1
|
Lee C, Kuo W, Chang Y, Hsu S, Wu C, Chen Y, Chang J, Wang AH. Structure-based development of a canine TNF-α-specific antibody using adalimumab as a template. Protein Sci 2024; 33:e4873. [PMID: 38111376 PMCID: PMC10804672 DOI: 10.1002/pro.4873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023]
Abstract
The canine anti-tumor necrosis factor-alpha (TNF-α) monoclonal antibody is a potential therapeutic option for treating canine arthritis. The current treatments for arthritis in dogs have limitations due to side effects, emphasizing the need for safer and more effective therapies. The crystal structure of canine TNF-α (cTNF-α) was successfully determined at a resolution of 1.85 Å, and the protein was shown to assemble as a trimer, with high similarity to the functional quaternary structure of human TNF-α (hTNF-α). Adalimumab (Humira), a known TNF-α inhibitor, effectively targets and neutralizes TNF-α to reduce inflammation and has been used to manage autoimmune conditions such as rheumatoid arthritis. By comparing the structure of cTNF-α with the complex structure of hTNF-α and adalimumab-Fab, the epitope of adalimumab on cTNF-α was identified. The significant structural similarities of epitopes in cTNF-α and hTNF-α indicate the potential of using adalimumab to target cTNF-α. Therefore, a canine/human chimeric antibody, Humivet-R1, was created by grafting the variable domain of adalimumab onto a canine antibody framework derived from ranevetmab. Humivet-R1 exhibits potent neutralizing ability (IC50 = 0.05 nM) and high binding affinity (EC50 = 0.416 nM) to cTNF-α, comparable to that of adalimumab for both hTNF-α and cTNF-α. These results strongly suggest that Humivet-R1 has the potential to provide effective treatment for canine arthritis with reduced side effects. Here, we propose a structure-guided antibody design for the use of a chimeric antibody to treat canine inflammatory disease. Our successful development strategy can speed up therapeutic antibody discovery for animals and has the potential to revolutionize veterinary medicine.
Collapse
Affiliation(s)
- Cheng‐Chung Lee
- Institute of Biological Chemistry, Academia SinicaTaipeiTaiwan
- The Ph.D. Program for Translational MedicineCollege of Medical Science and Technology, Taipei Medical UniversityTaipeiTaiwan
| | - Wen‐Chih Kuo
- Institute of Biological Chemistry, Academia SinicaTaipeiTaiwan
| | - Ya‐Wen Chang
- The Ph.D. Program for Translational MedicineCollege of Medical Science and Technology, Taipei Medical UniversityTaipeiTaiwan
| | - Shu‐Fang Hsu
- The Ph.D. Program for Translational MedicineCollege of Medical Science and Technology, Taipei Medical UniversityTaipeiTaiwan
| | - Chia‐Hung Wu
- Traditional Chinese Veterinary Medicine, China Medical UniversityTaichungTaiwan
| | - Ya‐Wen Chen
- The Ph.D. Program for Translational MedicineCollege of Medical Science and Technology, Taipei Medical UniversityTaipeiTaiwan
| | - Jui‐Jen Chang
- Graduate Institute of Integrated Medicine, China Medical UniversityTaichungTaiwan
- Department of Medical ResearchChina Medical University HospitalTaichungTaiwan
| | - Andrew H.‐J. Wang
- Institute of Biological Chemistry, Academia SinicaTaipeiTaiwan
- The Ph.D. Program for Translational MedicineCollege of Medical Science and Technology, Taipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
2
|
Cao Y, Harvey BP, Jin L, Westmoreland S, Wang J, Puri M, Yang Y, Robb HM, Tanriverdi S, Hu C, Wang X, Xin X, Liu Y, Macoritto MP, Smith KM, Tian Y, White K, Radstake TR, Kaymakcalan Z. Therapeutic TNF Inhibitors Exhibit Differential Levels of Efficacy in Accelerating Cutaneous Wound Healing. JID INNOVATIONS 2024; 4:100250. [PMID: 38226320 PMCID: PMC10788510 DOI: 10.1016/j.xjidi.2023.100250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 10/21/2023] [Accepted: 11/16/2023] [Indexed: 01/17/2024] Open
Abstract
Adalimumab but neither etanercept nor certolizumab-pegol has been reported to induce a wound-healing profile in vitro by regulating macrophage differentiation and matrix metalloproteinase expression, which may underlie the differences in efficacy between various TNF-α inhibitors in impaired wound healing in patients with hidradenitis suppurativa, a chronic inflammatory skin disease. To examine and compare the efficacy of various TNF inhibitors in cutaneous wound healing in vivo, a human TNF knock-in Leprdb/db mouse model was established to model the impaired cutaneous wound healing as seen in hidradenitis suppurativa. The vehicle group exhibited severe impairments in cutaneous wound healing. In contrast, adalimumab significantly accelerated healing, confirmed by both histologic assessment and a unique healing transcriptional profile. Moreover, adalimumab and infliximab showed similar levels of efficacy, but golimumab was less effective, along with etanercept and certolizumab-pegol. In line with histologic assessments, proteomics analyses from healing wounds exposed to various TNF inhibitors revealed distinct and differential wound-healing signatures that may underlie the differential efficacy of these inhibitors in accelerating cutaneous wound healing. Taken together, these data revealed that TNF inhibitors exhibited differential levels of efficacy in accelerating cutaneous wound healing in the impaired wound-healing model in vivo.
