1
|
Curvino EJ, Roe EF, Freire Haddad H, Anderson AR, Woodruff ME, Votaw NL, Segura T, Hale LP, Collier JH. Engaging natural antibody responses for the treatment of inflammatory bowel disease via phosphorylcholine-presenting nanofibres. Nat Biomed Eng 2024; 8:628-649. [PMID: 38012308 PMCID: PMC11128482 DOI: 10.1038/s41551-023-01139-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/17/2023] [Indexed: 11/29/2023]
Abstract
Inflammatory bowel disease lacks a long-lasting and broadly effective therapy. Here, by taking advantage of the anti-infection and anti-inflammatory properties of natural antibodies against the small-molecule epitope phosphorylcholine (PC), we show in multiple mouse models of colitis that immunization of the animals with self-assembling supramolecular peptide nanofibres bearing PC epitopes induced sustained levels of anti-PC antibodies that were both protective and therapeutic. The strength and type of immune responses elicited by the nanofibres could be controlled through the relative valency of PC epitopes and exogenous T-cell epitopes on the nanofibres and via the addition of the adjuvant CpG. The nanomaterial-assisted induction of the production of therapeutic antibodies may represent a durable therapy for inflammatory bowel disease.
Collapse
Affiliation(s)
| | - Emily F Roe
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Alexa R Anderson
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Mia E Woodruff
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nicole L Votaw
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Laura P Hale
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
2
|
Liu Y, Liao F. Vaccination therapy for inflammatory bowel disease. Hum Vaccin Immunother 2023; 19:2259418. [PMID: 37771317 PMCID: PMC10543345 DOI: 10.1080/21645515.2023.2259418] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
Recently, several novel medications, such as Ustekinumab, Infliximab, and Vedolizumab, have emerged as potential options for inflammatory bowel disease(IBD) management. Despite achieving some effects in clinical applications, these therapies are still plagued by inadequate response rates and adverse side effects. With rapid progress in immunological research, therapeutic vaccines are gaining traction as an alternative. These vaccines aim to activate the body's immune system to generate specific antibodies, thereby offering a potential avenue for treating IBD. The efficacy and safety of vaccines, coupled with their potential to mitigate the financial and healthcare burden associated with disease treatment, render therapeutic vaccines a more favorable approach for managing patients with IBD. In this review, we critically examine the existing literature pertaining to therapeutic vaccines for IBD, aiming to offer researchers a comprehensive understanding of their applications and prospects and stimulate novel vaccine development by presenting innovative ideas in this field.
Collapse
Affiliation(s)
- Yafei Liu
- Department of Gastroenterology, Wuhan University, Wuhan, Hubei, China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
3
|
Iversen PL, Kipshidze N, Kipshidze N, Dangas G, Ramacciotti E, Kakabadze Z, Fareed J. A novel therapeutic vaccine targeting the soluble TNFα receptor II to limit the progression of cardiovascular disease: AtheroVax™. Front Cardiovasc Med 2023; 10:1206541. [PMID: 37534280 PMCID: PMC10392828 DOI: 10.3389/fcvm.2023.1206541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/03/2023] [Indexed: 08/04/2023] Open
Abstract
The burden of atherosclerotic cardiovascular disease contributes to a large proportion of morbidity and mortality, globally. Vaccination against atherosclerosis has been proposed for over 20 years targeting different mediators of atherothrombosis; however, these have not been adequately evaluated in human clinical trials to assess safety and efficacy. Inflammation is a driver of atherosclerosis, but inflammatory mediators are essential components of the immune response. Only pathogenic forms of sTNFR2 are acted upon while preserving the membrane-bound (wild-type) TNFR2 contributions to a non-pathogenic immune response. We hypothesize that the inhibition of sTNRF2 will be more specific and offer long-term treatment options. Here we describe pre-clinical findings of an sTNFR2-targeting peptide vaccine (AtheroVax™) in a mouse model. The multiple pathways to synthesis of the soluble TNFRII receptor (sTNFRII) were identified as sTNFRII(PC), sTNFRII(Δ7), and sTNFRII(Δ7,9). The sTNFRII(Δ7) peptide, NH2-DFALPVEKPLCLQR-COOH is specific to sTNFR2 based on an mRNA splice-variant in which exon 6 is joined to exon 8. The role of sTNFRII(Δ7) as a mediator of prolonged TNFα activity by preventing degradation and clearance was investigated. Inflammation is a critical driver of onset, progression and expansion of atherosclerosis. The TNFα ligand represents a driver of inflammation that is mediated by a splice variant of TNFR2, referred to as sTNFRII(Δ7). The multiple forms of TNFRII, both membrane bound and soluble, are associated with distinctly different phenotypes. sTNFRII(PC) and sTNFRII(Δ7) are not equivalent to etanercept because they lack a clearance mechanism. The unique peptide associated with sTNFRII(Δ7) contains a linear B-cell epitope with amino acids from both exon 6 and exon 8 supporting the vaccine design. Animal studies to evaluate the vaccine are ongoing, and results will be forthcoming. We describe a peptide vaccine targeting sTNFR2 in limiting the progression of atherosclerosis. A therapeutic vaccine limiting the progression of atherosclerosis will greatly contribute to the reduction in morbidity and mortality from cardiovascular disease. It is likely the vaccine will be used in combination with the current standards of care and lifestyle modifications.
Collapse
Affiliation(s)
- Patrick L. Iversen
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | | | - Nodar Kipshidze
- Mailman School of Public Health, Columbia University, New York, NY, United States
| | - George Dangas
- Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Zurab Kakabadze
- Head Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - Jawed Fareed
- Department of Molecular Pharmacology and Neuroscience, Loyola University Medical Center, Maywood, IL, United States
| |
Collapse
|
4
|
Hernandez A, Hartgerink JD, Young S. Self-assembling peptides as immunomodulatory biomaterials. Front Bioeng Biotechnol 2023; 11:1139782. [PMID: 36937769 PMCID: PMC10014862 DOI: 10.3389/fbioe.2023.1139782] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
Self-assembling peptides are a type of biomaterial rapidly emerging in the fields of biomedicine and material sciences due to their promise in biocompatibility and effectiveness at controlled release. These self-assembling peptides can form diverse nanostructures in response to molecular interactions, making them versatile materials. Once assembled, the peptides can mimic biological functions and provide a combinatorial delivery of therapeutics such as cytokines and drugs. These self-assembling peptides are showing success in biomedical settings yet face unique challenges that must be addressed to be widely applied in the clinic. Herein, we describe self-assembling peptides' characteristics and current applications in immunomodulatory therapeutics.
Collapse
Affiliation(s)
- Andrea Hernandez
- Katz Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, United States
| | - Jeffrey D. Hartgerink
- Department of Chemistry and Department of Bioengineering, Rice University, Houston, TX, United States
| | - Simon Young
- Katz Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, United States
- *Correspondence: Simon Young,
| |
Collapse
|
5
|
From vaccines to nanovaccines: A promising strategy to revolutionize rheumatoid arthritis treatment. J Control Release 2022; 350:107-121. [PMID: 35977582 DOI: 10.1016/j.jconrel.2022.08.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Rheumatoid arthritis (RA) is a joint-related autoimmune disease that is difficult to cure. Most therapeutics act to alleviate the symptoms but not correct the causes of RA. Novel strategies that specifically target the causes are highly needed for RA management. Currently, early interruption of RA is increasingly suggested but the corresponding therapeutics are not available. Vaccines that have shown great success to combat infection, cancer, degenerative diseases, autoimmune diseases, etc. are ideal candidates for a new generation of anti-RA therapeutics to correct the causes and prevent RA or interrupt RA in early phases. Anti-RA vaccines can be divided into two major categories. One is to induce neutralizing antibodies and the other is to induce antigen-specific immune tolerance. The vaccines are inherently linked to nanotechnology because they usually need a biomacromolecule or carrier to provoke sufficient immune responses. In the past decade, designed nanocarriers such as nanoparticles, liposomes, nanoemulsion, etc., have been applied to optimize the vaccines for autoimmune disease treatment. Nanotechnology endows vaccines with a higher biostability, tunable in vivo behavior, better targeting, co-delivery with stimulatory agents, regulatory effects on immune responses, etc. In this review, unmet medical needs for RA treatment and anti-RA vaccinology are first introduced. The development of anti-RA therapies from vaccines to nanovaccines are then reviewed and perspectives on how nanotechnology promotes vaccine development and advancement are finally provided. In addition, challenges for anti-RA vaccine development are summarized and advantages of nanovaccines are analyzed. In conclusion, nanovaccines will be a promising strategy to revolutionize the treatment of RA by correcting the causes in an early phase of RA.
