1
|
de Lima JY, de Castro Andreassa E, Venturi Biembengut Í, de Arruda Campos Brasil de Souza T. Dissecting dual specificity: Identifying key residues in L-asparaginase for enhanced acute lymphoid leukemia therapy and reduced adverse effects. Int J Biol Macromol 2024; 254:127998. [PMID: 37949271 DOI: 10.1016/j.ijbiomac.2023.127998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
L-asparaginase from Escherichia coli (EcA) has been used for the treatment of acute lymphoid leukemia (ALL) since the 1970s. Nevertheless, the enzyme has a second specificity that results in glutaminase breakdown, resulting in depletion from the patient's body, causing severe adverse effects. Despite the huge interest in the use of this enzyme, the exact process of glutamine depletion is still unknown and there is no consensus regarding L-asparagine hydrolysis. Here, we investigate the role of T12, Y25, and T89 in asparaginase and glutaminase activities. We obtained individual clones containing mutations in the T12, Y25 or T89 residues. After the recombinant production of wild-type and mutated EcA, The purified samples were subjected to structural analysis using Nano Differential Scanning Fluorimetry, which revealed that all samples contained thermostable molecules in their active structural conformation, the homotetramer conformation. The quaternary conformation was confirmed by DLS and SEC. The activity enzymatic assay combined with molecular dynamics simulation identified the contribution of T12, Y25, and T89 residues in EcA glutaminase and asparaginase activities. Our results mapped the enzymatic behavior paving the way for the designing of improved EcA enzymes, which is important in the treatment of ALL.
Collapse
Affiliation(s)
- Jhenifer Yonara de Lima
- Structural and Computational Proteomics Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, PR 80320-290, Brazil.
| | - Emanuella de Castro Andreassa
- Structural and Computational Proteomics Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, PR 80320-290, Brazil
| | - Ísis Venturi Biembengut
- Structural and Computational Proteomics Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, PR 80320-290, Brazil
| | | |
Collapse
|
2
|
Yen CY, Wang PY, Chen KY, Tseng CC, Wu CC, Ou TT, Yen JH. HLA-DR genotypes in patients with systemic lupus erythematosus in Taiwan. J Chin Med Assoc 2023; 86:1060-1065. [PMID: 37801591 DOI: 10.1097/jcma.0000000000001009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Different human leukocyte antigen (HLA)-DR genotypes have been known to be associated with the risk of development of systemic lupus erythematosus (SLE) in different populations, although Lu et al. have reported previously that no correlation exists between the HLA-DR genotype and disease manifestation in SLE patients in Taiwan. We investigated the effects different HLA-DR genotypes had on SLE incidence in Taiwanese patients as to whether risk alleles were associated with different clinical manifestations, and the effects risk alleles had on the age of disease onset. METHODS Two hundred thirty-four SLE patients and 346 healthy controls were enrolled. HLA-DR genotyping was performed with the HLA FluoGene DRDQ kit for each subject. Chi-square tests and t tests were performed for statistical analysis. RESULTS HLA-DR2 was significantly more frequently found in SLE patients than in controls (odds ratio [OR] = 2.05, 95% CI, 1.44-2.92, p < 0.001). Notably, HLA-DR6 appeared to trend toward negative correlation with SLE, whereas HLA-DR8 appeared to trend toward positive correlation. HLA-DR2 patients had an earlier onset of disease as well as a higher prevalence of oral ulcer, avascular necrosis of bone, and renal involvement (lupus nephritis). CONCLUSION HLA-DR2 was associated with SLE susceptibility in this Taiwanese population as well as lower age of disease onset and more severe clinical manifestations.
Collapse
Affiliation(s)
- Chang-Yi Yen
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- Institute of Medical Informatics, College of Electrical Engineering and Computer Science, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Pin-Yi Wang
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Kuan-Yu Chen
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Chia-Chun Tseng
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Cheng-Chin Wu
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
| | - Tsan-Teng Ou
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
| | - Jeng-Hsien Yen
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan, ROC
- Department of Biomedical Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan, ROC
| |
Collapse
|
3
|
Ricotti S, Garay AS, Etcheverrigaray M, Amadeo GI, De Groot AS, Martin W, Mufarrege EF. Development of IFNβ-1a versions with reduced immunogenicity and full in vitro biological activity for the treatment of multiple sclerosis. Clin Immunol 2023; 257:109831. [PMID: 37931868 DOI: 10.1016/j.clim.2023.109831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/18/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
IFNβ (recombinant interferon Beta) has been widely used for the treatment of Multiple sclerosis for the last four decades. Despite the human origin of the IFNβ sequence, IFNβ is immunogenic, and unwanted immune responses in IFNβ-treated patients may compromise its efficacy and safety in the clinic. In this study, we applied the DeFT (De-immunization of Functional Therapeutics) approach to producing functional, de-immunized versions of IFNβ-1a. Two de-immunized versions of IFNβ-1a were produced in CHO cells and designated as IFNβ-1a(VAR1) and IFNβ-1a(VAR2). First, the secondary and tertiary protein structures were analyzed by circular dichroism spectroscopy. Then, the variants were also tested for functionality. While IFNβ-1a(VAR2) showed similar in vitro antiviral activity to the original protein, IFNβ-1a(VAR1) exhibited 40% more biological potency. Finally, in vivo assays using HLA-DR transgenic mice revealed that the de-immunized variants showed a markedly reduced immunogenicity when compared to the originator.