Collapse
Affiliation(s)
- Yonghao Cao
- Transformational and Translational Immunology Discovery, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Bohdan P. Harvey
- Transformational and Translational Immunology Discovery, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Liang Jin
- DMPK-BA, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Susan Westmoreland
- Phamacology and Pathology, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Jing Wang
- Immunology Computational Biology, AbbVie Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Munish Puri
- Phamacology and Pathology, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Yingli Yang
- Phamacology and Pathology, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Holly M. Robb
- Discovery Research, AbbVie, North Chicago, Illinois, USA
| | - Sultan Tanriverdi
- Transformational and Translational Immunology Discovery, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Chenqi Hu
- DMPK-BA, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Xue Wang
- DMPK-BA, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Xiaofeng Xin
- Global Biologics, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Yingchun Liu
- Immunology Computational Biology, AbbVie Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Michael P. Macoritto
- Immunology Computational Biology, AbbVie Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Kathleen M. Smith
- Immunology Computational Biology, AbbVie Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Yu Tian
- DMPK-BA, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Kevin White
- Transformational and Translational Immunology Discovery, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Timothy R.D.J. Radstake
- Transformational and Translational Immunology Discovery, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Zehra Kaymakcalan
- Transformational and Translational Immunology Discovery, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| |
Collapse
|
3
|
Vanamee ÉS, Faustman DL. The benefits of clustering in TNF receptor superfamily signaling. Front Immunol 2023; 14:1225704. [PMID: 37662920 PMCID: PMC10469783 DOI: 10.3389/fimmu.2023.1225704] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
The tumor necrosis factor (TNF) receptor superfamily is a structurally and functionally related group of cell surface receptors that play crucial roles in various cellular processes, including apoptosis, cell survival, and immune regulation. This review paper synthesizes key findings from recent studies, highlighting the importance of clustering in TNF receptor superfamily signaling. We discuss the underlying molecular mechanisms of signaling, the functional consequences of receptor clustering, and potential therapeutic implications of targeting surface structures of receptor complexes.
Collapse
Affiliation(s)
- Éva S. Vanamee
- Immunobiology Department, Massachusetts General Hospital, Boston, MA, United States
| | - Denise L. Faustman
- Immunobiology Department, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Kinzer JL, Halseth TA, Kang J, Kim SY, Kumaran P, Ford M, Saveliev S, Skilton SJ, Schwendeman A. Physicochemical characterization and functionality comparison of Humira®(adalimumab), Remicade®(infliximab) and Simponi Aria®(golimumab). Int J Pharm 2023; 635:122646. [PMID: 36709835 DOI: 10.1016/j.ijpharm.2023.122646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/14/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
FDA-approved anti-TNFα biopharmaceuticals are successful in treating a range of autoimmune diseases. However, not all anti-TNFα products are identical in their patient outcomes, suggesting that there may be product-specific differences stemming from protein structural differences, doses and routes of administration. In this work, we focus only on structural and functional differences across three full-length anti-TNFα mAbs (Humira®, Remicade®, and Simponi Aria®) to better understand the implications of such differences on the products' efficacy. For structural characterization, we quantified N-glycans using mass spectrometry and fluorescence labeling. From these studies, we observed that Remicade® had the highest percent of afucosylated glycans (15.5 ± 1.3 %) and the largest number of unique glycans, 28. While Humira® had the fewest unique glycans, 15, and 11.4 ± 0.8 % of afucosylated, high-mannose glycans. For the functional studies we tested TNFα binding via ELISA, FcγRIIIa binding via AlphaLISA and effector function using an ADCC bioreporter assay. Humira® had a significantly lower EC50 (1.9 ± 0.1 pM) for ELISA and IC50 (10.5 ± 1.1 nM) for AlphaLISA, suggesting that Humira® has higher TNFα and FcγRIIIa binding affinity than Remicade® and Simponi Aria®. Humira® was also the most potent in the bioreporter assay with an EC50 value of 0.55 ± 0.03 nM compared to Remicade® (0.64 ± 0.04 nM) and Simponi Aria® (0.67 ± 0.03 nM). This comparison is significant as it highlights functional differences between mAbs with shared mechanisms of action when examined in a single laboratory and under one set of conditions.
