1
|
Karnam S, Jindal AB, Paul AT. Quality by design-based optimization of teriflunomide and quercetin combinational topical transferosomes for the treatment of rheumatoid arthritis. Int J Pharm 2024; 666:124829. [PMID: 39406305 DOI: 10.1016/j.ijpharm.2024.124829] [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: 06/24/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Rheumatoid arthritis (RA) is an immune-mediated inflammatory disease. Combination therapy is anticipated to surpass monotherapy by targeting multiple pathways involved in RA progression. The present aim is to develop a combination of Teriflunomide (TFD) and Quercetin (QCN) loaded transferosomal gel to enhance permeability and achieve localized delivery to joint tissues. TFD or QCN transferosomes were optimized employing a 3-level, 3-factorial design Box-Behnken design (BBD). The transferosomes exhibited sustained in-vitro drug release. The topical combination gel underwent thorough evaluation of rheology, and also ex-vivo studies showed enhanced permeability through rat skin. The synergistic combination of TFD and QCN effectively suppressed NO, TNF-α and IL-6 levels in in-vitro RAW 264.7 cells. The cytotoxicity in HaCaT cell lines indicates non-toxicity of the gel, further confirmed by skin irritation study conducted in rats. The in-vivo anti-arthritic activity was evaluated in complete freund's adjuvant induced rat paw edema model illustrates the effectiveness of the combination transferosomal gel compared to other treatment groups. In conclusion, the topical delivery of TFD and QCN combination transferosomal gel demonstrated anti-arthritic activity through localized delivery whichallows for dose reduction, thereby may reduce the systemic drug exposure and mitigate the side effects associated with oral administration of TFD.
Collapse
Affiliation(s)
- Sriravali Karnam
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Pilani Campus, Rajasthan 333031, India
| | - Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Pilani Campus, Rajasthan 333031, India
| | - Atish T Paul
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
2
|
Zewail M. Leflunomide nanocarriers: a new prospect of therapeutic applications. J Microencapsul 2024; 41:715-738. [PMID: 39320955 DOI: 10.1080/02652048.2024.2407373] [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: 05/01/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Leflunomide (LEF) is a well-known disease-modifying anti-rheumatic agent (DMARDs) that was approved in 1998 for rheumatoid arthritis (RA) management. It is enzymatically converted into active metabolite teriflunomide (TER) inside the body. LEF and TER possess several pharmacological effects in a variety of diseases including multiple sclerosis, cancer, viral infections and neurobehavioral brain disorders. Despite the aforementioned pharmacological effects exploring these effects in nanomedicine applications has been focused mainly on RA and cancer treatment. This review summarises the main pharmacological, and pharmacokinetic effects of LEF along with highlighting the applications of nanoencapsulation of LEF and its metabolite in different diseases.
Collapse
Affiliation(s)
- Mariam Zewail
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| |
Collapse
|
3
|
Mirhani A, Auajjar N, Slimani C, Attarassi B. [Effectiveness and tolerance of azathioprine as first-line treatment for multiple sclerosis: a case study from Morocco]. Pan Afr Med J 2024; 49:16. [PMID: 39711832 PMCID: PMC11662217 DOI: 10.11604/pamj.2024.49.16.38051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 07/23/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction the purpose of our study is to evaluate the efficacy of azathioprine as first-line treatment in patients with relapsing-remitting multiple sclerosis (RRMS) or progressive multiple sclerosis (PMS), who were supposed to be treated with beta-interferons but, due to limited resources, received azathioprine instead. Method among the 31 patients, 17 had relapsing-remitting MS (RRMS), 11 had primary progressive MS (PPMS), and 3 had secondary progressive MS (SPMS). Patients received azathioprine orally at a dose of 3 mg/kg/day over 2 years. The effect of azathioprine was assessed using the Expanded Disability Status Scale (EDSS) score. Compare means t-test was used to determine the treatment performance. Results for RRMS, the mean EDSS score ranged between 4.2 and 3.6±1.4. A total of 82.4% of patients were stable or improved, with a very highly significant difference (P= 0.000). For PPMS, the mean EDSS score remained constant at 5.7 ±0.4. A total of 54.5% of patients showed stability or improvement, with a highly significant difference (P= 0.005). In the case of SPMS, the mean EDSS score ranged between 4.5 and 4.1±1.3. A total of 66.7% of patients were stable or improved but with a non-significant difference (P= 0.1). Conclusion the results of this study confirm that azathioprine is beneficial in the treatment of different types of multiple sclerosis in Morocco.