Collapse
Affiliation(s)
- Sonia Ricotti
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina
| | - Alberto Sergio Garay
- Laboratory of Molecular Modeling, FBCB (School of Biochemistry and Biological Sciences), Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina
| | - Marina Etcheverrigaray
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina
| | - Gabriel Ignacio Amadeo
- Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina
| | - Anne S De Groot
- EpiVax, Inc., Providence, RI 02903, USA; Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | | | - Eduardo Federico Mufarrege
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina.
| |
Collapse
|
4
|
Andlauer TFM, Link J, Martin D, Ryner M, Hermanrud C, Grummel V, Auer M, Hegen H, Aly L, Gasperi C, Knier B, Müller-Myhsok B, Jensen PEH, Sellebjerg F, Kockum I, Olsson T, Pallardy M, Spindeldreher S, Deisenhammer F, Fogdell-Hahn A, Hemmer B. Treatment- and population-specific genetic risk factors for anti-drug antibodies against interferon-beta: a GWAS. BMC Med 2020; 18:298. [PMID: 33143745 PMCID: PMC7641861 DOI: 10.1186/s12916-020-01769-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/28/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Upon treatment with biopharmaceuticals, the immune system may produce anti-drug antibodies (ADA) that inhibit the therapy. Up to 40% of multiple sclerosis patients treated with interferon β (IFNβ) develop ADA, for which a genetic predisposition exists. Here, we present a genome-wide association study on ADA and predict the occurrence of antibodies in multiple sclerosis patients treated with different interferon β preparations. METHODS We analyzed a large sample of 2757 genotyped and imputed patients from two cohorts (Sweden and Germany), split between a discovery and a replication dataset. Binding ADA (bADA) levels were measured by capture-ELISA, neutralizing ADA (nADA) titers using a bioassay. Genome-wide association analyses were conducted stratified by cohort and treatment preparation, followed by fixed-effects meta-analysis. RESULTS Binding ADA levels and nADA titers were correlated and showed a significant heritability (47% and 50%, respectively). The risk factors differed strongly by treatment preparation: The top-associated and replicated variants for nADA presence were the HLA-associated variants rs77278603 in IFNβ-1a s.c.- (odds ratio (OR) = 3.55 (95% confidence interval = 2.81-4.48), p = 2.1 × 10-26) and rs28366299 in IFNβ-1b s.c.-treated patients (OR = 3.56 (2.69-4.72), p = 6.6 × 10-19). The rs77278603-correlated HLA haplotype DR15-DQ6 conferred risk specifically for IFNβ-1a s.c. (OR = 2.88 (2.29-3.61), p = 7.4 × 10-20) while DR3-DQ2 was protective (OR = 0.37 (0.27-0.52), p = 3.7 × 10-09). The haplotype DR4-DQ3 was the major risk haplotype for IFNβ-1b s.c. (OR = 7.35 (4.33-12.47), p = 1.5 × 10-13). These haplotypes exhibit large population-specific frequency differences. The best prediction models were achieved for ADA in IFNβ-1a s.c.-treated patients. Here, the prediction in the Swedish cohort showed AUC = 0.91 (0.85-0.95), sensitivity = 0.78, and specificity = 0.90; patients with the top 30% of genetic risk had, compared to patients in the bottom 30%, an OR = 73.9 (11.8-463.6, p = 4.4 × 10-6) of developing nADA. In the German cohort, the AUC of the same model was 0.83 (0.71-0.92), sensitivity = 0.80, specificity = 0.76, with an OR = 13.8 (3.0-63.3, p = 7.5 × 10-4). CONCLUSIONS We identified several HLA-associated genetic risk factors for ADA against interferon β, which were specific for treatment preparations and population backgrounds. Genetic prediction models could robustly identify patients at risk for developing ADA and might be used for personalized therapy recommendations and stratified ADA screening in clinical practice. These analyses serve as a roadmap for genetic characterizations of ADA against other biopharmaceutical compounds.