Collapse
|
6
|
Rodríguez-Álvarez Y, Batista-Roche LG, Llopiz-Arzuaga A, Puente-Pérez P, Martínez-Castillo R, Castro-Velazco J, Santos-Savio A. Immunogenicity profile in African green monkeys of a vaccine candidate based on a mutated form of human Interleukin-15. BMC Immunol 2021; 22:79. [PMID: 34922462 PMCID: PMC8684083 DOI: 10.1186/s12865-021-00470-4] [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: 04/07/2021] [Accepted: 12/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Interleukin (IL)-15 is a proinflammatory T-cell growth factor overexpressed in several autoimmune diseases such as rheumatoid arthritis. Our initial strategy to neutralize the increased levels of IL-15 consisted in a vaccine candidate based on the recombinant modified human IL-15 (mhIL-15) mixed with the alum adjuvant. A previous study in non-human primates Macaca fascicularis has shown that vaccination induces neutralizing antibodies against native IL-15, without affecting animal behavior, clinical status, or the percentage of IL-15-dependent cell populations. However, the mhIL-15 used as an antigen was active in the IL-2-dependent cytotoxic T-cell line CTLL-2, which could hinder its therapeutic application. The current article evaluated the immunogenicity in African green monkeys of a vaccine candidate based on IL-15 mutant D8SQ108S, an inactive form of human IL-15. RESULTS IL-15 D8SQ108S was inactive in the CTLL-2 bioassay but was able to competitively inhibit the biological activity of human IL-15. Immunization with 200 µg of IL-15 mutant combined with alum elicited anti-IL-15 IgG antibodies after the second and third immunizations. The median values of anti-IL-15 antibody titers were slightly higher than those generated in animals immunized with 200 µg of mhIL-15. The highest antibody titers were induced after the third immunization in monkeys vaccinated with 350 µg of IL-15 D8SQ108S. In addition, sera from immunized animals inhibited the biological activity of human IL-15 in CTLL-2 cells. The maximum neutralizing effect was observed after the third immunization in sera of monkeys vaccinated with the highest dose of the IL-15 mutant. These sera also inhibited the proliferative activity of simian IL-15 in the CTLL-2 bioassay and did not affect the IL-2-induced proliferation of the aforementioned T-cell line. Finally, it was observed that vaccination neither affects the animal behavior nor the general clinical parameters of immunized monkeys. CONCLUSION Immunization with inactive IL-15 D8SQ108S mixed with alum generated neutralizing antibodies specific for human IL-15 in African green monkeys. Based on this fact, the current vaccine candidate could be more effective than the one based on biologically active mhIL-15 for treating autoimmune disorders involving an uncontrolled overproduction of IL-15.
Collapse
Affiliation(s)
- Yunier Rodríguez-Álvarez
- Pharmaceutical Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, 10 600, Havana, Cuba.
| | - Lino Gerardo Batista-Roche
- Pharmaceutical Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, 10 600, Havana, Cuba
| | - Alexey Llopiz-Arzuaga
- Chemistry and Physics Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, 10 600, Havana, Cuba
| | - Pedro Puente-Pérez
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, 10 600, Havana, Cuba
| | - Rafael Martínez-Castillo
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, 10 600, Havana, Cuba
| | - Jorge Castro-Velazco
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, 10 600, Havana, Cuba
| | - Alicia Santos-Savio
- Pharmaceutical Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, 10 600, Havana, Cuba
| |
Collapse
|
7
|
Mitkin NA, Ustiugova AS, Uvarova AN, Rumyantsev KA, Korneev KV, Pavshintsev VV. Serum of Mice Immunized with MT1-MMP Metalloproteinase Reduces Migration Potential of Pancreatic Cancer Cells. Mol Biol 2021. [DOI: 10.1134/s0026893321050095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
8
|
Alizadeh AA, Morris MB, Church WB, Yaqoubi S, Dastmalchi S. A mechanistic perspective, clinical applications, and phage-display-assisted discovery of TNFα inhibitors. Drug Discov Today 2021; 27:503-518. [PMID: 34628042 DOI: 10.1016/j.drudis.2021.09.024] [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: 04/03/2021] [Revised: 06/20/2021] [Accepted: 09/30/2021] [Indexed: 11/03/2022]
Abstract
TNFα participates in a variety of physiological processes, but at supra-physiological concentrations it has been implicated in the pathology of inflammatory and autoimmune diseases. Therefore, much attention has been devoted to the development of strategies that overcome the effects of aberrant TNFα concentration. Promising strategies include drugs that destabilize the active (trimeric) form of TNFα and antagonists of TNFα receptor type I. Underpinning these strategies is the successful application of phage-display technology to identify anti-TNFα peptides and antibodies. Here, we review the development of inhibitors of the TNFα-TNF receptor system, with particular focus on the phage-display-assisted identification of molecules that interfere with this system by acting as inhibitors of TNFα or by sequestering TNFα away from its receptor.
Collapse
Affiliation(s)
- Ali Akbar Alizadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael B Morris
- Discipline of Physiology and Bosch Institute, School of Medical Sciences, University of Sydney, NSW 2006, Australia
| | - W Bret Church
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy A15, University of Sydney, Sydney, NSW 2006, Australia
| | - Shadi Yaqoubi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Near East University, PO Box 99138, Nicosia, North Cyprus, Mersin 10, Turkey.
| |
Collapse
|
9
|
Modelling the linkage between influenza infection and cardiovascular events via thrombosis. Sci Rep 2020; 10:14264. [PMID: 32868834 PMCID: PMC7458909 DOI: 10.1038/s41598-020-70753-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 07/27/2020] [Indexed: 12/31/2022] Open
Abstract
There is a heavy burden associated with influenza including all-cause hospitalization as well as severe cardiovascular and cardiorespiratory events. Influenza associated cardiac events have been linked to multiple biological pathways in a human host. To study the contribution of influenza virus infection to cardiovascular thrombotic events, we develop a dynamic model which incorporates some key elements of the host immune response, inflammatory response, and blood coagulation. We formulate these biological systems and integrate them into a cohesive modelling framework to show how blood clotting may be connected to influenza virus infection. With blood clot formation inside an artery resulting from influenza virus infection as the primary outcome of this integrated model, we demonstrate how blood clot severity may depend on circulating prothrombin levels. We also utilize our model to leverage clinical data to inform the threshold level of the inflammatory cytokine TNFα which initiates tissue factor induction and subsequent blood clotting. Our model provides a tool to explore how individual biological components contribute to blood clotting events in the presence of influenza infection, to identify individuals at risk of clotting based on their circulating prothrombin levels, and to guide the development of future vaccines to optimally interact with the immune system.