Collapse
Affiliation(s)
- Ali Mirhani
- Laboratoire de Biologie et Santé, Faculté des Sciences, Université Ibn-Tofail, Kenitra, Maroc
| | - Nabila Auajjar
- Laboratoire de Biologie et Santé, Faculté des Sciences, Université Ibn-Tofail, Kenitra, Maroc
| | - Chouki Slimani
- Centre Hospitalier Universitaire, Service de Neurologie, Hôpital El Idrissi, Kenitra, Maroc
| | - Benaissa Attarassi
- Laboratoire de Biologie et Santé, Faculté des Sciences, Université Ibn-Tofail, Kenitra, Maroc
| |
Collapse
|
4
|
Dejbakht M, Akhzari M, Jalili S, Faraji F, Barazesh M. Multiple Sclerosis: New Insights into Molecular Pathogenesis and Novel Platforms for Disease Treatment. Curr Drug Res Rev 2024; 16:175-197. [PMID: 37724675 DOI: 10.2174/2589977516666230915103730] [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: 01/08/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Multiple sclerosis (MS), a chronic inflammatory disorder, affects the central nervous system via myelin degradation. The cause of MS is not fully known, but during recent years, our knowledge has deepened significantly regarding the different aspects of MS, including etiology, molecular pathophysiology, diagnosis and therapeutic options. Myelin basic protein (MBP) is the main myelin protein that accounts for maintaining the stability of the myelin sheath. Recent evidence has revealed that MBP citrullination or deamination, which is catalyzed by Ca2+ dependent peptidyl arginine deiminase (PAD) enzyme leads to the reduction of positive charge, and subsequently proteolytic cleavage of MBP. The overexpression of PAD2 in the brains of MS patients plays an essential role in new epitope formation and progression of the autoimmune disorder. Some drugs have recently entered phase III clinical trials with promising efficacy and will probably obtain approval in the near future. As different therapeutic platforms develop, finding an optimal treatment for each individual patient will be more challenging. AIMS This review provides a comprehensive insight into MS with a focus on its pathogenesis and recent advances in diagnostic methods and its present and upcoming treatment modalities. CONCLUSION MS therapy alters quickly as research findings and therapeutic options surrounding MS expand. McDonald's guidelines have created different criteria for MS diagnosis. In recent years, ever-growing interest in the development of PAD inhibitors has led to the generation of many reversible and irreversible PAD inhibitors against the disease with satisfactory therapeutic outcomes.
Collapse
Affiliation(s)
- Majid Dejbakht
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Morteza Akhzari
- School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Sajad Jalili
- Department of Orthopedics, School of Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Fouziyeh Faraji
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Mahdi Barazesh
- Department of Biotechnology, Cellular and Molecular Research Center, School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
5
|
Giovannoni G, Hawkes CH, Lechner-Scott J, Levy M, Yeh EA. Is it ethical to use teriflunomide as an active comparator in phase 3 trials? Mult Scler Relat Disord 2023; 78:104911. [PMID: 37582327 DOI: 10.1016/j.msard.2023.104911] [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: 07/08/2023] [Accepted: 07/21/2023] [Indexed: 08/17/2023]
Abstract
Ethical concerns have been raised about the practice of using teriflunomide, an oral licensed disease-modifying therapy, as an active comparator in phase 3 multiple sclerosis (MS) trials. The assumption is based on the perceived low efficacy of teriflunomide as judged by its effect on relapses and focal MRI activity. However, when you look beyond focal inflammation, teriflunomide has a robust impact on disability progression and a similar effect to the anti-CD20 monoclonal antibody therapies on slowing down the accelerated brain volume loss associated with MS. Teriflunomide is also more effective when used second or third line. The other classes of disease-modifying therapies have problems with their use as active comparators in clinical trials. Using a non-inferiority or equivalence trial design has its own unique set of regulatory and ethical challenges and is not necessarily a solution. There are also economic, altruistic and pragmatic reasons for continuing to use teriflunomide as an active comparator in MS clinical trials. An online survey indicates that the majority of the MS community feels it is still ethical to randomise subjects to teriflunomide and that procedures can be put in place to protect trial subjects randomised to teriflunomide. Therefore, we still have equipoise, and teriflunomide comparator trials are ethical.