Collapse
Affiliation(s)
- Till F M Andlauer
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str 22, 81675, Munich, Germany.
- Max Planck Institute of Psychiatry, Kraepelinstr 2-10, 80804, Munich, Germany.
| | - Jenny Link
- Department of Clinical Neuroscience, Karolinska Institutet, Visionsgatan 18, 17176, Stockholm, Sweden
| | - Dorothea Martin
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str 22, 81675, Munich, Germany
| | - Malin Ryner
- Department of Clinical Neuroscience, Karolinska Institutet, Visionsgatan 18, 17176, Stockholm, Sweden
| | - Christina Hermanrud
- Department of Clinical Neuroscience, Karolinska Institutet, Visionsgatan 18, 17176, Stockholm, Sweden
| | - Verena Grummel
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str 22, 81675, Munich, Germany
| | - Michael Auer
- Department of Neurology, Medical University of Innsbruck, Anichstr 35, 6020, Innsbruck, Austria
| | - Harald Hegen
- Department of Neurology, Medical University of Innsbruck, Anichstr 35, 6020, Innsbruck, Austria
| | - Lilian Aly
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str 22, 81675, Munich, Germany
- Institute of Experimental Neuroimmunology, Technical University of Munich, Trogerstr 9, 81675, Munich, Germany
| | - Christiane Gasperi
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str 22, 81675, Munich, Germany
| | - Benjamin Knier
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str 22, 81675, Munich, Germany
- Institute of Experimental Neuroimmunology, Technical University of Munich, Trogerstr 9, 81675, Munich, Germany
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Kraepelinstr 2-10, 80804, Munich, Germany
- Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377, Munich, Germany
| | | | - Finn Sellebjerg
- DMSC, Department of Neurology, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Visionsgatan 18, 17176, Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Visionsgatan 18, 17176, Stockholm, Sweden
| | - Marc Pallardy
- Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, INSERM, Faculté de Pharmacie, rue JB Clément, 92290, Châtenay-Malabry, France
| | - Sebastian Spindeldreher
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4056, Basel, Switzerland
- Integrated Biologix GmbH, Steinenvorstadt 33, 4051, Basel, Switzerland
| | - Florian Deisenhammer
- Department of Neurology, Medical University of Innsbruck, Anichstr 35, 6020, Innsbruck, Austria
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Visionsgatan 18, 17176, Stockholm, Sweden
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str 22, 81675, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377, Munich, Germany.
| |
Collapse
|
5
|
Stickler M, Reddy A, Xiong JM, Wong MH, Akamatsu Y, Hinton PR, Harding FA. Design, creation and in vitro testing of a reduced immunogenicity humanized anti-CD25 monoclonal antibody that retains functional activity. Protein Eng Des Sel 2019; 32:543-554. [PMID: 32725169 DOI: 10.1093/protein/gzaa017] [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: 04/04/2020] [Revised: 06/29/2020] [Accepted: 07/06/2020] [Indexed: 11/14/2022] Open
Abstract
Humanized and fully human sequence-derived therapeutic antibodies retain the capacity to induce anti-drug antibodies. Daclizumab (humanized version of the murine anti-Tac antibody; E.HAT) was selected for a proof of concept application of engineering approaches to reduce potential immunogenicity due to its demonstrated immunogenicity in the clinic. Reduced immunogenicity variants of E.HAT were created by identifying and modifying a CD4+ T cell epitope region in the VH region. Variant epitope region peptides were selected for their reduced capacity to induce CD4+ T cell proliferative responses in vitro. Variant antibody molecules were created, and CD25 affinity and potency were similar to the unmodified parent antibody. Fab fragments from the variant antibodies induced a lower frequency and magnitude of responses in human peripheral blood mononuclear cells proliferation tests. By the empirical selection of two amino acid mutations, fully functional humanized E.HAT antibodies with reduced potential to induce immune responses in vitro were created.