Collapse
|
10
|
Shores LS, Kelly SH, Hainline KM, Suwanpradid J, MacLeod AS, Collier JH. Multifactorial Design of a Supramolecular Peptide Anti-IL-17 Vaccine Toward the Treatment of Psoriasis. Front Immunol 2020; 11:1855. [PMID: 32973764 PMCID: PMC7461889 DOI: 10.3389/fimmu.2020.01855] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022] Open
Abstract
Current treatments for chronic immune-mediated diseases such as psoriasis, rheumatoid arthritis, or Crohn's disease commonly rely on cytokine neutralization using monoclonal antibodies; however, such approaches have drawbacks. Frequent repeated dosing can lead to the formation of anti-drug antibodies and patient compliance issues, and it is difficult to identify a single antibody that is broadly efficacious across diverse patient populations. As an alternative to monoclonal antibody therapy, anti-cytokine immunization is a potential means for long-term therapeutic control of chronic inflammatory diseases. Here we report a supramolecular peptide-based approach for raising antibodies against IL-17 and demonstrate its efficacy in a murine model of psoriasis. B-cell epitopes from IL-17 were co-assembled with the universal T-cell epitope PADRE using the Q11 self-assembling peptide nanofiber system. These materials, with or without adjuvants, raised antibody responses against IL-17. Exploiting the modularity of the system, multifactorial experimental designs were used to select formulations maximizing titer and avidity. In a mouse model of psoriasis induced by imiquimod, unadjuvanted nanofibers had therapeutic efficacy, which could be enhanced with alum adjuvant but reversed with CpG adjuvant. Measurements of antibody subclass induced by adjuvanted and unadjuvanted formulations revealed strong correlations between therapeutic efficacy and titers of IgG1 (improved efficacy) or IgG2b (worsened efficacy). These findings have important implications for the development of anti-cytokine active immunotherapies and suggest that immune phenotype is an important metric for eliciting therapeutic anti-cytokine antibody responses.
Collapse
Affiliation(s)
- Lucas S Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Sean H Kelly
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Jutamas Suwanpradid
- Department of Dermatology, Duke University School of Medicine, Durham, NC, United States
| | - Amanda S MacLeod
- Department of Dermatology, Duke University School of Medicine, Durham, NC, United States.,Department of Immunology, Duke University School of Medicine, Durham, NC, United States.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, United States.,Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
11
|
Peptide-Based Vaccination Therapy for Rheumatic Diseases. J Immunol Res 2020; 2020:8060375. [PMID: 32258176 PMCID: PMC7104265 DOI: 10.1155/2020/8060375] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/28/2020] [Indexed: 02/06/2023] Open
Abstract
Rheumatic diseases are extremely heterogeneous diseases with substantial risks of morbidity and mortality, and there is a pressing need in developing more safe and cost-effective treatment strategies. Peptide-based vaccination is a highly desirable strategy in treating noninfection diseases, such as cancer and autoimmune diseases, and has gained increasing attentions. This review is aimed at providing a brief overview of the recent advances in peptide-based vaccination therapy for rheumatic diseases. Tremendous efforts have been made to develop effective peptide-based vaccinations against rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), while studies in other rheumatic diseases are still limited. Peptide-based active vaccination against pathogenic cytokines such as TNF-α and interferon-α (IFN-α) is shown to be promising in treating RA or SLE. Moreover, peptide-based tolerogenic vaccinations also have encouraging results in treating RA or SLE. However, most studies available now have been mainly based on animal models, while evidence from clinical studies is still lacking. The translation of these advances from experimental studies into clinical therapy remains impeded by some obstacles such as species difference in immunity, disease heterogeneity, and lack of safe delivery carriers or adjuvants. Nevertheless, advances in high-throughput technology, bioinformatics, and nanotechnology may help overcome these impediments and facilitate the successful development of peptide-based vaccination therapy for rheumatic diseases.
Collapse
|
12
|
Mitkin NA, Anokhin PK, Belopolskaya MV, Frolova OY, Kushnir EA, Lovat ML, Pavshintsev VV. Active immunization against serum alcohol dehydrogenase normalizes brain dopamine metabolism disturbed during chronic alcohol consumption. Alcohol 2020; 83:17-28. [PMID: 31260795 DOI: 10.1016/j.alcohol.2019.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 06/17/2019] [Accepted: 06/25/2019] [Indexed: 10/26/2022]
Abstract
Chronic ethanol consumption in high doses is associated with constitutively elevated activity of the serum alcohol dehydrogenase I (ADH I) isoform, which demonstrates a high affinity not only for ethanol but also for a number of bioamine metabolites. Such excessive ADH activity is probably associated with disruptions in the metabolism of neurotransmitters (dopamine, serotonin, and norepinephrine) and subsequent long-term changes in the activity of their receptors. Ultimately, a stable depressive-like condition contributes to the development of patients' craving for ethanol intake, frequent disruptions during therapy, and low efficacy of treatment. We applied active immunization against ADH to investigate its efficacy in the reduction of excessive serum ADH activity and regulation of ethanol consumption by chronically ethanol-fed Wistar rats (15% ethanol, 4 months, free-choice method), and we analyzed its ability to influence the levels of bioamines in the brain. Immunization (2 injections, 2-week intervals) was performed using a combination of recombinant horse ADH isozyme as an antigen and 2% aluminum hydroxide-based adjuvant. The efficacy of immunization was demonstrated by the production of high titers of ADH-specific antibodies, which was consistent with the significantly reduced ADH activity in the serum of chronically ethanol-fed rats. On the 26th day after the first vaccine injection, we registered significantly lower levels of alcohol consumption compared to ethanol-fed control animals, and the difference reached 16% on the 49th day of the experiment. These observations were accompanied by data that showed reduced levels of ethanol preference in immunized rats. Chronic alcohol drinking led to a decrease in dopamine and DOPAL (a direct dopamine metabolite and a high-affinity ADH substrate) levels in the striatum,while immunization neutralized this effect. Additionally, we observed that inhibition of serum ADH activity caused a decrease in peak dopamine levels during acute alcohol intake in chronically ethanol-fed rats during ethanol withdrawal that was associated with reduced tyrosine hydroxylase activity in the striatum. The obtained data suggest a significant contribution of ADH to the changes in neurotransmitter systems during chronic alcohol consumption and make available new prospects for developing innovative strategies for treatment of excessive alcohol intake.
Collapse
|
13
|
Rostami-Nejad M, Yazdi MH, Nikfar S, Rezaie A, Abdollahi M. Potential Vaccines for Treating Crohn's Disease. IRANIAN BIOMEDICAL JOURNAL 2020; 24:1-14. [PMCID: PMC6900481 DOI: 10.29252/ibj.24.1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/08/2019] [Indexed: 11/28/2022]
Abstract
CD is an inflammatory disease of the GIT and can affect several parts of the digestive system. There is a relationship between impaired mucosal barrier in the GIT of IBD patients and the role of bacteria such as MAP in CD. Apart from different therapeutic approaches for treating CD, development of a vaccine is a novel modality. In the present article, most available therapeutic opportunities in the last decade, especially the possibility of vaccines against CD, are reviewed. According to the search, availability of a new generation of vaccines against CD is expected specially tolerogenic ex vivo -derived DC-based vaccines. Regarding different locations of the challenge and the variety of clinical manifests of CD and also the type of resident antigen-presenting cells and their traffic in different parts of GIT, the results of immunotherapy with DC-based vaccines may vary case by case.
Collapse
Affiliation(s)
- Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Yazdi
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Shekoufeh Nikfar
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Rezaie
- Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mohammad Abdollahi
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
A New Venue of TNF Targeting. Int J Mol Sci 2018; 19:ijms19051442. [PMID: 29751683 PMCID: PMC5983675 DOI: 10.3390/ijms19051442] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
The first Food and Drug Administration-(FDA)-approved drugs were small, chemically-manufactured and highly active molecules with possible off-target effects, followed by protein-based medicines such as antibodies. Conventional antibodies bind a specific protein and are becoming increasingly important in the therapeutic landscape. A very prominent class of biologicals are the anti-tumor necrosis factor (TNF) drugs that are applied in several inflammatory diseases that are characterized by dysregulated TNF levels. Marketing of TNF inhibitors revolutionized the treatment of diseases such as Crohn’s disease. However, these inhibitors also have undesired effects, some of them directly associated with the inherent nature of this drug class, whereas others are linked with their mechanism of action, being pan-TNF inhibition. The effects of TNF can diverge at the level of TNF format or receptor, and we discuss the consequences of this in sepsis, autoimmunity and neurodegeneration. Recently, researchers tried to design drugs with reduced side effects. These include molecules with more specificity targeting one specific TNF format or receptor, or that neutralize TNF in specific cells. Alternatively, TNF-directed biologicals without the typical antibody structure are manufactured. Here, we review the complications related to the use of conventional TNF inhibitors, together with the anti-TNF alternatives and the benefits of selective approaches in different diseases.