Collapse
Affiliation(s)
- Jill L Kinzer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States
| | - Troy A Halseth
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States
| | - Jukyung Kang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States
| | - Sang Yeop Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States
| | - Preethi Kumaran
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States
| | - Michael Ford
- MS Bioworks, 3950 Varsity Dr, Ann Arbor, MI 48108, United States
| | - Sergei Saveliev
- Promega Corporation, 2800 Woods Hollow Rd, Madison, WI 53711, United States
| | - St John Skilton
- Protein Metrics, 20863 Stevens Creek Blvd #450, Cupertino, CA 95014, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States; Biointerfaces Institute, NCRC, 2800 Plymouth Rd, Ann Arbor, MI 48109, United States.
| |
Collapse
|
5
|
Casasola-LaMacchia A, Seward RJ, Tourdot S, Willetts M, Kruppa G, Agostino MJ, Bergeron G, Ahyi-Amendah N, Ciarla A, Lu Z, Kim HY, Hickling TP, Neubert H. HLAII peptide presentation of infliximab increases when complexed with TNF. Front Immunol 2022; 13:932252. [PMID: 36177046 PMCID: PMC9513746 DOI: 10.3389/fimmu.2022.932252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
CD4+ T-cell activation through recognition of Human Leukocyte Antigen II (HLAII)-presented peptides is a key step in the development of unwanted immune response against biotherapeutics, such as the generation of anti-drug antibodies (ADA). Therefore, the identification of HLAII-presented peptides derived from biotherapeutics is a crucial part of immunogenicity risk assessment and mitigation strategies during drug development. To date, numerous CD4+ T-cell epitopes have been identified by HLAII immunopeptidomics in antibody-based biotherapeutics using either their native or aggregated form. Antibody-target immune complexes have been detected in patients with ADA and are thought to play a role in ADA development by enhancing the presentation of CD4+ T-cell epitopes at the surface of antigen presenting cells (APCs). The aim of this study was to investigate the effect of biotherapeutic antibody-target immune complexes on the HLAII peptide presentation of biotherapeutics in human primary monocyte-derived dendritic cells (DCs). The trimeric tumor necrosis factor (TNF) and its biotherapeutic antagonists infliximab (INFL), adalimumab (ADAL), and a single armed Fab' were used as a model system. The HLAII immunopeptidome of DCs loaded with antagonists or their immune complexes with TNF was analyzed by trapped ion mobility time-of-flight mass spectrometry (timsTOF MS) leading to the identification of ~ 12,000 unique HLAII-associated peptides per preparation. Anti-TNF sequences were detected at a median of 0.3% of the total immunopeptidome, against a majority background of peptides from endogenous and media-derived proteins. TNF antagonist presentation spanned the variable and constant regions in a widespread manner in both light and heavy chains, consistent with previously discovered HLAII peptides. This investigation extends the collection of observed HLAII peptides from anti-TNF biotherapeutics to include sequences that at least partially span the complementary determining regions (CDRs), such as the LCDR1 for both INFL and ADAL. Although antagonist presentation varied significantly across donors, peptides from both bivalent antagonists INFL and ADAL were more highly presented relative to the Fab'. While TNF immune complexes did not alter overall HLAII presentation, a moderate increase in presentation of a subset of peptide clusters was observed in the case of INFL-TNF, which included HCDR2, HCDR3 and LCDR2 sequences.