Collapse
Affiliation(s)
| | | | | | | | | | - Paul R Hinton
- Formerly of AbbVie, Redwood City, CA, USA.,IGM Biosciences, Mountain View, CA, USA
| | | |
Collapse
|
6
|
Schultz HS, Reedtz-Runge SL, Bäckström BT, Lamberth K, Pedersen CR, Kvarnhammar AM. Quantitative analysis of the CD4+ T cell response to therapeutic antibodies in healthy donors using a novel T cell:PBMC assay. PLoS One 2017; 12:e0178544. [PMID: 28562666 PMCID: PMC5451071 DOI: 10.1371/journal.pone.0178544] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 05/15/2017] [Indexed: 12/22/2022] Open
Abstract
Many biopharmaceuticals (BPs) are known to be immunogenic in the clinic, which can result in modified pharmacokinetics, reduced efficacy, allergic reactions and anaphylaxis. During recent years, several technologies to predict immunogenicity have been introduced, but the predictive value is still considered low. Thus, there is an unmet medical need for optimization of such technologies. The generation of T cell dependent high affinity anti-drug antibodies plays a key role in clinical immunogenicity. This study aimed at developing and evaluating a novel in vitro T cell:PBMC assay for prediction of the immunogenicity potential of BPs. To this end, we assessed the ability of infliximab (anti-TNF-α), rituximab (anti-CD20), adalimumab (anti-TNF-α) and natalizumab (anti-α4-integrin), all showing immunogenicity in the clinic, to induce a CD4+ T cells response. Keyhole limpet hemocyanin (KLH) and cytomegalovirus pp65 protein (CMV) were included as neo-antigen and recall antigen positive controls, respectively. By analyzing 26 healthy donors having HLA-DRB1 alleles matching the European population, we calculated the frequency of responding donors, the magnitude of the response, and the frequency of BP-specific T cells, as measured by 3[H]-thymidine incorporation and ELISpot IL-2 secretion. KLH and CMV demonstrated a strong T cell response in all the donors analyzed. The frequency of responding donors to the BPs was 4% for infliximab, 8% for adalimumab, 19% for rituximab and 27% for natalizumab, which is compared to and discussed with their respective observed clinical immunogenicity. This study further complements predictive immunogenicity testing by quantifying the in vitro CD4+ T cell responses to different BPs. Even though the data generated using this modified method does not directly translate to the clinical situation, a high sensitivity and immunogenic potential of most BPs is demonstrated.
Collapse
Affiliation(s)
- Heidi S. Schultz
- Immunogenicity Prediction and Tolerance, Global Research, Novo Nordisk A/S, Måløv, Denmark
- * E-mail:
| | | | - B. Thomas Bäckström
- Immunogenicity Prediction and Tolerance, Global Research, Novo Nordisk A/S, Måløv, Denmark
| | - Kasper Lamberth
- Immunogenicity Prediction and Tolerance, Global Research, Novo Nordisk A/S, Måløv, Denmark
| | | | - Anne M. Kvarnhammar
- Immunogenicity Prediction and Tolerance, Global Research, Novo Nordisk A/S, Måløv, Denmark
| | | |
Collapse
|
7
|
Barbosa MD, Smith DD. Channeling postmarketing patient data into pharmaceutical regulatory systems. Drug Discov Today 2014; 19:1897-912. [DOI: 10.1016/j.drudis.2014.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/24/2014] [Accepted: 07/24/2014] [Indexed: 12/15/2022]
|
8
|
Baker MP, Reynolds HM, Lumicisi B, Bryson CJ. Immunogenicity of protein therapeutics: The key causes, consequences and challenges. SELF NONSELF 2014; 1:314-322. [PMID: 21487506 DOI: 10.4161/self.1.4.13904] [Citation(s) in RCA: 265] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 09/20/2010] [Accepted: 10/07/2010] [Indexed: 12/15/2022]
Abstract
The immunogenicity of protein therapeutics has so far proven to be difficult to predict in patients, with many biologics inducing undesirable immune responses directed towards the therapeutic resulting in reduced efficacy, anaphylaxis and occasionally life threatening autoimmunity. The most common effect of administrating an immunogenic protein therapeutic is the development of a high affinity anti-therapeutic antibody response. Furthermore, it is clear from clinical studies that protein therapeutics derived from endogenous human proteins are capable of stimulating undesirable immune responses in patients, and as a consequence, the prediction and reduction of immunogenicity has been the focus of intense research. This review will outline the principle causes of the immunogenicity in protein therapeutics, and describe the development of pre-clinical models that can be used to aid in the prediction of the immunogenic potential of novel protein therapeutics prior to administration in man.