Collapse
Affiliation(s)
- Gavin Giovannoni
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Christopher H Hawkes
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Michael Levy
- Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - E Ann Yeh
- Department of Paediatrics, Dalla Lana School of Public Health, University of Toronto, Canada
| |
Collapse
|
6
|
Letarouilly JG, Vermersch P, Flipo RM. Therapeutic consequences in patients with both inflammatory rheumatic diseases and multiple sclerosis. Rheumatology (Oxford) 2023; 62:2352-2359. [PMID: 36440887 DOI: 10.1093/rheumatology/keac665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/19/2022] [Indexed: 07/20/2023] Open
Abstract
Dealing with patients with both multiple sclerosis (MS) and inflammatory rheumatic disorders (IRDs) is not uncommon for a rheumatologist, as there is a statistical association between SpA and MS. As several CNS demyelinating events have been reported in patients treated with TNF inhibitor (TNFi), the pre-existing demyelinating disease was considered a contraindication for TNFi. However, this contraindication is mainly based on a randomized controlled trial in MS and not on large epidemiological studies. According to the last epidemiological studies, TNFi might not be an inducer of MS. Moreover, there are no clear recommendations on the use of the other DMARDs in patients suffering from an IRD and MS. In this review, we summarize the link between MS and IRDs and the impact of DMARDs on MS, especially TNFi. We also look at the impact of disease-modifying drugs for adults with MS and IRDs.
Collapse
Affiliation(s)
| | - Patrick Vermersch
- Université de Lille, CHU Lille, INSERM UMR1172 LilNCog, FHU PRECISE, Service de Neurologie, Lille, France
| | - René-Marc Flipo
- Université de Lille, CHU Lille, FHU PRECISE, Service de Rhumatologie, Lille, France
| |
Collapse
|
7
|
Kihara Y, Chun J. Molecular and neuroimmune pharmacology of S1P receptor modulators and other disease-modifying therapies for multiple sclerosis. Pharmacol Ther 2023; 246:108432. [PMID: 37149155 DOI: 10.1016/j.pharmthera.2023.108432] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023]
Abstract
Multiple sclerosis (MS) is a neurological, immune-mediated demyelinating disease that affects people in the prime of life. Environmental, infectious, and genetic factors have been implicated in its etiology, although a definitive cause has yet to be determined. Nevertheless, multiple disease-modifying therapies (DMTs: including interferons, glatiramer acetate, fumarates, cladribine, teriflunomide, fingolimod, siponimod, ozanimod, ponesimod, and monoclonal antibodies targeting ITGA4, CD20, and CD52) have been developed and approved for the treatment of MS. All the DMTs approved to date target immunomodulation as their mechanism of action (MOA); however, the direct effects of some DMTs on the central nervous system (CNS), particularly sphingosine 1-phosphate (S1P) receptor (S1PR) modulators, implicate a parallel MOA that may also reduce neurodegenerative sequelae. This review summarizes the currently approved DMTs for the treatment of MS and provides details and recent advances in the molecular pharmacology, immunopharmacology, and neuropharmacology of S1PR modulators, with a special focus on the CNS-oriented, astrocyte-centric MOA of fingolimod.
Collapse
Affiliation(s)
- Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, United States of America.
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, United States of America
| |
Collapse
|
8
|
Wu Q, Wang Q, Yang J, Mills EA, Chilukuri P, Saad A, Dowling CA, Fisher C, Kirch B, Mao-Draayer Y. Teriflunomide modulates both innate and adaptive immune capacities in multiple sclerosis. Mult Scler Relat Disord 2023; 75:104719. [PMID: 37172367 DOI: 10.1016/j.msard.2023.104719] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/02/2023] [Accepted: 04/15/2023] [Indexed: 05/14/2023]
Abstract
BACKGROUND Teriflunomide (TER) (Aubagio™) is an FDA-approved disease-modifying therapy (DMT) for relapsing-remitting multiple sclerosis (RRMS). The mechanism of action of TER is thought to be related to the inhibition of dihydroorotate dehydrogenase (DHODH), a key mitochondrial enzyme in the de novo pyrimidine synthesis pathway required by rapidly dividing lymphocytes. Several large pivotal studies have established the efficacy and safety of TER in patients with RRMS. Despite this, little is known about how the adaptive and innate immune cell subsets are affected by the treatment in patients with MS. METHODS We recruited 20 patients with RRMS who were newly started on TER and performed multicolor flow cytometry and functional assays on peripheral blood samples. A paired t-test was used for the statistical analysis and comparison. RESULTS Our data showed that TER promoted a tolerogenic environment by shifting the balance between activated pathogenic and naïve or immunosuppressive immune cell subsets. In our cohort, TER increased the expression of the immunosuppressive marker CD39 on regulatory T cells (Tregs) while it decreased the expression of the activation marker CXCR3 on CD4+ T helper cells. TER treatment also reduced switched memory (sm) B cells while it increased naïve B cells and downregulated the expression of co-stimulatory molecules CD80 and CD86. Additionally, TER reduced the percentage and absolute numbers of natural killer T (NKT) cells, as well as the percentage of natural killer (NK) cells and showed a trend toward reducing the CD56dim NK pathogenic subset. CONCLUSION TER promotes the tolerogenic immune response and suppresses the pathogenic immune response in patients with RRMS.