Collapse
|
15
|
Zhang Y, Xu S, Huang E, Zhou H, Li B, Shao C, Yang Y. MicroRNA-130a regulates chondrocyte proliferation and alleviates osteoarthritis through PTEN/PI3K/Akt signaling pathway. Int J Mol Med 2018. [PMID: 29532889 DOI: 10.3892/ijmm.2018.3551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The function of microRNA‑130a in development and progression of osteoarthritis was determined. In osteoarthritis patients, the serum levels of microRNA‑130a were decreased, compared with normal group. Overexpression of microRNA‑130a increased cell proliferation and decreased apoptosis of chondrocytes, and downregulation of microRNA‑130a also decreased cell proliferation and induced apoptosis in chondrocytes. Downregulation of microRNA‑130a promoted Bax and caspase‑3/9 protein expression, increased inflammation divisors and suppressed the PTEN/PI3K/Akt signaling pathway. PTEN inhibitor, VO‑Ohpic trihydrate increased the destructive effect of microRNA‑130a on cell proliferation of chondrocytes. PI3K inhibitor, wortmannin also increased the destructive effect of microRNA‑130a on osteoarthritis. In conclusion, microRNA‑130a is an important regulator of osteoarthritis in chondrocytes through PTEN/PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Orthopaedics, Shanghai Tongji Hospital, Shanghai 200065, P.R. China
| | - Shaochen Xu
- Department of Orthopaedics, Shanghai Tongji Hospital, Shanghai 200065, P.R. China
| | - Eric Huang
- Department of Orthopaedics, Shanghai Tongji Hospital, Shanghai 200065, P.R. China
| | - Haichao Zhou
- Department of Orthopaedics, Shanghai Tongji Hospital, Shanghai 200065, P.R. China
| | - Bing Li
- Department of Orthopaedics, Shanghai Tongji Hospital, Shanghai 200065, P.R. China
| | - Chenni Shao
- Shanghai Jiading Nanxiang Hospital, Shanghai 200065, P.R. China
| | - Yunfeng Yang
- Department of Orthopaedics, Shanghai Tongji Hospital, Shanghai 200065, P.R. China
| |
Collapse
|
16
|
Belmellat N, Semerano L, Segueni N, Damotte D, Decker P, Ryffel B, Quesniaux V, Boissier MC, Assier E. Tumor Necrosis Factor-Alpha Targeting Can Protect against Arthritis with Low Sensitization to Infection. Front Immunol 2017; 8:1533. [PMID: 29184553 PMCID: PMC5694445 DOI: 10.3389/fimmu.2017.01533] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/27/2017] [Indexed: 01/12/2023] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) blockade is an effective treatment for rheumatoid arthritis (RA) and other inflammatory diseases, but in patients, it is associated with reduced resistance to the infectious agents Mycobacterium tuberculosis and Listeria monocytogenes, among others. Our goal was to model infection and arthritis in mice and to compare etanercept, a currently used anti-TNF-α inhibitor, to an anti-TNF-α vaccine. We developed a murine surrogate of the TNF-α kinoid and produced an anti-murine TNF-α vaccine (TNFKi) composed of keyhole limpet hemocyanin conjugated to TNF-α, which resulted in anti-TNF-α antibody production in mice. We also used etanercept (a soluble receptor of TNF commonly used to treat RA) as a control of TNF neutralization. In a mouse model of collagen-induced arthritis, TNFKi protected against inflammation similar to etanercept. In a mouse model of acute L. monocytogenes infection, all TNFKi-treated mice showed cleared bacterial infection and survived, whereas etanercept-treated mice showed large liver granulomas and quickly died. Moreover, TNFKi mice infected with the virulent H37Rv M. tuberculosis showed resistance to infection, in contrast with etanercept-treated mice or controls. Depending on the TNF-α blockade strategy, treating arthritis with a TNF-α inhibitor could result in a different profile of infection suceptibility. Our TNFKi vaccine allowed for a better remaining host defense than did etanercept.
Collapse
Affiliation(s)
- Nadia Belmellat
- UMR 1125 INSERM, Bobigny, France.,Sorbonne Paris Cité Université Paris 13, Bobigny, France
| | - Luca Semerano
- UMR 1125 INSERM, Bobigny, France.,Sorbonne Paris Cité Université Paris 13, Bobigny, France.,Service de Rhumatologie, Groupe Hospitalier Avicenne-Jean Verdier-René Muret, APHP, Bobigny, France
| | - Noria Segueni
- INEM UMR7355, CNRS, University of Orléans, Orléans, France
| | - Diane Damotte
- Service de pathologie Hôpitaux Universitaires Paris Centre, APHP, Université Paris Descartes, Paris, France
| | - Patrice Decker
- UMR 1125 INSERM, Bobigny, France.,Sorbonne Paris Cité Université Paris 13, Bobigny, France
| | - Bernhard Ryffel
- INEM UMR7355, CNRS, University of Orléans, Orléans, France.,IDM, University of Cape Town, Cape Town, South Africa
| | | | - Marie-Christophe Boissier
- UMR 1125 INSERM, Bobigny, France.,Sorbonne Paris Cité Université Paris 13, Bobigny, France.,Service de Rhumatologie, Groupe Hospitalier Avicenne-Jean Verdier-René Muret, APHP, Bobigny, France
| | - Eric Assier
- UMR 1125 INSERM, Bobigny, France.,Sorbonne Paris Cité Université Paris 13, Bobigny, France
| |
Collapse
|
17
|
Pavshintsev VV, Mitkin NA, Frolova OY, Kushnir EA, Averina OA, Lovat ML. Individual roles of brain and serum alcohol dehydrogenase isoforms in regulation of alcohol consumption in SPF Wistar rats. Physiol Behav 2017; 179:458-466. [DOI: 10.1016/j.physbeh.2017.07.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/29/2017] [Accepted: 07/13/2017] [Indexed: 12/31/2022]
|
18
|
Ying HZ, Chen Q, Zhang WY, Zhang HH, Ma Y, Zhang SZ, Fang J, Yu CH. PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics (Review). Mol Med Rep 2017; 16:7879-7889. [PMID: 28983598 PMCID: PMC5779870 DOI: 10.3892/mmr.2017.7641] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 07/20/2017] [Indexed: 02/06/2023] Open
Abstract
The platelet‑derived growth factor (PDFG) signaling pathway exerts persistent activation in response to a variety of stimuli and facilitates the progression of hepatic fibrosis. Since this pathway modulates a broad spectrum of cellular processes, including cell growth, differentiation, inflammation and carcinogenesis, it has emerged as a therapeutic target for hepatic fibrosis and liver‑associated disorders. The present review exhibits the current knowledge of the role of the PDGF signaling pathway and its pathological profiles in hepatic fibrosis, and assesses the potential of inhibitors which have been investigated in the experimental hepatic fibrosis model, in addition to the clinical challenges associated with these inhibitors.