Collapse
Affiliation(s)
- Matthew P Baker
- Antitope Ltd.; Babraham Research Campus; Babraham, Cambridge UK
| | | | | | | |
Collapse
|
9
|
Link J, Lundkvist Ryner M, Fink K, Hermanrud C, Lima I, Brynedal B, Kockum I, Hillert J, Fogdell-Hahn A. Human leukocyte antigen genes and interferon beta preparations influence risk of developing neutralizing anti-drug antibodies in multiple sclerosis. PLoS One 2014; 9:e90479. [PMID: 24608124 PMCID: PMC3946519 DOI: 10.1371/journal.pone.0090479] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 02/01/2014] [Indexed: 11/18/2022] Open
Abstract
A significant proportion of patients with multiple sclerosis who receive interferon beta (IFNβ) therapy develop neutralizing antibodies (NAbs) that reduce drug efficacy. To investigate if HLA class I and II alleles are associated with development of NAbs against IFNβ we analyzed whether NAb status and development of NAb titers high enough to be biologically relevant (>150 tenfold reduction units/ml) correlated with the HLA allele group carriage in a cohort of 903 Swedish patients with multiple sclerosis treated with either intramuscular IFNβ-1a, subcutaneous IFNβ-1a or subcutaneous IFNβ-1b. Carriage of HLA-DRB1*15 was associated with increased risk of developing NAbs and high NAb titers. After stratification based on type of IFNβ preparation, HLA-DRB1*15 carriage was observed to increase the risk of developing NAbs as well as high NAb titers against both subcutaneous and intramuscular IFNβ-1a. Furthermore, in patients receiving subcutaneous IFNβ-1a carriage of HLA-DQA1*05 decreased the risk for high NAb titers. In IFNβ-1b treated patients, HLA-DRB1*04 increased the risk of developing high NAb titers, and in a subgroup analysis of DRB1*04 alleles the risk for NAbs was increased in DRB1*04:01 carriers. In conclusion, there is a preparation-specific genetically determined risk to develop NAbs against IFNβ high enough to be clinically relevant in treatment decisions for patients with multiple sclerosis if confirmed in future studies. However, choice of IFNβ preparation still remains the single most significant determinant for the risk of developing NAbs.
Collapse
Affiliation(s)
- Jenny Link
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Malin Lundkvist Ryner
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Katharina Fink
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Christina Hermanrud
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Izaura Lima
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Boel Brynedal
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Chester KA, Baker M, Mayer A. Overcoming the immunologic response to foreign enzymes in cancer therapy. Expert Rev Clin Immunol 2014; 1:549-59. [DOI: 10.1586/1744666x.1.4.549] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
11
|
Jawa V, Cousens LP, Awwad M, Wakshull E, Kropshofer H, De Groot AS. T-cell dependent immunogenicity of protein therapeutics: Preclinical assessment and mitigation. Clin Immunol 2013; 149:534-55. [PMID: 24263283 DOI: 10.1016/j.clim.2013.09.006] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/13/2013] [Accepted: 09/14/2013] [Indexed: 02/07/2023]
Abstract
Protein therapeutics hold a prominent and rapidly expanding place among medicinal products. Purified blood products, recombinant cytokines, growth factors, enzyme replacement factors, monoclonal antibodies, fusion proteins, and chimeric fusion proteins are all examples of therapeutic proteins that have been developed in the past few decades and approved for use in the treatment of human disease. Despite early belief that the fully human nature of these proteins would represent a significant advantage, adverse effects associated with immune responses to some biologic therapies have become a topic of some concern. As a result, drug developers are devising strategies to assess immune responses to protein therapeutics during both the preclinical and the clinical phases of development. While there are many factors that contribute to protein immunogenicity, T cell- (thymus-) dependent (Td) responses appear to play a critical role in the development of antibody responses to biologic therapeutics. A range of methodologies to predict and measure Td immune responses to protein drugs has been developed. This review will focus on the Td contribution to immunogenicity, summarizing current approaches for the prediction and measurement of T cell-dependent immune responses to protein biologics, discussing the advantages and limitations of these technologies, and suggesting a practical approach for assessing and mitigating Td immunogenicity.
Collapse
|
12
|
Immunogenicity to biologics: mechanisms, prediction and reduction. Arch Immunol Ther Exp (Warsz) 2012; 60:331-44. [PMID: 22930363 DOI: 10.1007/s00005-012-0189-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 05/11/2012] [Indexed: 01/06/2023]
Abstract
Currently, there is a significant rise in the development and clinical use of a unique class of pharmaceuticals termed as Biopharmaceuticals or Biologics, in the management of a range of disease conditions with, remarkable therapeutic benefits. However, there is an equally growing concern regarding development of adverse effects like immunogenicity in the form of anti-drug antibodies (ADA) production and hypersensitivity. Immunogenicity to biologics represents a significant hurdle in the continuing therapy of patients in a number of disease settings. Efforts focussed on the identification of factors that contribute towards the onset of immunogenic response to biologics have led to reductions in the incidence of immunogenicity. An in-depth understanding of the cellular and molecular mechanism underpinning immunogenic responses will likely improve the safety profile of biologics. This review addresses the mechanistic basis of ADA generation to biologics, with emphasis on the role of antigen processing and presentation in this process. The article also addresses the potential contribution of complement system in augmenting or modulating this response. Identifying specific factors that influences processing and presentation of biologic-derived antigens in different genotype and disease background may offer additional options for intervention in the immunogenic process and consequently, the management of immunogenicity to biologics.