Collapse
Affiliation(s)
- Qi Wu
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Qin Wang
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jennifer Yang
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Elizabeth A Mills
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pavani Chilukuri
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Aiya Saad
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Catherine A Dowling
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Caitlyn Fisher
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Brittany Kirch
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA; Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Pyrimidine de novo synthesis inhibition selectively blocks effector but not memory T cell development. Nat Immunol 2023; 24:501-515. [PMID: 36797499 DOI: 10.1038/s41590-023-01436-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 01/13/2023] [Indexed: 02/18/2023]
Abstract
Blocking pyrimidine de novo synthesis by inhibiting dihydroorotate dehydrogenase is used to treat autoimmunity and prevent expansion of rapidly dividing cell populations including activated T cells. Here we show memory T cell precursors are resistant to pyrimidine starvation. Although the treatment effectively blocked effector T cells, the number, function and transcriptional profile of memory T cells and their precursors were unaffected. This effect occurred in a narrow time window in the early T cell expansion phase when developing effector, but not memory precursor, T cells are vulnerable to pyrimidine starvation. This vulnerability stems from a higher proliferative rate of early effector T cells as well as lower pyrimidine synthesis capacity when compared with memory precursors. This differential sensitivity is a drug-targetable checkpoint that efficiently diminishes effector T cells without affecting the memory compartment. This cell fate checkpoint might therefore lead to new methods to safely manipulate effector T cell responses.
Collapse
|
10
|
Ma X, Ma R, Zhang M, Qian B, Wang B, Yang W. Recent Progress in Multiple Sclerosis Treatment Using Immune Cells as Targets. Pharmaceutics 2023; 15:pharmaceutics15030728. [PMID: 36986586 PMCID: PMC10057470 DOI: 10.3390/pharmaceutics15030728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/02/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune-mediated demyelinating disease of the central nervous system. The main pathological features are inflammatory reaction, demyelination, axonal disintegration, reactive gliosis, etc. The etiology and pathogenesis of the disease have not been clarified. The initial studies believed that T cell-mediated cellular immunity is the key to the pathogenesis of MS. In recent years, more and more evidence has shown that B cells and their mediated humoral immune and innate immune cells (such as microglia, dendritic cells, macrophages, etc.) also play an important role in the pathogenesis of MS. This article mainly reviews the research progress of MS by targeting different immune cells and analyzes the action pathways of drugs. The types and mechanisms of immune cells related to the pathogenesis are introduced in detail, and the mechanisms of drugs targeting different immune cells are discussed in depth. This article aims to clarify the pathogenesis and immunotherapy pathway of MS, hoping to find new targets and strategies for the development of therapeutic drugs for MS.
Collapse
Affiliation(s)
- Xiaohong Ma
- Department of Neuroscience, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Rong Ma
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Mengzhe Zhang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Baicheng Qian
- Department of Neuroscience, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Baoliang Wang
- Department of Neuroscience, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
- Correspondence: (B.W.); (W.Y.)
| | - Weijing Yang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (B.W.); (W.Y.)