Collapse
Affiliation(s)
- Hua-Zhong Ying
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Qin Chen
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Wen-You Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Huan-Huan Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Yue Ma
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Song-Zhao Zhang
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jie Fang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
19
|
Mora-Solano C, Wen Y, Han H, Chen J, Chong AS, Miller ML, Pompano RR, Collier JH. Active immunotherapy for TNF-mediated inflammation using self-assembled peptide nanofibers. Biomaterials 2017; 149:1-11. [PMID: 28982051 DOI: 10.1016/j.biomaterials.2017.09.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 11/18/2022]
Abstract
Active immunotherapies raising antibody responses against autologous targets are receiving increasing interest as alternatives to the administration of manufactured antibodies. The challenge in such an approach is generating protective and adjustable levels of therapeutic antibodies while at the same time avoiding strong T cell responses that could lead to autoimmune reactions. Here we demonstrate the design of an active immunotherapy against TNF-mediated inflammation using short synthetic peptides that assemble into supramolecular peptide nanofibers. Immunization with these materials, without additional adjuvants, was able to break B cell tolerance and raise protective antibody responses against autologous TNF in mice. The strength of the anti-TNF antibody response could be tuned by adjusting the epitope content in the nanofibers, and the T-cell response was focused on exogenous and non-autoreactive T-cell epitopes. Immunization with unadjuvanted peptide nanofibers was therapeutic in a lethal model of acute inflammation induced by intraperitoneally delivered lipopolysaccharide, whereas formulations adjuvanted with CpG showed comparatively poorer protection that correlated with a more Th1-polarized response. Additionally, immunization with peptide nanofibers did not diminish the ability of mice to clear infections of Listeria monocytogenes. Collectively this work suggests that synthetic self-assembled peptides can be attractive platforms for active immunotherapies against autologous targets.
Collapse
Affiliation(s)
- Carolina Mora-Solano
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States; Molecular Pathogenesis Program, University of Chicago, Chicago, IL, 60637, United States
| | - Yi Wen
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Huifang Han
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Jianjun Chen
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Anita S Chong
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Michelle L Miller
- Molecular Pathogenesis Program, University of Chicago, Chicago, IL, 60637, United States
| | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22904, United States
| | - Joel H Collier
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States; Molecular Pathogenesis Program, University of Chicago, Chicago, IL, 60637, United States.
| |
Collapse
|
20
|
Struja T, Kutz A, Fischli S, Meier C, Mueller B, Recher M, Schuetz P. Is Graves' disease a primary immunodeficiency? New immunological perspectives on an endocrine disease. BMC Med 2017; 15:174. [PMID: 28942732 PMCID: PMC5611589 DOI: 10.1186/s12916-017-0939-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/04/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Uncertainty about factors influencing the susceptibility and triggers for Graves' disease persists, along with a wide variation in the response to anti-thyroid drugs, currently at approximately 50% of non-responders. The aim of this narrative review is to summarize immunological concepts, with a combined endocrine and immunological perspective, to highlight potential new areas of research. MAIN TEXT Relevant studies were identified through a systematic literature search using the PubMed and EMBASE databases in March 2016. No cut-offs regarding dates were imposed. We used the terms "Graves' Disease" or "Basedow" or "thyrotoxicosis" together with the terms "etiology", "pathophysiology", "immunodeficiency", "causality", and "autoimmunity". The terms "orbitopathy", "ophthalmopathy", and "amiodarone" were excluded. Articles in English, French, German, Croatian, Spanish, and Italian were eligible for inclusion. CONCLUSIONS While concepts such as the impact of iodine, smoking, human leucocyte antigen, infections, and ethnicity are established, new ideas have emerged. Pertaining evidence suggests the involvement of autoimmunity and immunodeficiency in the pathophysiology of Graves' disease. Recent studies point to specific immunological mechanisms triggering the onset of disease, which may also serve as targets for more specific therapies.
Collapse
Affiliation(s)
- Tristan Struja
- Medical University Department, Clinic for Endocrinology, Diabetes & Metabolism, Kantonsspital Aarau, Aarau, Switzerland.
| | - Alexander Kutz
- Medical University Department, Clinic for Endocrinology, Diabetes & Metabolism, Kantonsspital Aarau, Aarau, Switzerland
| | - Stefan Fischli
- Medical Clinic, Department for Endocrinology, Diabetes & Metabolism, Kantonsspital Luzern, Luzern, Switzerland
| | - Christian Meier
- Medical Faculty of the University of Basel, Basel, Switzerland.,Division of Endocrinology, Diabetes & Metabolism, University Hospital and University Basel, Basel, Switzerland
| | - Beat Mueller
- Medical University Department, Clinic for Endocrinology, Diabetes & Metabolism, Kantonsspital Aarau, Aarau, Switzerland.,Medical Faculty of the University of Basel, Basel, Switzerland
| | - Mike Recher
- Medical Faculty of the University of Basel, Basel, Switzerland.,Medical Outpatient Clinic and Immunodeficiency Laboratory, Department of Biomedicine, University Hospital and University Basel, Basel, Switzerland
| | - Philipp Schuetz
- Medical University Department, Clinic for Endocrinology, Diabetes & Metabolism, Kantonsspital Aarau, Aarau, Switzerland.,Medical Faculty of the University of Basel, Basel, Switzerland
| |
Collapse
|
21
|
Do we need animal models to advance research on inflammatory joint disease? Joint Bone Spine 2017; 84:381-383. [PMID: 28456601 DOI: 10.1016/j.jbspin.2017.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2017] [Indexed: 11/20/2022]
|
22
|
Venuturupalli S. Immune Mechanisms and Novel Targets in Rheumatoid Arthritis. Immunol Allergy Clin North Am 2017; 37:301-313. [PMID: 28366478 DOI: 10.1016/j.iac.2017.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Significant progress has been made in understanding the pathophysiology of rheumatoid arthritis. It is widely recognized that early institution of treatment is one of the best predictive factors for response to therapy. Several novel approaches are currently being investigated and several novel biologics are in various stages of development. With advances in the fields of proteomics, genomics, and metabolomics, specific phenotypes of the disease can be better identified and specific therapies for particular phases of the disease and specific patients will allow for improved control of this condition.
Collapse
Affiliation(s)
- Swamy Venuturupalli
- Division of Rheumatology- Cedars Sinai Medical Center, 8750 Wilshire Blvd, Suite 350, Beverly Hills, CA 90211, USA.
| |
Collapse
|
23
|
Assier E, Bessis N, Zagury JF, Boissier MC. IL-1 Vaccination Is Suitable for Treating Inflammatory Diseases. Front Pharmacol 2017; 8:6. [PMID: 28197099 PMCID: PMC5281538 DOI: 10.3389/fphar.2017.00006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/04/2017] [Indexed: 01/25/2023] Open
Affiliation(s)
- Eric Assier
- UMR 1125 Institut National de la Santé et de la Recherche MédicaleBobigny, France; Sorbonne Paris Cité Université Paris 13Bobigny, France
| | - Natacha Bessis
- UMR 1125 Institut National de la Santé et de la Recherche MédicaleBobigny, France; Sorbonne Paris Cité Université Paris 13Bobigny, France
| | | | - Marie-Christophe Boissier
- UMR 1125 Institut National de la Santé et de la Recherche MédicaleBobigny, France; Sorbonne Paris Cité Université Paris 13Bobigny, France; Assistance Publique-Hôpitaux de Paris, HUPSSD, Service de RhumatologieBobigny, France
| |
Collapse
|
24
|
Targeted vaccination against the bevacizumab binding site on VEGF using 3D-structured peptides elicits efficient antitumor activity. Proc Natl Acad Sci U S A 2016; 113:12532-12537. [PMID: 27791128 DOI: 10.1073/pnas.1610258113] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Therapeutic targeting of the VEGF signaling axis by the VEGF-neutralizing monoclonal antibody bevacizumab has clearly demonstrated clinical benefit in cancer patients. To improve this strategy using a polyclonal approach, we developed a vaccine targeting VEGF using 3D-structured peptides that mimic the bevacizumab binding site. An in-depth study on peptide optimization showed that the antigen's 3D structure is essential to achieve neutralizing antibody responses. Peptide 1 adopts a clear secondary, native-like structure, including the typical cysteine-knot fold, as evidenced by CD spectroscopy. Binding and competition studies with bevacizumab in ELISA and surface plasmon resonance analysis revealed that peptide 1 represents the complete bevacizumab binding site, including the hairpin loop (β5-turn-β6) and the structure-supporting β2-α2-β3 loop. Vaccination with peptide 1 elicited high titers of cross-reactive antibodies to VEGF, with potent neutralizing activity. Moreover, vaccination-induced antisera displayed strong angiostatic and tumor-growth-inhibiting properties in a preclinical mouse model for colorectal carcinoma, whereas antibodies raised with peptides exclusively encompassing the β5-turn-β6 loop (peptides 15 and 20) did not. Immunization with peptide 1 or 7 (murine analog of 1) in combination with the potent adjuvant raffinose fatty acid sulfate ester (RFASE) showed significant inhibition of tumor growth in the B16F10 murine melanoma model. Based on these data, we conclude that this vaccination technology, which is currently being investigated in a phase I clinical trial (NCT02237638), can potentially outperform currently applied anti-VEGF therapeutics.