Collapse
|
13
|
Biosimilars and biobetters as tools for understanding and mitigating the immunogenicity of biotherapeutics. Drug Discov Today 2012; 17:1282-8. [PMID: 22796124 DOI: 10.1016/j.drudis.2012.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 06/11/2012] [Accepted: 07/04/2012] [Indexed: 11/21/2022]
Abstract
In this article, we review key steps for the development of biosimilars and biobetters and related bioanalytical challenges, with a focus on how they are associated with immunogenicity. We analyze the factors that can impact antidrug antibody (ADA) responses and their correlations with preclinical and clinical outcomes to provide relevant insights and to answer questions, including what types of aggregate are immunogenic. We also address strategies for developing less-immunogenic biotherapeutics. Using interferon-β (IFN-β) as a case study, we explore the correlation between aggregation and immunogenicity. We dissect and integrate with clinical data the IFN-β preclinical immunogenicity and aggregation predictions and discuss the feasibility of developing an IFN-β with lower aggregation and/or immunogenicity.
Collapse
|
14
|
Barbosa MD. Immunogenicity of biotherapeutics in the context of developing biosimilars and biobetters. Drug Discov Today 2011; 16:345-53. [DOI: 10.1016/j.drudis.2011.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 01/07/2011] [Accepted: 01/31/2011] [Indexed: 11/24/2022]
|
15
|
Enevold C, Oturai AB, Sørensen PS, Ryder LP, Koch-Henriksen N, Bendtzen K. Polymorphisms of innate pattern recognition receptors, response to interferon-beta and development of neutralizing antibodies in multiple sclerosis patients. Mult Scler 2010; 16:942-9. [PMID: 20595247 DOI: 10.1177/1352458510373264] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Interferon-beta therapy of patients with relapsing-remitting multiple sclerosis involves repeated 'immunizations' with exogenous protein solutions. Innate pattern recognition receptors play an important role in immune responses towards foreign substances and may thus be related to treatment outcome. OBJECTIVE To determine the genotypes at 42 single nucleotide polymorphism loci in selected pattern recognition receptors for 567 prospectively followed relapsing-remitting multiple sclerosis patients treated with recombinant interferon-beta, and test for relationships to several outcome parameters, including formation of interferon-beta neutralizing antibodies. RESULTS The results suggest an association between the rs5743810 polymorphism (Ser249Pro) of TLR6 and development of neutralizing antibodies after 24 months of therapy in males (p = 0.00002), but not in females (p = 0.2). This association survived crude Bonferroni correction (p (corrected) = 0.02). Additional associations were observed in carriers of the TLR2-rs5743708 and NOD2-rs3135499 SNPs (time to relapse), the TLR7-rs179008 and NOD1-rs2075820 SNPs (time to disease progression) and the TLR4-rs7873784, TLR9-rs5743836, and NOD2-rs2066842 SNPs (frequency of neutralizing antibodies development). All of these, however, failed to survive correction for multiple testing. There were no significant differences between interferon-beta responders and non-responders for any of the investigated single nucleotide polymorphisms. CONCLUSIONS The rs5743810 polymorphism of TLR6 may be involved in development of anti-interferon-beta antibodies in males, although further studies are required to firmly establish this.
Collapse
Affiliation(s)
- Christian Enevold
- Institute for Inflammation Research, Department of Rheumatology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
16
|
Harding FA, Stickler MM, Razo J, DuBridge RB. The immunogenicity of humanized and fully human antibodies: residual immunogenicity resides in the CDR regions. MAbs 2010; 2:256-65. [PMID: 20400861 DOI: 10.4161/mabs.2.3.11641] [Citation(s) in RCA: 513] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Monoclonal antibodies represent an attractive therapeutic tool as they are highly specific for their targets, convey effector functions and enjoy robust manufacturing procedures. Humanization of murine monoclonal antibodies has vastly improved their in vivo tolerability. Humanization, the replacement of mouse constant regions and V framework regions for human sequences, results in a significantly less immunogenic product. However, some humanized and even fully human sequence-derived antibody molecules still carry immunological risk. To more fully understand the immunologic potential of humanized and human antibodies, we analyzed CD4(+) helper T cell epitopes in a set of eight humanized antibodies. The antibodies studied represented a number of different VH and VL family members carrying unique CDR regions. In spite of these differences, CD4(+) T cell epitopes were found only in CDR-sequence containing regions. We were able to incorporate up to two amino acid modifications in a single epitope that reduced the immunogenic potential while retaining full biologic function. We propose that immunogenicity will always be present in some antibody molecules due to the nature of the antigen-specific combining sites. A consequence of this result is modifications to reduce immunogenicity will be centered on the affinity-determining regions. Modifications to CDR regions can be designed that reduce the immunogenic potential while maintaining the bioactivity of the antibody molecule.