| |
Collapse
|
11
|
Kohle F, Dalakas MC, Lehmann HC. Repurposing MS immunotherapies for CIDP and other autoimmune neuropathies: unfulfilled promise or efficient strategy? Ther Adv Neurol Disord 2023; 16:17562864221137129. [PMID: 36620728 PMCID: PMC9810996 DOI: 10.1177/17562864221137129] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/19/2022] [Indexed: 01/03/2023] Open
Abstract
Despite advances in the treatment of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) and other common autoimmune neuropathies (AN), still-many patients with these diseases do not respond satisfactorily to the available treatments. Repurposing of disease-modifying therapies (DMTs) from other autoimmune conditions, particularly multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD), is a promising strategy that may accelerate the establishment of novel treatment choices for AN. This approach appears attractive due to homologies in the pathogenesis of these diseases and the extensive post-marketing experience that has been gathered from treating MS and NMOSD patients. The idea is also strengthened by a number of studies that explored the efficacy of DMTs in animal models of AN but also in some CIDP patients. We here review the available preclinical and clinical data of approved MS therapeutics in terms of their applicability to AN, especially CIDP. Promising therapeutic approaches appear to be B cell-directed and complement-targeting strategies, such as anti-CD20/anti-CD19 agents, Bruton's tyrosine kinase inhibitors and anti-C5 agents, as they exert their effects in the periphery. This is a major advantage because, in contrast to MS, their action in the periphery is sufficient to exert significant immunomodulation.
Collapse
Affiliation(s)
- Felix Kohle
- Department of Neurology, Faculty of Medicine,
University of Cologne and University Hospital Cologne, Cologne,
Germany
| | - Marinos C. Dalakas
- Department of Neurology, Thomas Jefferson
University, Philadelphia, PA, USA
- Neuroimmunology Unit, National and Kapodistrian
University of Athens Medical School, Athens, Greece
| | - Helmar C. Lehmann
- Department of Neurology, Faculty of Medicine,
University of Cologne and University Hospital Cologne, Kerpener Strasse, 62,
50937 Cologne, Germany
| |
Collapse
|
12
|
Khodadadi Z, Ektefa F, Naderi F, Fathi F. A computational evidence of the intermolecular hydrogen bonding in leflunomide: chemical shielding tensors. COMPUT THEOR CHEM 2023. [DOI: 10.1016/j.comptc.2023.114027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
13
|
Hyroššová P, Milošević M, Škoda J, Vachtenheim Jr J, Rohlena J, Rohlenová K. Effects of metabolic cancer therapy on tumor microenvironment. Front Oncol 2022; 12:1046630. [PMID: 36582801 PMCID: PMC9793001 DOI: 10.3389/fonc.2022.1046630] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Targeting tumor metabolism for cancer therapy is an old strategy. In fact, historically the first effective cancer therapeutics were directed at nucleotide metabolism. The spectrum of metabolic drugs considered in cancer increases rapidly - clinical trials are in progress for agents directed at glycolysis, oxidative phosphorylation, glutaminolysis and several others. These pathways are essential for cancer cell proliferation and redox homeostasis, but are also required, to various degrees, in other cell types present in the tumor microenvironment, including immune cells, endothelial cells and fibroblasts. How metabolism-targeted treatments impact these tumor-associated cell types is not fully understood, even though their response may co-determine the overall effectivity of therapy. Indeed, the metabolic dependencies of stromal cells have been overlooked for a long time. Therefore, it is important that metabolic therapy is considered in the context of tumor microenvironment, as understanding the metabolic vulnerabilities of both cancer and stromal cells can guide new treatment concepts and help better understand treatment resistance. In this review we discuss recent findings covering the impact of metabolic interventions on cellular components of the tumor microenvironment and their implications for metabolic cancer therapy.
Collapse
Affiliation(s)
- Petra Hyroššová
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Mirko Milošević
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Josef Škoda
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Jiří Vachtenheim Jr
- 3rd Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jakub Rohlena
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Kateřina Rohlenová
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
14
|
Peterson S, Jalil A, Beard K, Kakara M, Sriwastava S. Updates on efficacy and safety outcomes of new and emerging disease modifying therapies and stem cell therapy for Multiple Sclerosis: A review. Mult Scler Relat Disord 2022; 68:104125. [PMID: 36057173 DOI: 10.1016/j.msard.2022.104125] [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: 05/10/2022] [Revised: 08/08/2022] [Accepted: 08/16/2022] [Indexed: 12/15/2022]
Abstract
Multiple Sclerosis (MS) is a chronic neurodegenerative autoimmune disorder of the central nervous system (CNS) and the most common cause of serious physical disability in working-age adults. Drug development and research in this field have rapidly evolved over the past two decades, leading to the broad array of treatment options available today. These disease-modifying therapies (DMTs) work through distinct mechanisms of action and exhibit varying safety and efficacy profiles to help manage symptoms and reduce exacerbations in MS patients. Our extensive understanding of this condition has also led to novel approaches, such as the discovery of specific biomarkers that allow us to monitor the therapeutic response towards DMTs. The development of new DMTs continues to progress quickly today, and it can be difficult for clinicians to remain up to date on the most recent advancements and new treatment options for their patients. In this comprehensive review, we provide an outline of current MS medications in the pipeline including emerging DMTs and stem cell therapy, as well as the unique characteristics of these medications, including their indications, pharmacokinetic effects, and the relevant advancements.