Collapse
Affiliation(s)
- Andrea Casasola-LaMacchia
- BioMedicine Design, Worldwide Research, Development and Medical, Pfizer Inc., Andover, MA, United States
| | - Robert Joseph Seward
- BioMedicine Design, Worldwide Research, Development and Medical, Pfizer Inc., Andover, MA, United States
| | - Sophie Tourdot
- BioMedicine Design, Worldwide Research, Development and Medical, Pfizer Inc., Andover, MA, United States
| | | | - Gary Kruppa
- Bruker Daltonics, Billerica, MA, United States
| | | | - Gabrielle Bergeron
- BioMedicine Design, Worldwide Research, Development and Medical, Pfizer Inc., Andover, MA, United States
| | - Nathalie Ahyi-Amendah
- BioMedicine Design, Worldwide Research, Development and Medical, Pfizer Inc., Andover, MA, United States
| | - Andrew Ciarla
- BioMedicine Design, Worldwide Research, Development and Medical, Pfizer Inc., Andover, MA, United States
| | - Zhaojiang Lu
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, MA, United States
| | - Hai-Young Kim
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, MA, United States
| | - Timothy P. Hickling
- BioMedicine Design, Worldwide Research, Development and Medical, Pfizer Inc., Andover, MA, United States
| | - Hendrik Neubert
- BioMedicine Design, Worldwide Research, Development and Medical, Pfizer Inc., Andover, MA, United States
| |
Collapse
|
6
|
Marcantonio DH, Matteson A, Presler M, Burke JM, Hagen DR, Hua F, Apgar JF. Early Feasibility Assessment: A Method for Accurately Predicting Biotherapeutic Dosing to Inform Early Drug Discovery Decisions. Front Pharmacol 2022; 13:864768. [PMID: 35754500 PMCID: PMC9214263 DOI: 10.3389/fphar.2022.864768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/16/2022] [Indexed: 11/18/2022] Open
Abstract
The application of model-informed drug discovery and development (MID3) approaches in the early stages of drug discovery can help determine feasibility of drugging a target, prioritize between targets, or define optimal drug properties for a target product profile (TPP). However, applying MID3 in early discovery can be challenging due to the lack of pharmacokinetic (PK) and pharmacodynamic (PD) data at this stage. Early Feasibility Assessment (EFA) is the application of mechanistic PKPD models, built from first principles, and parameterized by data that is readily available early in drug discovery to make effective dose predictions. This manuscript demonstrates the ability of EFA to make accurate predictions of clinical effective doses for nine approved biotherapeutics and outlines the potential of extending this approach to novel therapeutics to impact early drug discovery decisions.
Collapse
|
7
|
Ishiwatari-Ogata C, Kyuuma M, Ogata H, Yamakawa M, Iwata K, Ochi M, Hori M, Miyata N, Fujii Y. Ozoralizumab, a Humanized Anti-TNFα NANOBODY ® Compound, Exhibits Efficacy Not Only at the Onset of Arthritis in a Human TNF Transgenic Mouse but Also During Secondary Failure of Administration of an Anti-TNFα IgG. Front Immunol 2022; 13:853008. [PMID: 35273620 PMCID: PMC8902368 DOI: 10.3389/fimmu.2022.853008] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/01/2022] [Indexed: 12/19/2022] Open
Abstract
Although the introduction of tumor necrosis factor (TNF) inhibitors represented a significant advance in the treatment of rheumatoid arthritis (RA), traditional anti-TNFα antibodies are somewhat immunogenic, and their use results in the formation of anti-drug antibodies (ADAs) and loss of efficacy (secondary failure). Ozoralizumab is a trivalent, bispecific NANOBODY® compound that differs structurally from IgGs. In this study we investigated the suppressant effect of ozoralizumab and adalimumab, an anti-TNFα IgG, on arthritis and induction of ADAs in human TNF transgenic mice. Ozoralizumab markedly suppressed arthritis progression and did not induce ADAs during long-term administration. We also developed an animal model of secondary failure by repeatedly administering adalimumab and found that switching from adalimumab to ozoralizumab was followed by superior anti-arthritis efficacy in the secondary-failure animal model. Moreover, ozoralizumab did not form large immune complexes that might lead to ADA formation. The results of our studies suggest that ozoralizumab, which exhibited low immunogenicity in the animal model used and has a different antibody structure from that of IgGs, is a promising candidate for the treatment of RA patients not only at the onset of RA but also during secondary failure of anti-TNFα treatment.
Collapse
Affiliation(s)
| | - Masanao Kyuuma
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Hitoshi Ogata
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Machi Yamakawa
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Katsuya Iwata
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Motoki Ochi
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Miyuki Hori
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Noriyuki Miyata
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Yasuyuki Fujii
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| |
Collapse
|
8
|
Ternant D, Le Tilly O, Picon L, Moussata D, Passot C, Bejan-Angoulvant T, Desvignes C, Mulleman D, Goupille P, Paintaud G. Infliximab Efficacy May Be Linked to Full TNF-α Blockade in Peripheral Compartment-A Double Central-Peripheral Target-Mediated Drug Disposition (TMDD) Model. Pharmaceutics 2021; 13:pharmaceutics13111821. [PMID: 34834236 PMCID: PMC8623740 DOI: 10.3390/pharmaceutics13111821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/03/2022] Open
Abstract
Infliximab is an anti-TNF-α monoclonal antibody approved in chronic inflammatory bowel diseases (IBD). This study aimed at providing an in-depth description of infliximab target-mediated pharmacokinetics in 133 IBD patients treated with 5 mg/kg infliximab at weeks 0, 2, 14, and 22. A two-compartment model with double target-mediated drug disposition (TMDD) in both central and peripheral compartments was developed, using a rich database of 26 ankylosing spondylitis patients as a reference for linear elimination kinetics. Population approach and quasi-steady-state (QSS) approximation were used. Concentration-time data were satisfactorily described using the double-TMDD model. Target-mediated parameters of central and peripheral compartments were respectively baseline TNF concentrations (RC0 = 3.3 nM and RP0 = 0.46 nM), steady-stated dissociation rates (KCSS = 15.4 nM and KPSS = 0.49 nM), and first-order elimination rates of complexes (kCint = 0.17 day−1 and kPint = 0.0079 day−1). This model showed slower turnover of targets and infliximab-TNF complex elimination rate in peripheral compartment than in central compartment. This study allowed a better understanding of the multi-scale target-mediated pharmacokinetics of infliximab. This model could be useful to improve model-based therapeutic drug monitoring of infliximab in IBD patients.
