtonian behavior, shear thinning was exhibited at the highest concentration (480 g/l) and lowest temperatures at shear rates above approximately 10(4 )s(-1). The onset shear rate for thinning and a structural relaxation rate estimated from a slow-mode measured by DLS are compared. These measurements provide insights into the properties of protein solutions and their microscopic structural origins.
Collapse
Affiliation(s)
| | | | | | - Steven D Hudson
- Polymers and Complex Fluids Group, Materials Science and Engineering Division, National Institute of Standards and Technology , Gaithersburg, Maryland 20899, USA
| |
Collapse
|
36
|
Vultaggio A, Matucci A, Nencini F, Pratesi S, Maggi E. Hypersensitivity Reactions to Biologicals: True Allergy? CURRENT TREATMENT OPTIONS IN ALLERGY 2016. [DOI: 10.1007/s40521-016-0082-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
37
|
Sediq AS, van Duijvenvoorde RB, Jiskoot W, Nejadnik MR. No Touching! Abrasion of Adsorbed Protein Is the Root Cause of Subvisible Particle Formation During Stirring. J Pharm Sci 2016; 105:519-529. [PMID: 26869415 DOI: 10.1016/j.xphs.2015.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 08/07/2015] [Accepted: 10/05/2015] [Indexed: 01/15/2023]
Abstract
This study addressed the effect of contact sliding during stirring of a monoclonal antibody solution on protein aggregation, in particular, in the nanometer and micrometer size range. An overhead stirring set-up was designed in which the presence and magnitude of the contact between the stir bar and the container could be manipulated. A solution of 0.1 mg/mL of a monoclonal antibody (IgG) in phosphate buffered saline was stirred at 300 rpm at room temperature. At different time points, samples were taken and analyzed by nanoparticle tracking analysis, flow imaging microscopy, and size-exclusion chromatography. In contrast to non-contact-stirred and unstirred samples, the contact-stirred sample contained several-fold more particles and showed a significant loss of monomer. No increase in oligomer content was detected. The number of particles formed was proportional to the contact area and the magnitude of the normal pressure between the stir bar and the glass container. Extrinsic 9-(2,2-dicyanovinyl) julolidine fluorescence indicated a conformational change for contact-stirred protein samples. Presence of polysorbate 20 inhibited the formation of micron-sized aggregates. We suggest a model in which abrasion of the potentially destabilized, adsorbed protein leads to aggregation and renewal of the surface for adsorption of a fresh protein layer.
Collapse
Affiliation(s)
- Ahmad S Sediq
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300 RA, The Netherlands
| | - R B van Duijvenvoorde
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300 RA, The Netherlands
| | - Wim Jiskoot
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300 RA, The Netherlands.
| | - M Reza Nejadnik
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300 RA, The Netherlands.
| |
Collapse
|
38
|
Gahoual R, Biacchi M, Chicher J, Kuhn L, Hammann P, Beck A, Leize-Wagner E, François YN. Monoclonal antibodies biosimilarity assessment using transient isotachophoresis capillary zone electrophoresis-tandem mass spectrometry. MAbs 2015; 6:1464-73. [PMID: 25484058 DOI: 10.4161/mabs.36305] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Out of all categories, monoclonal antibody (mAb) therapeutics attract the most interest due to their strong therapeutic potency and specificity. Six of the 10 top-selling drugs are antibody-based therapeutics that will lose patent protection soon. The European Medicines Agency has pioneered the regulatory framework for approval of biosimilar products and approved the first biosimilar antibodies by the end of 2013. As highly complex glycoproteins with a wide range of micro-variants, mAbs require extensive characterization through multiple analytical methods for structure assessment rendering manufacturing control and biosimilarity studies particularly product and time-consuming. Here, capillary zone electrophoresis coupled to mass spectrometry by a sheathless interface (CESI-MS) was used to characterize marketed reference mAbs and their respective biosimilar candidate simultaneously over different facets of their primary structure. CESI-MS/MS data were compared between approved mAbs and their biosimilar candidates to prove/disconfirm biosimilarity regarding recent regulation directives. Using only a single sample injection of 200 fmol, CESI-MS/MS data enabled 100% amino acids (AA) sequence characterization, which allows a difference of even one AA between 2 samples to be distinguished precisely. Simultaneously glycoforms were characterized regarding their structures and position through fragmentation spectra and glycoforms semiquantitative analysis was established, showing the capacity of the developed methodology to detect up to 16 different glycans. Other posttranslational modifications hotspots were characterized while their relative occurrence levels were estimated and compared to biosimilars. These results proved the value of using CESI-MS because the separation selectivity and ionization efficiency provided by the system allowed substantial improvement in the characterization workflow robustness and accuracy. Biosimilarity assessment could be performed routinely with a single injection of each candidate enabling improvements in the biosimilar development pipeline.