Collapse
|
25
|
Rodríguez-Álvarez Y, Morera-Díaz Y, Gerónimo-Pérez H, Castro-Velazco J, Martínez-Castillo R, Puente-Pérez P, Besada-Pérez V, Hardy-Rando E, Chico-Capote A, Martínez-Cordovez K, Santos-Savio A. Active immunization with human interleukin-15 induces neutralizing antibodies in non-human primates. BMC Immunol 2016; 17:30. [PMID: 27671547 PMCID: PMC5036325 DOI: 10.1186/s12865-016-0168-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/12/2016] [Indexed: 02/07/2023] Open
Abstract
Background Interleukin-15 is an immunostimulatory cytokine overexpressed in several autoimmune and inflammatory diseases such as Rheumatoid Arthritis, psoriasis and ulcerative colitis; thus, inhibition of IL-15-induced signaling could be clinically beneficial in these disorders. Our approach to neutralize IL-15 consisted in active immunization with structurally modified human IL-15 (mhIL-15) with the aim to induce neutralizing antibodies against native IL-15. In the present study, we characterized the antibody response in Macaca fascicularis, non-human primates that were immunized with a vaccine candidate containing mhIL-15 in Aluminum hydroxide (Alum), Montanide and Incomplete Freund’s Adjuvant. Results Immunization with mhIL-15 elicited a specific antibodies response that neutralized native IL-15-dependent biologic activity in a CTLL-2 cell proliferation assay. The highest neutralizing response was obtained in macaques immunized with mhIL-15 adjuvanted in Alum. This response, which was shown to be transient, also inhibited the activity of simian IL-15 and did not affect the human IL-2-induced proliferation of CTLL-2 cells. Also, in a pool of synovial fluid cells from two Rheumatoid Arthritis patients, the immune sera slightly inhibited TNF-α secretion. Finally, it was observed that this vaccine candidate neither affect animal behavior, clinical status, blood biochemistry nor the percentage of IL-15-dependent cell populations, specifically CD56+ NK and CD8+ T cells. Conclusion Our results indicate that vaccination with mhIL-15 induced neutralizing antibodies to native IL-15 in non-human primates. Based on this fact, we propose that this vaccine candidate could be potentially beneficial for treatment of diseases where IL-15 overexpression is associated with their pathogenesis.
Collapse
Affiliation(s)
- Yunier Rodríguez-Álvarez
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba.
| | - Yanelys Morera-Díaz
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba
| | - Haydee Gerónimo-Pérez
- Quality Control Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Jorge Castro-Velazco
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Rafael Martínez-Castillo
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Pedro Puente-Pérez
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Vladimir Besada-Pérez
- Chemistry and Physics Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Eugenio Hardy-Rando
- Biotechnology Laboratory, Study Center for Research and Biological Evaluations, Institute of Pharmacy and Foods, Havana University, Avenue 222, PO Box 13600, Havana, 10600, Cuba
| | - Araceli Chico-Capote
- Rheumatology Department, Hermanos Ameijeiras Hospital, San Lazaro 701, PO Box 6122, Havana, 10600, Cuba
| | - Klaudia Martínez-Cordovez
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba
| | - Alicia Santos-Savio
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba
| |
Collapse
|
26
|
A Rationally Designed TNF-α Epitope-Scaffold Immunogen Induces Sustained Antibody Response and Alleviates Collagen-Induced Arthritis in Mice. PLoS One 2016; 11:e0163080. [PMID: 27658047 PMCID: PMC5033357 DOI: 10.1371/journal.pone.0163080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 09/04/2016] [Indexed: 12/29/2022] Open
Abstract
The TNF-α biological inhibitors have significantly improved the clinical outcomes of many autoimmune diseases, in particular rheumatoid arthritis. However, the practical uses are limited due to high costs and the risk of anti-drug antibody responses. Attempts to develop anti-TNF-α vaccines have generated encouraging data in animal models, however, data from clinical trials have not met expectations. In present study, we designed a TNF-α epitope-scaffold immunogen DTNF7 using the transmembrane domain of diphtheria toxin, named DTT as a scaffold. Molecular dynamics simulation shows that the grafted TNF-α epitope is entirely surface-exposed and presented in a native-like conformation while the rigid helical structure of DTT is minimally perturbed, thereby rendering the immunogen highly stable. Immunization of mice with alum formulated DTNF7 induced humoral responses against native TNF-α, and the antibody titer was sustained for more than 6 months, which supports a role of the universal CD4 T cell epitopes of DTT in breaking self-immune tolerance. In a mouse model of rheumatoid arthritis, DTNF7-alum vaccination markedly delayed the onset of collagen-induced arthritis, and reduced incidence as well as clinical score. DTT is presumed safe as an epitope carrier because a catalytic inactive mutant of diphtheria toxin, CRM197 has good clinical safety records as an active vaccine component. Taken all together, we show that DTT-based epitope vaccine is a promising strategy for prevention and treatment of autoimmune diseases.
Collapse
|
27
|
A virus-like particle-based connective tissue growth factor vaccine suppresses carbon tetrachloride-induced hepatic fibrosis in mice. Sci Rep 2016; 6:32155. [PMID: 27562139 PMCID: PMC4999884 DOI: 10.1038/srep32155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/03/2016] [Indexed: 12/30/2022] Open
Abstract
Connective tissue growth factor (CTGF) has been recognized as a central mediator and promising therapeutic target in hepatic fibrosis. In this study, we generated a novel virus-like particle (VLP) CTGF vaccine by inserting the 138–159 amino acid (aa) fragment of CTGF into the central c/e1 epitope of C-terminus truncated hepatitis B virus core antigen (HBc, aa 1–149) using a prokaryotic expression system. Immunization of BALB/c mice with the VLP vaccine efficiently elicited the production of anti-CTGF neutralizing antibodies. Vaccination with this CTGF vaccine significantly protected BALB/c mice from carbon tetrachloride (CCl4)-induced hepatic fibrosis, as indicated by decreased hepatic hydroxyproline content and lower fibrotic score. CCl4 intoxication-induced hepatic stellate cell activation was inhibited by the vaccination, as indicated by decreased α-smooth muscle actin expression and Smad2 phosphorylation. Vaccination against CTGF also attenuated the over-expression of some profibrogenic factors, such as CTGF, transforming growth factor-β1, platelet-derived growth factor-B and tissue inhibitor of metalloproteinase-1 in the fibrotic mouse livers, decreased hepatocyte apoptosis and accelerated hepatocyte proliferation in the fibrotic mouse livers. Our results clearly indicate that vaccination against CTGF inhibits fibrogenesis, alleviates hepatocyte apoptosis and facilitate hepatic regeneration. We suggest that the vaccine should be developed into an effective therapeutic measure for hepatic fibrosis.