Collapse
Affiliation(s)
- David Ternant
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
- Correspondence:
| | - Olivier Le Tilly
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
| | - Laurence Picon
- Department of Gastroenterology, CHRU de Tours, 37044 Tour, France;
| | - Driffa Moussata
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Gastroenterology, CHRU de Tours, 37044 Tour, France;
| | - Christophe Passot
- Département de Biopathologie, Institut de Cancérologie de l’Ouest, 49055 Angers, France;
| | - Theodora Bejan-Angoulvant
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
| | - Céline Desvignes
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
| | - Denis Mulleman
- EA 7501 “Groupe Innovation et Ciblage Cellulaire”, Université de Tours, 37044 Tour, France;
- Department of Rheumatology, CHRU de Tours, 37044 Tour, France;
| | | | - Gilles Paintaud
- EA 4245 “Transplantation, Immunology, Inflammation”, Université de Tours, 37044 Tours, France; (O.L.T.); (D.M.); (T.B.-A.); (C.D.); (G.P.)
- Department of Clinical Pharmacology, CHRU de Tours, 37044 Tour, France
| |
Collapse
|
9
|
Rouby G, Tran NT, Leblanc Y, Taverna M, Bihoreau N. Investigation of monoclonal antibody dimers in a final formulated drug by separation techniques coupled to native mass spectrometry. MAbs 2021; 12:e1781743. [PMID: 32633190 PMCID: PMC7531515 DOI: 10.1080/19420862.2020.1781743] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are highly complex proteins that must be exhaustively characterized according to the regulatory authorities' recommendations. MAbs display micro-heterogeneity mainly due to their post-translational modifications, but also to their susceptibility to chemical and physical degradations. Among these degradations, aggregation is quite frequent, initiated by protein denaturation and then dimer formation. Here, we investigated the nature and structure of the high molecular weight species (HMW) present at less than 1% in an unstressed formulated roledumab biopharmaceutical, as a model of high purity mAb. HMW species were first purified through preparative size-exclusion chromatography (SEC) and then analyzed by a combination of chromatographic methods (ion-exchange chromatography (IEX), SEC) coupled to native mass spectrometry (MS), as well as sodium dodecyl sulfate–polyacrylamide gel electrophoresis and capillary gel electrophoresis under non-reducing conditions. Both covalently and non-covalently bound dimers were identified at a proportion of 50/50. In-depth characterization of the HMW fraction by SEC and IEX hyphenated to native MS revealed the presence of three mAb dimer forms having the same mass, but differing by their charge and size. They were attributed to different compact and elongated dimers. Finally, high-resolution middle-up approaches using different enzymes (IdeS and IgdE) were performed to determine the mAb domains implicated in the dimerization. Our results revealed that the roledumab dimers were associated mainly by a single Fab-to-Fab arm-bound association.
Collapse
Affiliation(s)
- G Rouby
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay , 92296, Châtenay-Malabry, France.,Analytical Department, LFB , Courtaboeuf (Les Ulis), France
| | - N T Tran
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay , 92296, Châtenay-Malabry, France
| | - Y Leblanc
- Analytical Department, LFB , Courtaboeuf (Les Ulis), France
| | - M Taverna
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay , 92296, Châtenay-Malabry, France.,Institut Universitaire de France , Paris, France
| | - N Bihoreau
- Analytical Department, LFB , Courtaboeuf (Les Ulis), France
| |
Collapse
|
10
|
Cao Y, Harvey BP, Hong F, Ruzek M, Wang J, Murphy ER, Kaymakcalan Z. Adalimumab Induces a Wound Healing Profile in Patients with Hidradenitis Suppurativa by Regulating Macrophage Differentiation and Matrix Metalloproteinase Expression. J Invest Dermatol 2021; 141:2730-2740.e9. [PMID: 33965402 DOI: 10.1016/j.jid.2021.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 12/11/2022]
Abstract
Adalimumab (ADA) is the only Food and Drug Administration‒approved treatment for moderate-to-severe hidradenitis suppurativa, whereas etanercept and certolizumab-pegol have been shown to be ineffective, suggesting that the mechanism of action of ADA is distinct in hidradenitis suppurativa and may contribute to improved wound healing. Given that macrophages (Mϕs) play pivotal roles throughout the wound healing process, an in vitro Mϕ differentiation assay was carried out to assess the impact of TNF‒anti-TNF complexes on these cells. TNF‒ADA complexes exhibited stronger inhibitory effects on inflammatory Mϕ differentiation. Moreover, RNA sequencing revealed several unique wound healing profiles for TNF‒ADA‒treated inflammatory Mϕs, which were not observed for those treated with either TNF‒etanercept or TNF‒certolizumab-pegol complexes, including the inhibition of the matrix metalloproteinase (MMP) pathway. In addition, ADA administration was found to significantly reduce the levels of inflammatory MMP-1 and MMP-9 while promoting wound-healing MMP-13 and tissue inhibitor of metalloproteinases 2 levels in the circulation of the patients with hidradenitis suppurativa who responded to treatment. Our in vitro findings show that TNF‒ADA‒treated inflammatory Mϕs exhibit a distinct profile resembling wound healing. Moreover, ADA not only differentially regulates MMP expression in patients with hidradenitis suppurativa responding to the therapy but also potentially induces a transition to a profile suggestive of wound healing.