Collapse
|
17
|
Immunogenicity of biologically-derived therapeutics: assessment and interpretation of nonclinical safety studies. Regul Toxicol Pharmacol 2009; 54:164-82. [PMID: 19345250 DOI: 10.1016/j.yrtph.2009.03.012] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 03/24/2009] [Accepted: 03/25/2009] [Indexed: 11/20/2022]
Abstract
An evaluation of potential antibody formation to biologic therapeutics during the course of nonclinical safety studies and its impact on the toxicity profile is expected under current regulatory guidance and is accepted standard practice. However, approaches for incorporating this information in the interpretation of nonclinical safety studies are not clearly established. Described here are the immunological basis of anti-drug antibody formation to biopharmaceuticals (immunogenicity) in laboratory animals, and approaches for generating and interpreting immunogenicity data from nonclinical safety studies of biotechnology-derived therapeutics to support their progression to clinical evaluation. We subscribe that immunogenicity testing strategies should be adapted to the specific needs of each therapeutic development program, and data generated from such analyses should be integrated with available clinical and anatomic pathology, pharmacokinetic, and pharmacodynamic data to properly interpret nonclinical studies.
Collapse
|
18
|
Perry LCA, Jones TD, Baker MP. New approaches to prediction of immune responses to therapeutic proteins during preclinical development. Drugs R D 2009; 9:385-96. [PMID: 18989990 DOI: 10.2165/0126839-200809060-00004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Clinical studies show that immunogenicity observed against therapeutic proteins can limit efficacy and reduce the safety of the treatment. It is therefore beneficial to be able to predict the immunogenicity of therapeutic proteins before they enter the clinic. Studies using deimmunized proteins have highlighted the importance of T-cell epitopes in the generation of undesirable immunogenicity. In silico, in vitro, ex vivo and in vivo methods have therefore been developed that focus on identification of CD4+ T-cell epitopes in the sequence of therapeutic proteins. A case study of existing therapeutic proteins is presented to review these different approaches in order to assess their utility in predicting immunogenic potential.
Collapse
Affiliation(s)
- Laura C A Perry
- Antitope Ltd, Babraham Research Campus, Babraham, Cambridge, UK
| | | | | |
Collapse
|
19
|
Garg N, Weinstock-Guttman B, Bhasi K, Locke J, Ramanathan M. An association between autoreactive antibodies and anti-interferon-beta antibodies in multiple sclerosis. Mult Scler 2007; 13:895-9. [PMID: 17468449 DOI: 10.1177/1352458507076968] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Approximately 5-25% of interferon-beta (IFN-beta) treated multiple sclerosis (MS) patients develop anti-IFN-beta neutralizing antibodies (NAb) but the patient-specific variables associated with the risk of developing anti-IFN-beta antibodies are poorly understood. Anti-IFN-beta NAb are a subset of anti-IFN-beta binding antibodies (BAb) and all patients with NAb generally have high levels of associated BAb. The purpose of this research was to assess the association between autoreactive antibodies (ARAB) and the risk of developing anti-IFN-beta BAb in MS patients. This was a retrospective study that included consecutive patients diagnosed with clinically definite MS evaluated at our center and considered appropriate for IFN-beta therapy. The patients were tested for various subtypes of antiphospholipid antibodies (APLA) including anti-phosphatidylethanolamine (APE), anti-phosphatidylserine (APS), and anti-cardiolipin (ACA) antibodies, and other ARAB, antinuclear and anti-neutrophilic cytoplasmic antibodies, anti-thyroid peroxidase antibodies (ATA), anti-SS-A and anti-SS-B antibodies. BAb levels were assessed using a commercial binding ELISA assay. A total of 33 patients (mean age: 45.4 years, 85% female) were enrolled; 15 patients were negative and 18 patients were positive for BAb. APLA or ATA were present in 95% (17 of 18 patients) of patients positive for BAb. In comparison, APLA or ATA occurred in only 27% (four of 15 patients) of patients in the BAb negative group. The associations between the occurrence of BAb and the occurrence of high APLA or ATA were significant (chi2 = 13.4, P<0.001in Fisher exact test). The odds ratio for the association was 46.8 (with a 95% confidence interval range of 4.6-475). No significant correlations were found for other ARAB. The presence of autoreactive antibodies, particularly APLA and ATA is associated with increased risk of occurrence of IFN-beta BAb in MS patients on long-term IFN-beta therapy.