Collapse
Affiliation(s)
- Sarah Peterson
- West Virginia University, School of Medicine, Morgantown, WV, USA; Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA; Department of Neurology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Amaris Jalil
- West Virginia University, School of Medicine, Morgantown, WV, USA; Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Katherine Beard
- West Virginia University, School of Medicine, Morgantown, WV, USA; Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Mihir Kakara
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, PA, USA; Depratment of Neurology, Wayne State University, Detroit, MI, USA
| | - Shitiz Sriwastava
- West Virginia University, School of Medicine, Morgantown, WV, USA; Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA; Depratment of Neurology, Wayne State University, Detroit, MI, USA; West Virginia Clinical and Translational Science Institute, Morgantown, WV, USA; Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, McGovern Medical School (UT Health), University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
15
|
Pontikos MA, Leija C, Zhao Z, Wang X, Kilgore J, Tornesi B, Adenmatten N, Phillips MA, Williams NS. Development of a biomarker to monitor target engagement after treatment with dihydroorotate dehydrogenase inhibitors. Biochem Pharmacol 2022; 204:115237. [PMID: 36055381 PMCID: PMC9547971 DOI: 10.1016/j.bcp.2022.115237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/22/2022]
Abstract
Dihydroorotate dehydrogenase (DHODH) catalyzes a key step in pyrimidine biosynthesis and has recently been validated as a therapeutic target for malaria through clinical studies on the triazolopyrimidine-based Plasmodium DHODH inhibitor DSM265. Selective toxicity towards Plasmodium species could be achieved because malaria parasites lack pyrimidine salvage pathways, and DSM265 selectively inhibits Plasmodium DHODH over the human enzyme. However, while DSM265 does not inhibit human DHODH, it inhibits DHODH from several preclinical species, including mice, suggesting that toxicity could result from on-target DHODH inhibition in those species. We describe here the use of dihydroorotate (DHO) as a biomarker of DHODH inhibition. Treatment of mammalian cells with DSM265 or the mammalian DHODH inhibitor teriflunomide led to increases in DHO where the extent of biomarker buildup correlated with both dose and inhibitor potency on DHODH. Treatment of mice with leflunomide (teriflunomide prodrug) caused a large dose-dependent buildup of DHO in blood (up to 16-fold) and urine (up to 5,400-fold) that was not observed for mice treated with DSM265. Unbound plasma teriflunomide levels reached 20-85-fold above the mouse DHODH IC50, while free DSM265 levels were only 1.6-4.2-fold above, barely achieving ∼ IC90 concentrations, suggesting that unbound DSM265 plasma levels are not sufficient to block the pathway in vivo. Thus, any toxicity associated with DSM265 treatment in mice is likely caused by off-target mechanisms. The identification of a robust biomarker for mammalian DHODH inhibition represents an important advance to generally monitor for on-target effects in preclinical and clinical applications of DHODH inhibitors used to treat human disease.
Collapse
Affiliation(s)
- Michael A Pontikos
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX 75390-9135, United States
| | - Christopher Leija
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX 75390-9135, United States
| | - Zhiyu Zhao
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX 75390, United States
| | - Xiaoyu Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX 75390-9135, United States
| | - Jessica Kilgore
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX 75390-9135, United States
| | - Belen Tornesi
- Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | | | - Margaret A Phillips
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX 75390-9135, United States.