Collapse
Affiliation(s)
- Yonghao Cao
- Immunology Discovery, AbbVie Bioresearch Center, Worcester, Massachusetts, USA.
| | - Bohdan P Harvey
- Immunology Discovery, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Feng Hong
- Discovery and Early Pipeline Statistics, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Melanie Ruzek
- Translational Immunology, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Jing Wang
- Immunology Systems Computational Biology, Genomic Research Center, AbbVie Inc, Cambridge, Massachusetts, USA
| | - Erin R Murphy
- Genomic Research Center, AbbVie Inc, North Chicago, Illinois, USA
| | - Zehra Kaymakcalan
- Immunology Discovery, AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| |
Collapse
|
11
|
Pedersen ME, Haegebaert RMS, Østergaard J, Jensen H. Size-based characterization of adalimumab and TNF-α interactions using flow induced dispersion analysis: assessment of avidity-stabilized multiple bound species. Sci Rep 2021; 11:4754. [PMID: 33637878 PMCID: PMC7910425 DOI: 10.1038/s41598-021-84113-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/10/2021] [Indexed: 12/16/2022] Open
Abstract
The understanding and characterization of protein interactions is crucial for elucidation of complicated biomolecular processes as well as for the development of new biopharmaceutical therapies. Often, protein interactions involve multiple binding, avidity, oligomerization, and are dependent on the local environment. Current analytical methodologies are unable to provide a detailed mechanistic characterization considering all these parameters, since they often rely on surface immobilization, cannot measure under biorelevant conditions, or do not feature a structurally-related readout for indicating formation of multiple bound species. In this work, we report the use of flow induced dispersion analysis (FIDA) for in-solution characterization of complex protein interactions under in vivo like conditions. FIDA is an immobilization-free ligand binding methodology employing Taylor dispersion analysis for measuring the hydrodynamic radius (size) of biomolecular complexes. Here, the FIDA technology is utilized for a size-based characterization of the interaction between TNF-α and adalimumab. We report concentration-dependent complex sizes, binding affinities (Kd), kinetics, and higher order stoichiometries, thus providing essential information on the TNF-α-adalimumab binding mechanism. Furthermore, it is shown that the avidity stabilized complexes involving formation of multiple non-covalent bonds are formed on a longer timescale than the primary complexes formed in a simple 1 to 1 binding event.
Collapse
Affiliation(s)
- Morten E Pedersen
- Fida Biosystems ApS, Fruebjergvej 3, 2100, Copenhagen O, Denmark
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen O, Denmark
| | - Ragna M S Haegebaert
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen O, Denmark
| | - Jesper Østergaard
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen O, Denmark
| | - Henrik Jensen
- Fida Biosystems ApS, Fruebjergvej 3, 2100, Copenhagen O, Denmark.
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen O, Denmark.