Collapse
|
28
|
Pouillon L, Bossuyt P, Peyrin-Biroulet L. Considerations, challenges and future of anti-TNF therapy in treating inflammatory bowel disease. Expert Opin Biol Ther 2016; 16:1277-90. [PMID: 27329436 DOI: 10.1080/14712598.2016.1203897] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Crohn's disease (CD) and ulcerative colitis (UC) are chronic disabling conditions. Monoclonal antibody therapy directed against tumor necrosis factor-alpha (anti-TNF) has revolutionized the care of patients with inflammatory bowel disease (IBD). AREAS COVERED Considerations before starting anti-TNF therapy are highlighted: the best time to start with anti-TNF therapy, either alone or in combination with an immunomodulator, the choice of an anti-TNF agent and the contra-indications to anti-TNF therapy. Primary nonresponse and secondary loss of response are discussed. De-escalating therapy, the role of therapeutic drug monitoring and the use of biosimilars, are handled. Finally, the future directions of anti-TNF therapy are emphasized. EXPERT OPINION Anti-TNF therapy remains the cornerstone in the treatment of IBD. When initiating long-term therapy, safety and cost issues are of great importance. The therapeutic armamentarium in the treatment of IBD is rapidly growing. Therefore, the challenge is to optimize the use and refine the exact position of anti-TNF therapy in the near future, with personalized medicine as the ultimate goal.
Collapse
Affiliation(s)
- Lieven Pouillon
- a Department of Hepato-Gastroenterology , University Hospitals Leuven, Uz Gasthuisberg , Leuven , Belgium
| | - Peter Bossuyt
- b Imelda GI Clinical Research Centre , Imeldaziekenhuis Bonheiden , Bonheiden , Belgium
| | - Laurent Peyrin-Biroulet
- c Inserm U954 and Department of Gastroenterology , Nancy University Hospital, Université de Lorraine , Vandœuvre-lès-Nancy , France
| |
Collapse
|
29
|
Favoino E, Prete M, Marzullo A, Millo E, Shoenfeld Y, Perosa F. CD20-Mimotope Peptide Active Immunotherapy in Systemic Lupus Erythematosus and a Reappraisal of Vaccination Strategies in Rheumatic Diseases. Clin Rev Allergy Immunol 2016; 52:217-233. [DOI: 10.1007/s12016-016-8551-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
30
|
Sánchez Ramírez J, Morera Díaz Y, Musacchio Lasa A, Bequet-Romero M, Muñoz Pozo Y, Pérez Sánchez L, Hernández-Bernal F, Mendoza Fuentes O, Selman-Housein KH, Gavilondo Cowley JV, Ayala Avila M. Indirect and competitive enzyme-linked immunosorbent assays for monitoring the humoral response against human VEGF. J Immunoassay Immunochem 2016; 37:636-58. [PMID: 27143151 DOI: 10.1080/15321819.2016.1184164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
CIGB-247, a VEGF-based vaccine, was studied in a clinical trial. This advance demands the refinement of the methodologies for assessment of vaccine immune responses. This study aimed to improve the performance of ELISAs for detecting IgG antibodies against human VEGF and the blocking activity of the serum to inhibit the VEGF/VEGFR2 interaction. The best experimental conditions were established through the evaluation of several blocking buffers, immobilization surfaces, and plate suppliers using human sera as test samples. As a result, two controlled ELISAs were used in testing of elicited immune response against VEGF in patients immunized with CIGB-247.
Collapse
Affiliation(s)
- Javier Sánchez Ramírez
- a Department of Pharmaceuticals , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| | - Yanelys Morera Díaz
- a Department of Pharmaceuticals , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| | - Alexis Musacchio Lasa
- b Department of Bioinformatics , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| | - Mónica Bequet-Romero
- a Department of Pharmaceuticals , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| | - Yasmiana Muñoz Pozo
- a Department of Pharmaceuticals , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| | - Lincidio Pérez Sánchez
- a Department of Pharmaceuticals , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| | - Francisco Hernández-Bernal
- c Department of Clinical Research , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| | - Osmany Mendoza Fuentes
- d Animal Facility , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| | | | - Jorge Víctor Gavilondo Cowley
- a Department of Pharmaceuticals , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| | - Marta Ayala Avila
- a Department of Pharmaceuticals , Center for Genetic Engineering and Biotechnology (CIGB) , Playa Cubanacán, Havana , Cuba
| |
Collapse
|
31
|
Novel Immunotherapeutic Avenues for Rheumatoid Arthritis. Trends Mol Med 2016; 22:214-229. [PMID: 26875450 DOI: 10.1016/j.molmed.2016.01.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 01/07/2023]
Abstract
Rheumatoid arthritis (RA) is the most common inflammatory rheumatic disease. It leads to irreversible joint damage, physical handicap, and reduced life expectancy. The past two decades have seen considerable therapeutic advances with the development of biologic treatments to block proinflammatory cytokines or modulate lymphocyte function, followed by the development of small molecules to target intracellular signaling. Nevertheless, only a minority of patients can achieve disease remission, especially long term, warranting further investigation into newer therapeutic options. Targeting single proinflammatory pathways may not be sufficient, as suggested by variable results with T helper (Th)-17-related cytokine blockade. Multilevel information from 'omics' techniques along with data from mechanistic studies might facilitate the identification of pivotal checkpoints in RA disease pathogenesis and the subsequent development of new effective treatments.
Collapse
|
32
|
Kalliolias GD, Ivashkiv LB. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol 2015; 12:49-62. [PMID: 26656660 DOI: 10.1038/nrrheum.2015.169] [Citation(s) in RCA: 907] [Impact Index Per Article: 90.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
TNF is a pleiotropic cytokine with important functions in homeostasis and disease pathogenesis. Recent discoveries have provided insights into TNF biology that introduce new concepts for the development of therapeutics for TNF-mediated diseases. The model of TNF receptor signalling has been extended to include linear ubiquitination and the formation of distinct signalling complexes that are linked with different functional outcomes, such as inflammation, apoptosis and necroptosis. Our understanding of TNF-induced gene expression has been enriched by the discovery of epigenetic mechanisms and concepts related to cellular priming, tolerization and induction of 'short-term transcriptional memory'. Identification of distinct homeostatic or pathogenic TNF-induced signalling pathways has introduced the concept of selectively inhibiting the deleterious effects of TNF while preserving its homeostatic bioactivities for therapeutic purposes. In this Review, we present molecular mechanisms underlying the roles of TNF in homeostasis and inflammatory disease pathogenesis, and discuss novel strategies to advance therapeutic paradigms for the treatment of TNF-mediated diseases.