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX 75390-9135, United States.
| |
Collapse
|
16
|
Nytrova P, Dolezal O. Sex bias in multiple sclerosis and neuromyelitis optica spectrum disorders: How it influences clinical course, MRI parameters and prognosis. Front Immunol 2022; 13:933415. [PMID: 36016923 PMCID: PMC9396644 DOI: 10.3389/fimmu.2022.933415] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
This review is a condensed summary of representative articles addressing the sex/gender bias in multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD). The strong effects of sex on the incidence and possibly also the activity and progression of these disorders should be implemented in the evaluation of any phase of clinical research and also in treatment choice consideration in clinical practice and evaluation of MRI parameters. Some relationships between clinical variables and gender still remain elusive but with further understanding of sex/gender-related differences, we should be able to provide appropriate patient-centered care and research.
Collapse
Affiliation(s)
- Petra Nytrova
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czechia
- *Correspondence: Petra Nytrova,
| | - Ondrej Dolezal
- Department of Neurology, Dumfries and Galloway Royal Infirmary, NHS Scotland, Dumfries, United Kingdom
| |
Collapse
|
17
|
Yamaoka S, Weisend CM, Swenson VA, Ebihara H. Development of accelerated high-throughput antiviral screening systems for emerging orthomyxoviruses. Antiviral Res 2022; 200:105291. [PMID: 35296419 PMCID: PMC9259280 DOI: 10.1016/j.antiviral.2022.105291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 11/19/2022]
Abstract
Bourbon virus (BRBV) is an emerging tick-borne orthomyxovirus that causes severe febrile illness in humans. There are no specific treatments for BRBV disease currently available. Here, we developed a highly accessible and robust, quantitative fluorescence-based BRBV minigenome (MG) system and applied it to high-throughput antiviral drug screening. We demonstrated that human dihydroorotate dehydrogenase (DHODH) inhibitors, hDHODH-IN-4 and brequinar, efficiently reduced BRBV RNA synthesis, and validated these findings using infectious BRBV in vitro. The DHODH inhibitors also exhibited high potency in inhibiting MG activities of other orthomyxoviruses with emerging zoonotic potential, including bat influenza A virus, swine influenza D virus, and Thogoto virus. Our newly developed MG system is a powerful platform for antiviral drug screening across the Orthomyxoviridae family, enabling rapid development and deployment of antivirals against future emerging orthomyxoviruses.
Collapse
Affiliation(s)
- Satoko Yamaoka
- Mayo Clinic, Department of Infectious Diseases, Rochester, MN, 55905, USA
| | - Carla M Weisend
- Mayo Clinic, Department of Infectious Diseases, Rochester, MN, 55905, USA; Mayo Clinic, Department of Molecular Medicine, Rochester, MN, 55905, USA
| | - Vaille A Swenson
- Mayo Clinic Graduate School of Biomedical Sciences, Virology and Gene Therapy Graduate Program, Rochester, MN, 55905, USA
| | - Hideki Ebihara
- National Institute of Infectious Diseases, Department of Virology I, Tokyo, 162-8640, Japan.
| |
Collapse
|
18
|
Au KM, Tisch R, Wang AZ. Immune Checkpoint Ligand Bioengineered Schwann Cells as Antigen-Specific Therapy for Experimental Autoimmune Encephalomyelitis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107392. [PMID: 34775659 PMCID: PMC8813901 DOI: 10.1002/adma.202107392] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/08/2021] [Indexed: 05/05/2023]
Abstract
Failure to establish immune tolerance leads to the development of autoimmune disease. The ability to regulate autoreactive T cells without inducing systemic immunosuppression represents a major challenge in the development of new strategies to treat autoimmune disease. Here, a translational method for bioengineering programmed death-ligand 1 (PD-L1)- and cluster of differentiation 86 (CD86)-functionalized mouse Schwann cells (SCs) to prevent and ameliorate multiple sclerosis (MS) in established mouse models of chronic and relapsing-remitting experimental autoimmune encephalomyelitis (EAE) is described. It is shown that the intravenous (i.v.) administration of immune checkpoint ligand functionalized mouse SCs modifies the course of disease and ameliorates EAE. Further, it is found that such bioengineered mouse SCs inhibit the differentiation of myelin-specific helper T cells into pathogenic T helper type-1 (Th 1) and type-17 (Th 17) cells, promote the development of tolerogenic myelin-specific regulatory T (Treg ) cells, and resolve inflammatory central nervous system microenvironments without inducing systemic immunosuppression.