| |
Collapse
|
12
|
Singrang N, Laophetsakunchai S, Tran BN, Matsudaira PT, Tassanakajon A, Wangkanont K. Biochemical and structural characterization of a recombinant fibrinogen-related lectin from Penaeus monodon. Sci Rep 2021; 11:2934. [PMID: 33536457 PMCID: PMC7858579 DOI: 10.1038/s41598-021-82301-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 01/19/2021] [Indexed: 11/25/2022] Open
Abstract
Fibrinogen-related lectins are carbohydrate-binding proteins of the innate immune system that recognize glycan structures on microbial surfaces. These innate immune lectins are crucial for invertebrates as they do not rely on adaptive immunity for pathogen clearance. Here, we characterize a recombinant fibrinogen-related lectin PmFREP from the black tiger shrimp Penaeus monodon expressed in the Trichoplusia ni insect cell. Electron microscopy and cross-linking experiments revealed that PmFREP is a disulfide-linked dimer of pentamers distinct from other fibrinogen-related lectins. The full-length protein binds N-acetyl sugars in a Ca2+ ion-independent manner. PmFREP recognized and agglutinated Pseudomonas aeruginosa. Weak binding was detected with other bacteria, including Vibrio parahaemolyticus, but no agglutination activity was observed. The biologically active PmFREP will not only be a crucial tool to elucidate the innate immune signaling in P. monodon and other economically important species, but will also aid in detection and prevention of shrimp bacterial infectious diseases.
Collapse
Affiliation(s)
- Nongnuch Singrang
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand.,Molecular Crop Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Sirasit Laophetsakunchai
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Bich Ngoc Tran
- Department of Biological Sciences, Faculty of Science, Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Paul T Matsudaira
- Department of Biological Sciences, Faculty of Science, Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Anchalee Tassanakajon
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Kittikhun Wangkanont
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand. .,Molecular Crop Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
13
|
de Oliveira TM, van Beek L, Shilliday F, Debreczeni JÉ, Phillips C. Cryo-EM: The Resolution Revolution and Drug Discovery. SLAS DISCOVERY 2020; 26:17-31. [PMID: 33016175 DOI: 10.1177/2472555220960401] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Single-particle cryogenic electron microscopy (cryo-EM) has been elevated to the mainstream of structural biology propelled by technological advancements in numerous fronts, including imaging analysis and the development of direct electron detectors. The drug discovery field has watched with (initial) skepticism and wonder at the progression of the technique and how it revolutionized the molecular understanding of previously intractable targets. This article critically assesses how cryo-EM has impacted drug discovery in diverse therapeutic areas. Targets that have been brought into the realm of structure-based drug design by cryo-EM and are thus reviewed here include membrane proteins like the GABAA receptor, several TRP channels, and G protein-coupled receptors, and multiprotein complexes like the ribosomes, the proteasome, and eIF2B. We will describe these studies highlighting the achievements, challenges, and caveats.
Collapse
Affiliation(s)
| | - Lotte van Beek
- Structure, Biophysics and FBLG, Discovery Sciences, AstraZeneca R&D, Cambridge, UK
| | - Fiona Shilliday
- Structure, Biophysics and FBLG, Discovery Sciences, AstraZeneca R&D, Cambridge, UK
| | - Judit É Debreczeni
- Structure, Biophysics and FBLG, Discovery Sciences, AstraZeneca R&D, Cambridge, UK
| | - Chris Phillips
- Structure, Biophysics and FBLG, Discovery Sciences, AstraZeneca R&D, Cambridge, UK
| |
Collapse
|
14
|
New insights on the interaction mechanism of rhTNFα with its antagonists Adalimumab and Etanercept. Biochem J 2020; 477:3299-3311. [DOI: 10.1042/bcj20200568] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 12/31/2022]
Abstract
TNFα is a pro-inflammatory cytokine that is a therapeutic target for inflammatory autoimmune disorders. Thus, TNFα antagonists are successfully used for the treatment of these disorders. Here, new association patterns of rhTNFα and its antagonists Adalimumab and Etanercept are disclosed. Active rhTNFα was purified by IMAC from the soluble fraction of transformed Escherichia coli. Protein detection was assessed by SDS–PAGE and Western blot. The KD values for rhTNFα interactions with their antagonists were obtained by non-competitive ELISA and by microscale thermophoresis (MST). Molecular sizes of the complexes were evaluated by size-exclusion chromatography-high performance liquid chromatography (SEC-HPLC). Surprisingly, both antagonists recognized the monomeric form of rhTNFα under reducing and non-reducing conditions, indicating unexpected bindings of the antagonists to linear epitopes and to rhTNFα monomers. For the first time, the interactions of rhTNFα with Adalimumab and Etanercept were assessed by MST, which allows evaluating molecular interactions in solution with a wide range of concentrations. Biphasic binding curves with low and high KD values (<10−9 M and >10−8 M) were observed during thermophoresis experiments, suggesting the generation of complexes with different stoichiometry, which were confirmed by SEC-HPLC. Our results demonstrated the binding of TNFα-antagonists with rhTNFα monomers and linear epitopes. Also, complexes of high molecular mass were observed. This pioneer investigation constitutes valuable data for future approaches into the study of the interaction mechanism of TNFα and its antagonists.