Collapse
Affiliation(s)
- George D Kalliolias
- Arthritis &Tissue Degeneration Program and David Z. Rosensweig Center for Genomics Research, Hospital for Special Surgery, 535 E 70th Street, New York, New York 10021, USA
| | - Lionel B Ivashkiv
- Arthritis &Tissue Degeneration Program and David Z. Rosensweig Center for Genomics Research, Hospital for Special Surgery, 535 E 70th Street, New York, New York 10021, USA
| |
Collapse
|
33
|
Semerano L, Duvallet E, Belmellat N, Marival N, Schall N, Monteil M, Grouard-Vogel G, Bernier E, Lecouvey M, Hlawaty H, Muller S, Boissier MC, Assier E. Targeting VEGF-A with a vaccine decreases inflammation and joint destruction in experimental arthritis. Angiogenesis 2015; 19:39-52. [PMID: 26419779 DOI: 10.1007/s10456-015-9487-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 09/16/2015] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Inflammation and angiogenesis are two tightly linked processes in arthritis, and therapeutic targeting of pro-angiogenic factors may contribute to control joint inflammation and synovitis progression. In this work, we explored whether vaccination against vascular endothelial growth factor (VEGF) ameliorates collagen-induced arthritis (CIA). METHODS Anti-VEGF vaccines were heterocomplexes consisting of the entire VEGF cytokine (or a VEGF-derived peptide) linked to the carrier protein keyhole limpet hemocyanin (KLH). Two kinds of vaccines were separately tested in two independent experiments of CIA. In the first, we tested a kinoid of the murine cytokine VEGF (VEGF-K), obtained by conjugating VEGF-A to KLH. For the second, we selected two VEGF-A-derived peptide sequences to produce heterocomplexes (Vpep1-K and Vpep2-K). DBA/1 mice were immunized with either VEGF-K, Vpep1-K, or Vpep2-K, before CIA induction. Clinical and histological scores of arthritis, anti-VEGF, anti-Vpep Ab titers, and anti-VEGF Abs neutralizing capacity were determined. RESULTS Both VEGF-K and Vpep1-K significantly ameliorated clinical arthritis scores and reduced synovial inflammation and joint destruction at histology. VEGF-K significantly reduced synovial vascularization. None of the vaccines reduced anti-collagen Ab response in mice. Both VEGF-K and Vpep1-K induced persistently high titers of anti-VEGF Abs capable of inhibiting VEGF-A bioactivity. CONCLUSION Vaccination against the pro-angiogenic factor VEGF-A leads to the production of anti-VEGF polyclonal Abs and has a significant anti-inflammatory effect in CIA. Restraining Ab response to a single peptide sequence (Vpep1) with a peptide vaccine effectively protects immunized mice from joint inflammation and destruction.
Collapse
Affiliation(s)
- Luca Semerano
- Inserm UMR 1125, Sorbonne Paris Cité - Université Paris 13, 74, rue Marcel Cachin, 93017, Bobigny, France. .,Sorbonne Paris Cité - Université Paris 13, 74 rue Marcel Cachin, 93000, Bobigny, France. .,Service de Rhumatologie, Assistance Publique - Hôpitaux de Paris (AP-HP) Groupe hospitalier Avicenne - Jean Verdier - René Muret, 125 rue de Stalingrad, 93000, Bobigny, France.
| | - Emilie Duvallet
- Inserm UMR 1125, Sorbonne Paris Cité - Université Paris 13, 74, rue Marcel Cachin, 93017, Bobigny, France. .,Sorbonne Paris Cité - Université Paris 13, 74 rue Marcel Cachin, 93000, Bobigny, France.
| | - Nadia Belmellat
- Inserm UMR 1125, Sorbonne Paris Cité - Université Paris 13, 74, rue Marcel Cachin, 93017, Bobigny, France. .,Sorbonne Paris Cité - Université Paris 13, 74 rue Marcel Cachin, 93000, Bobigny, France.
| | - Nicolas Marival
- Sorbonne Paris Cité - Université Paris 13, 74 rue Marcel Cachin, 93000, Bobigny, France. .,Inserm UMR 1148, 74 rue Marcel Cachin, 93000, Bobigny, France.
| | - Nicolas Schall
- CNRS, Immunopathologie et chimie thérapeutique/Laboratory of excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, 15 rue René Descartes, 67000, Strasbourg, France.
| | - Maëlle Monteil
- Sorbonne Paris Cité - Université Paris 13, 74 rue Marcel Cachin, 93000, Bobigny, France. .,CNRS UMR 7244, 74 rue Marcel Cachin, 93000, Bobigny, France.
| | | | | | - Marc Lecouvey
- Sorbonne Paris Cité - Université Paris 13, 74 rue Marcel Cachin, 93000, Bobigny, France. .,CNRS UMR 7244, 74 rue Marcel Cachin, 93000, Bobigny, France.
| | - Hanna Hlawaty
- Sorbonne Paris Cité - Université Paris 13, 74 rue Marcel Cachin, 93000, Bobigny, France. .,Inserm UMR 1148, 74 rue Marcel Cachin, 93000, Bobigny, France.
| | - Sylviane Muller
- CNRS, Immunopathologie et chimie thérapeutique/Laboratory of excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, 15 rue René Descartes, 67000, Strasbourg, France.
| | - Marie-Christophe Boissier
- Inserm UMR 1125, Sorbonne Paris Cité - Université Paris 13, 74, rue Marcel Cachin, 93017, Bobigny, France. .,Sorbonne Paris Cité - Université Paris 13, 74 rue Marcel Cachin, 93000, Bobigny, France. .,Service de Rhumatologie, Assistance Publique - Hôpitaux de Paris (AP-HP) Groupe hospitalier Avicenne - Jean Verdier - René Muret, 125 rue de Stalingrad, 93000, Bobigny, France.
| | - Eric Assier
- Inserm UMR 1125, Sorbonne Paris Cité - Université Paris 13, 74, rue Marcel Cachin, 93017, Bobigny, France. .,Sorbonne Paris Cité - Université Paris 13, 74 rue Marcel Cachin, 93000, Bobigny, France.
| |
Collapse
|
34
|
Liu C, Zhao Y, He W, Wang W, Chen Y, Zhang S, Ma Y, Gohda J, Ishida T, Walter TS, Owens RJ, Stuart DI, Ren J, Gao B. A RANKL mutant used as an inter-species vaccine for efficient immunotherapy of osteoporosis. Sci Rep 2015; 5:14150. [PMID: 26412210 PMCID: PMC4585926 DOI: 10.1038/srep14150] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 08/14/2015] [Indexed: 01/01/2023] Open
Abstract
Anti-cytokine therapeutic antibodies have been demonstrated to be effective in the treatment of several auto-immune disorders. However, The problems in antibody manufacture and the immunogenicity caused by multiple doses of antibodies inspire people to use auto-cytokine as immunogen to induce anti-cytokine antibodies. Nevertheless, the tolerance for inducing immune response against self-antigen has hindered the wide application of the strategy. To overcome the tolerance, here we proposed a strategy using the inter-species cytokine as immunogen for active immunization (TISCAI) to induce anti-cytokine antibody. As a proof of concept, an inter-species cytokine RANKL was successfully used as immunogen to induce anti-RANKL immune response. Furthermore, to prevent undesirable side-effects, the human RANKL was mutated based on the crystal structure of the complex of human RANKL and its rodent counterpart receptor RANK. We found, the antibodies produced blocked the osteoclast development in vitro and osteoporosis in OVX rat models. The results demonstrated this strategy adopted is very useful for general anti-cytokine immunotherapy for different diseases settings.
Collapse
Affiliation(s)
- Changzhen Liu
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, NO.16, Dongzhimennei South Street, Dongcheng District, Beijing 100700, China
| | - Yunfeng Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 1 Beichen Xilu, Beijing 100101, China
| | - Wen He
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 1 Beichen Xilu, Beijing 100101, China
| | - Wei Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 1 Beichen Xilu, Beijing 100101, China
| | - Yuan Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 1 Beichen Xilu, Beijing 100101, China
| | - Shiqian Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 1 Beichen Xilu, Beijing 100101, China
| | - Yijing Ma
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 1 Beichen Xilu, Beijing 100101, China
| | - Jin Gohda
- Research Center for Asian Infectious Diseases, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai Minato-ku, Tokyo 108-8639, Japan
| | - Takaomi Ishida
- Research Center for Asian Infectious Diseases, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai Minato-ku, Tokyo 108-8639, Japan
| | - Thomas S Walter
- Division of Structural Biology, The Wellcome Trust Centre for Human Genetics, The Henry Welcome Building for Genomic Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Raymond J Owens
- Oxford Protein Production Facility UK, Research Complex at Harwell, Rutherford Appleton Laboratory Harwell, Science and Innovation Campus, Oxfordshire, OX11 0FA, UK
| | - David I Stuart
- Division of Structural Biology, The Wellcome Trust Centre for Human Genetics, The Henry Welcome Building for Genomic Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Jingshan Ren
- Division of Structural Biology, The Wellcome Trust Centre for Human Genetics, The Henry Welcome Building for Genomic Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | | |
Collapse
|