Collapse
Affiliation(s)
- Rabah Gahoual
- a Laboratoire de spectrométrie de masse des interactions et des systèmes (LSMIS); CNRS - UMR 7140 , Université de Strasbourg , Strasbourg Cedex , France
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Giavina-Bianchi P, Aun MV, Galvão VR, Castells M. Rapid Desensitization in Immediate Hypersensitivity Reaction to Drugs. CURRENT TREATMENT OPTIONS IN ALLERGY 2015. [DOI: 10.1007/s40521-015-0060-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
40
|
Therapeutic applications of nanomedicine in autoimmune diseases: From immunosuppression to tolerance induction. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1003-18. [DOI: 10.1016/j.nano.2014.12.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/17/2014] [Accepted: 12/03/2014] [Indexed: 01/13/2023]
|
41
|
Hifumi E, Arakawa M, Matsumoto S, Yamamoto T, Katayama Y, Uda T. Biochemical features and antiviral activity of a monomeric catalytic antibody light-chain 23D4 against influenza A virus. FASEB J 2015; 29:2347-58. [PMID: 25713031 DOI: 10.1096/fj.14-264275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/26/2015] [Indexed: 02/02/2023]
Abstract
Catalytic antibodies have exhibited interesting functions against some infectious viruses such as HIV, rabies virus, and influenza virus in vitro as well as in vivo. In some cases, a catalytic antibody light chain takes on several structures from the standpoint of molecular size (monomer, dimer, etc.) and/or isoelectronic point. In this study, we prepared a monomeric 23D4 light chain by mutating the C-terminal Cys to Ala of the wild-type. The mutated 23D4 molecule took a simple monomeric form, which could hydrolyze synthetic 4-methyl-coumaryl-7-amide substrates and a plasmid DNA. Because the monomeric 23D4 light chain suppressed the infection of influenza virus A/Hiroshima/37/2001 in an in vitro assay, the corresponding experiments were conducted in vivo, after the virus strain (which was taken from a human patient) was successfully adapted into BALB/cN Sea mice. In the experiments, a mixture of the monomeric 23D4 and the virus was nasally administered 1) with preincubation and 2) without preincubation. As a result, the monomeric 23D4 clearly exhibited the ability to suppress the influenza virus infection in both cases, indicating a potential drug for preventing infection of the influenza A virus.
Collapse
Affiliation(s)
- Emi Hifumi
- *Research Promotion Institute, Oita University, Oita-shi, Oita, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Kawaguchi, Saitama, Japan; Faculty of Medicine, Oita University, Yufu-city, Oita, Japan; Department of Applied Chemistry; Oita University, Oita-shi, Oita, Japan; Graduate School of Systems Life Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan; and Institute of Systems, Information Technologies and Nanotechnologies, Nanotechnology Laboratory, Fukuoka, Japan
| | - Mitsue Arakawa
- *Research Promotion Institute, Oita University, Oita-shi, Oita, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Kawaguchi, Saitama, Japan; Faculty of Medicine, Oita University, Yufu-city, Oita, Japan; Department of Applied Chemistry; Oita University, Oita-shi, Oita, Japan; Graduate School of Systems Life Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan; and Institute of Systems, Information Technologies and Nanotechnologies, Nanotechnology Laboratory, Fukuoka, Japan
| | - Shingo Matsumoto
- *Research Promotion Institute, Oita University, Oita-shi, Oita, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Kawaguchi, Saitama, Japan; Faculty of Medicine, Oita University, Yufu-city, Oita, Japan; Department of Applied Chemistry; Oita University, Oita-shi, Oita, Japan; Graduate School of Systems Life Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan; and Institute of Systems, Information Technologies and Nanotechnologies, Nanotechnology Laboratory, Fukuoka, Japan
| | - Tatsuhiro Yamamoto
- *Research Promotion Institute, Oita University, Oita-shi, Oita, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Kawaguchi, Saitama, Japan; Faculty of Medicine, Oita University, Yufu-city, Oita, Japan; Department of Applied Chemistry; Oita University, Oita-shi, Oita, Japan; Graduate School of Systems Life Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan; and Institute of Systems, Information Technologies and Nanotechnologies, Nanotechnology Laboratory, Fukuoka, Japan
| | - Yoshiki Katayama
- *Research Promotion Institute, Oita University, Oita-shi, Oita, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Kawaguchi, Saitama, Japan; Faculty of Medicine, Oita University, Yufu-city, Oita, Japan; Department of Applied Chemistry; Oita University, Oita-shi, Oita, Japan; Graduate School of Systems Life Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan; and Institute of Systems, Information Technologies and Nanotechnologies, Nanotechnology Laboratory, Fukuoka, Japan
| | - Taizo Uda