Collapse
|
15
|
Abstract
Biologic drugs have revolutionized the treatment of certain hematologic, autoimmune, and malignant diseases, but they may place patients at risk for reactivation or acquisition of tuberculosis. This risk is highest with the tumor necrosis factor-alpha (TNF-α) inhibitors. Amongst this class of drugs, the monoclonal antibodies (infliximab, adalimumab, golimumab) and antibody fragment (certolizumab) carry an increased risk compared to the soluble receptor fusion molecule, etanercept. Treatment of latent TB is critical to decrease the risk of reactivation. Data continues to emerge regarding tuberculosis risk associated with novel biologics targeting cytokines involved in tuberculosis control.
Collapse
|
16
|
Ono M, Horita S, Sato Y, Nomura Y, Iwata S, Nomura N. Structural basis for tumor necrosis factor blockade with the therapeutic antibody golimumab. Protein Sci 2018; 27:1038-1046. [PMID: 29575262 DOI: 10.1002/pro.3407] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/09/2018] [Accepted: 03/21/2018] [Indexed: 12/29/2022]
Abstract
Tumor necrosis factor α (TNFα) is a proinflammatory cytokine, and elevated levels of TNFα in serum are associated with various autoimmune diseases, including rheumatoid arthritis (RA), ankylosing spondylitis (AS), Crohn's disease (CD), psoriasis, and systemic lupus erythaematosus. TNFα performs its pleiotropic functions by binding to two structurally distinct transmembrane receptors, TNF receptor (TNFR) 1 and TNFR2. Antibody-based therapeutic strategies that block excessive TNFα signaling have been shown to be effective in suppressing such harmful inflammatory conditions. Golimumab (Simponi®) is an FDA-approved fully human monoclonal antibody targeting TNFα that has been widely used for the treatment of RA, AS, and CD. However, the structural basis underlying the inhibitory action of golimumab remains unclear. Here, we report the crystal structure of the Fv fragment of golimumab in complex with TNFα at a resolution of 2.73 Å. The resolved structure reveals that golimumab binds to a distinct epitope on TNFα that does not overlap with the binding residues of TNFR2. Golimumab exerts its inhibitory effect by preventing binding of TNFR1 and TNFR2 to TNFα by steric hindrance. Golimumab does not induce conformational changes in TNFα that could affect receptor binding. This mode of action is specific to golimumab among the four anti-TNFα therapeutic antibodies currently approved for clinical use.
Collapse
Affiliation(s)
- Masatsugu Ono
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shoichiro Horita
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yumi Sato
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yayoi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - So Iwata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.,RIKEN SPring-8 Center, Kouto, Sayo-cho, Sayo-gun, Hyogo, 679-5148, Japan
| | - Norimichi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
17
|
Lim H, Lee SH, Lee HT, Lee JU, Son JY, Shin W, Heo YS. Structural Biology of the TNFα Antagonists Used in the Treatment of Rheumatoid Arthritis. Int J Mol Sci 2018. [PMID: 29518978 PMCID: PMC5877629 DOI: 10.3390/ijms19030768] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The binding of the tumor necrosis factor α (TNFα) to its cognate receptor initiates many immune and inflammatory processes. The drugs, etanercept (Enbrel®), infliximab (Remicade®), adalimumab (Humira®), certolizumab-pegol (Cimzia®), and golimumab (Simponi®), are anti-TNFα agents. These drugs block TNFα from interacting with its receptors and have enabled the development of breakthrough therapies for the treatment of several autoimmune inflammatory diseases, including rheumatoid arthritis, Crohn's disease, and psoriatic arthritis. In this review, we describe the latest works on the structural characterization of TNFα-TNFα antagonist interactions related to their therapeutic efficacy at the atomic level. A comprehensive comparison of the interactions of the TNFα blockers would provide a better understanding of the molecular mechanisms by which they neutralize TNFα. In addition, an enhanced understanding of the higher order complex structures and quinary structures of the TNFα antagonists can support the development of better biologics with the improved pharmacokinetic properties. Accumulation of these structural studies can provide a basis for the improvement of therapeutic agents against TNFα for the treatment of rheumatoid arthritis and other autoimmune inflammatory diseases in which TNFα plays an important role in pathogenesis.
Collapse
Affiliation(s)
- Heejin Lim
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Sang Hyung Lee
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Hyun Tae Lee
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Jee Un Lee
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Ji Young Son
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Woori Shin
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Yong-Seok Heo
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| |
Collapse
|
18
|
Marino A, Real-Fernández F, Rovero P, Giani T, Pagnini I, Cimaz R, Simonini G. Anti-adalimumab antibodies in a cohort of patients with juvenile idiopathic arthritis: incidence and clinical correlations. Clin Rheumatol 2018; 37:1407-1411. [DOI: 10.1007/s10067-018-4057-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/21/2018] [Accepted: 02/27/2018] [Indexed: 10/17/2022]
|