Collapse
Affiliation(s)
- N Garg
- Jacobs Neurological Institute, Buffalo General Hospital, Buffalo, NY, USA.
| | | | | | | | | |
Collapse
|
20
|
Barbosa MDFS, Vielmetter J, Chu S, Smith DD, Jacinto J. Clinical link between MHC class II haplotype and interferon-beta (IFN-beta) immunogenicity. Clin Immunol 2005; 118:42-50. [PMID: 16260183 DOI: 10.1016/j.clim.2005.08.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Revised: 08/25/2005] [Accepted: 08/31/2005] [Indexed: 02/06/2023]
Abstract
Interferon-beta (IFN-beta) is currently the first-line therapy for the treatment of multiple sclerosis (MS). However, a significant percentage of MS patients develop anti-IFN-beta antibodies, which can reduce the efficacy of the drug. We describe an association between a common MHC class II allele (DRB1*0701), present in 23% of the patients studied, and the anti-IFN-beta antibody response. We identified IFN-beta epitopes using a peptide-binding assay with B cell lines expressing this allele. Moreover, epitope-specific activation responses obtained with peripheral blood mononuclear cells (PBMCs) from IFN-beta treated patients with the DRB1*0701 allele indicated a role for T-cell activation in IFN-beta immunogenicity. These results suggest that HLA typing of MS patients may provide an accurate screen for subjects who are likely to develop anti-IFN-beta antibodies and should therefore be considered for alternative therapies. In addition, elucidation of the factors underlying the anti-IFN-beta antibody response should accelerate the engineering of less immunogenic IFN-beta therapeutics.
Collapse
|
21
|
Yuasa T, Tsuji H, Kimura S, Niwa N, Yurugi K, Egawa H, Tanaka K, Maruya E, Saji HO, Asano H, Maekawa T. Human leukocyte antigens in Japanese patients with biliary atresia: Retrospective analysis of patients who underwent living donor liver transplantation. Hum Immunol 2005; 66:295-300. [PMID: 15784468 DOI: 10.1016/j.humimm.2004.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2004] [Revised: 11/10/2004] [Accepted: 11/16/2004] [Indexed: 11/23/2022]
Abstract
Biliary atresia (BA) is a neonatal obstructive cholangiopathy characterized by a fibrosclerosing obliteration of the extrahepatic bile duct. The aim of this study was to investigate the relationship between human leukocyte antigens (HLA) and susceptibility to BA. We retrospectively analyzed 392 Japanese patients with BA and without extrahepatic anomalies who underwent living donor liver transplantations at our institute. Healthy Japanese volunteers (n = 828) served as normal controls. A significant positive association was observed between BA and HLA-DR2 (39.0% of patients vs. 30.4% of controls, odds ratio = 1.46, p = 0.029). Two-locus analyses disclosed that DR2 was not independently associated with BA, but the increased frequency of HLA-A24 and -B52 reflected the linkage disequilibrium between -A24, -B52, and -DR2. Moreover, the frequency of the haplotype HLA-A24-B52-DR2 was significantly higher in patients with BA than in the general Japanese populations described in the literature (odds ratio = 2.20, p = 0.00124). These results indicate that the gene for BA susceptibility is in close linkage disequilibrium with the HLA-A24-B52-DR2 haplotype observed in the Japanese population. We speculate that a gene at the locus close to HLA plays an important role in the pathogenesis of BA.
Collapse
Affiliation(s)
- Takeshi Yuasa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yeung VP, Chang J, Miller J, Barnett C, Stickler M, Harding FA. Elimination of an immunodominant CD4+ T cell epitope in human IFN-beta does not result in an in vivo response directed at the subdominant epitope. THE JOURNAL OF IMMUNOLOGY 2004; 172:6658-65. [PMID: 15153481 DOI: 10.4049/jimmunol.172.11.6658] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The BALB/cByJ mouse strain displays an immunodominant T cell response directed at the same CD4(+) T cell epitope peptide region in human IFN-beta, as detected in a human population-based assay. BALB/cByJ mice also recognize a second region of the protein with a lesser magnitude proliferative response. Critical residue testing of the immunodominant peptide showed that both BALB/cByJ mice and the human population response were dependent on an isoleucine residue at position 129. A variant IFN-beta molecule was constructed containing the single amino acid modification, I129V, in the immunodominant epitope. The variant displayed 100% of control antiproliferation activity. Mice immunized with unmodified IFN-beta responded weakly in vitro to the I129V variant. However, BALB/cByJ mice immunized with the I129V variant were unable to respond to either the I129V variant or the unmodified IFN-beta molecule by either T cell proliferation or Ag-specific IgG1 Ab production. This demonstrates that a single amino acid change in an immunodominant epitope can eliminate an immune response to an otherwise intact therapeutic protein. The elimination of the immunodominant epitope response also eliminated the response to the subdominant epitope in the protein. Modifying functionally immunodominant T cell epitopes within proteins may obviate the need for additional subdominant epitope modifications.
Collapse
|