- *Research Promotion Institute, Oita University, Oita-shi, Oita, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Kawaguchi, Saitama, Japan; Faculty of Medicine, Oita University, Yufu-city, Oita, Japan; Department of Applied Chemistry; Oita University, Oita-shi, Oita, Japan; Graduate School of Systems Life Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan; and Institute of Systems, Information Technologies and Nanotechnologies, Nanotechnology Laboratory, Fukuoka, Japan
| |
Collapse
|
42
|
Ovacik AM. Network biology in development of monoclonal antibody therapeutics. Math Biosci 2014; 260:6-10. [PMID: 25311982 DOI: 10.1016/j.mbs.2014.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022]
Abstract
Monoclonal antibodies (mAbs) are large glycoproteins that recognize and remove/neutralize a specific target. Inflammation and inflammatory diseases are often treated with mAb-based therapeutics. Mathematical modeling is widely used in development of mAbs. Bioinformatics and structural modeling is used for humanization of mAbs and PK/PD modeling is extensively used in preclinical and clinical development. The objective of this commentary is to introduce systems biology-based modeling that can accelerate and improve development of mAbs.
Collapse
Affiliation(s)
- Ayse Meric Ovacik
- Merck Research Laboratories, 901 S. California Avenue, Palo Alto, CA 94304, USA .
| |
Collapse
|
43
|
|
44
|
Rajaii F, McCoy AN, Smith TJ. Cytokines are both villains and potential therapeutic targets in thyroid-associated ophthalmopathy: From bench to bedside. EXPERT REVIEW OF OPHTHALMOLOGY 2014; 9:227-234. [PMID: 25544859 DOI: 10.1586/17469899.2014.917960] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The pathophysiology underlying Graves' disease and its ocular manifestation, thyroid associated ophthalmopathy (TAO) is incompletely understood. Characterization of the mononuclear cells driving the disease and the cytokines they produce has led to significant advances in our understanding of TAO. This in turn has resulted in the identification of potentially attractive drug targets. For instance, development of inhibitors of specific cytokine pathways for use in other autoimmune diseases now presents an opportunity for their application in TAO. In this paper, we review the rationale for considering anti-cytokine therapy in TAO, evidence linking specific cytokines such as interleukin-6, tumor necrosis factor-α, and interleukin-17 pathways to TAO, and explore the potential for targeting of these pathways as therapy.
Collapse
Affiliation(s)
- Fatemeh Rajaii
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Allison N McCoy
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Terry J Smith
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI 48105.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105
| |
Collapse
|
45
|
Blocking monocyte transmigration in in vitro system by a human antibody scFv anti-CD99. Efficient large scale purification from periplasmic inclusion bodies in E. coli expression system. J Immunol Methods 2014; 408:35-45. [PMID: 24798881 DOI: 10.1016/j.jim.2014.04.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 04/24/2014] [Accepted: 04/25/2014] [Indexed: 11/21/2022]
Abstract
Migration of leukocytes into site of inflammation involves several steps mediated by various families of adhesion molecules. CD99 play a significant role in transendothelial migration (TEM) of leukocytes. Inhibition of TEM by specific monoclonal antibody (mAb) can provide a potent therapeutic approach to treating inflammatory conditions. However, the therapeutic utilization of whole IgG can lead to an inappropriate activation of Fc receptor-expressing cells, inducing serious adverse side effects due to cytokine release. In this regard, specific recombinant antibody in single chain variable fragments (scFvs) originated by phage library may offer a solution by affecting TEM function in a safe clinical context. However, this consideration requires large scale production of functional scFv antibodies and the absence of toxic reagents utilized for solubilization and refolding step of inclusion bodies that may discourage industrial application of these antibody fragments. In order to apply the scFv anti-CD99 named C7A in a clinical setting, we herein describe an efficient and large scale production of the antibody fragments expressed in E. coli as periplasmic insoluble protein avoiding gel filtration chromatography approach, and laborious refolding step pre- and post-purification. Using differential salt elution which is a simple, reproducible and effective procedure we are able to separate scFv in monomer format from aggregates. The purified scFv antibody C7A exhibits inhibitory activity comparable to an antagonistic conventional mAb, thus providing an excellent agent for blocking CD99 signaling. This protocol can be useful for the successful purification of other monomeric scFvs which are expressed as periplasmic inclusion bodies in bacterial systems.
Collapse
|