Collapse
Affiliation(s)
- Kin Man Au
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75230, USA
| | - Roland Tisch
- Department of Microbiology and Immunology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75230, USA
| |
Collapse
|
19
|
Chen JA, Ma H, Liu Z, Tian J, Lu S, Fang W, Ze S, Lu W, Xie Q, Huang J, Wang Y. Discovery of Orally Available Retinoic Acid Receptor-Related Orphan Receptor γ-t/Dihydroorotate Dehydrogenase Dual Inhibitors for the Treatment of Refractory Inflammatory Bowel Disease. J Med Chem 2021; 65:592-615. [PMID: 34957834 DOI: 10.1021/acs.jmedchem.1c01746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel disease (IBD) is a multifactorial autoimmune disease, representing a major clinical challenge. Herein, a strategy of dual-targeting approach employing retinoic acid receptor-related orphan receptor γ-t (RORγt) and dihydroorotate dehydrogenase (DHODH) was proposed for the treatment of IBD. Dual RORγt/DHODH inhibitors are expected not only to reduce RORγt-driven Th17 cell differentiation but also to mitigate the expansion and activation of T cells, which may enhance anti-inflammatory effects. Starting from 2-aminobenzothiazole hit 1, a series of 2-aminotetrahydrobenzothiazoles were discovered as potent dual RORγt/DHODH inhibitors. Compound 14d stands out with IC50 values of 0.110 μM for RORγt and of 0.297 μM for DHODH. With acceptable mouse pharmacokinetic profiles, 14d exhibited remarkable in vivo anti-inflammatory activity and dose-dependently alleviated the severity of dextran sulfate sodium (DSS)-induced acute colitis in mice. Taken together, the present study provides a novel framework for the development of therapeutic agents for the treatment of IBD.
Collapse
Affiliation(s)
- Ji-An Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Hui Ma
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zehui Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jinlong Tian
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Sisi Lu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Wenqing Fang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Shuyin Ze
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.,Fudan Zhangjiang Institute, 666 Zhangheng Road, Shanghai 201203, China
| | - Jin Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yonghui Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
20
|
Lymphocyte Counts and Multiple Sclerosis Therapeutics: Between Mechanisms of Action and Treatment-Limiting Side Effects. Cells 2021; 10:cells10113177. [PMID: 34831400 PMCID: PMC8625745 DOI: 10.3390/cells10113177] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 01/18/2023] Open
Abstract
Although the detailed pathogenesis of multiple sclerosis (MS) is not completely understood, a broad range of disease-modifying therapies (DMTs) are available. A common side effect of nearly every MS therapeutic agent is lymphopenia, which can be both beneficial and, in some cases, treatment-limiting. A sound knowledge of the underlying mechanism of action of the selected agent is required in order to understand treatment-associated changes in white blood cell counts, as well as monitoring consequences. This review is a comprehensive summary of the currently available DMTs with regard to their effects on lymphocyte count. In the first part, we describe important general information about the role of lymphocytes in the course of MS and the essentials of lymphopenic states. In the second part, we introduce the different DMTs according to their underlying mechanism of action, summarizing recommendations for lymphocyte monitoring and definitions of lymphocyte thresholds for different therapeutic regimens.
Collapse
|
21
|
Barešić M, Reihl Crnogaj M, Zadro I, Anić B. Demyelinating disease (multiple sclerosis) in a patient with psoriatic arthritis treated with adalimumab: a case-based review. Rheumatol Int 2021; 41:2233-2239. [PMID: 34557936 DOI: 10.1007/s00296-021-04995-0] [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: 05/31/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
Over the past two decades, tumor necrosis factor-α (TNF-α) inhibitors became one of the most important drugs in the treatment of patients with psoriatic arthritis. Unfortunately, some of the patients exhibit unwanted side effects of the treatment. We describe a patient with psoriasis, psoriatic arthritis and uveitis who was treated with adalimumab and after 4 months of the treatment developed clinical and neuroradiological signs of demyelinating disease of the central nervous system. She experienced no signs and symptoms of neurological disease prior to adalimumab administration. After a detailed neurological work-up she was diagnosed with relapsing-remitting type of multiple sclerosis and treated with oral and pulse glucocorticoids and later with dimethyl fumarate. Adalimumab was discontinued. The question remains was the demyelination induced by the TNF-α blockade or was it unmasked by the introduction of the cytokine blocking agent. In patients suffering from inflammatory arthritis, treating disease to target as well as a close follow-up and knowledge of potential side effects of treatment remains crucial in good clinical practice.
Collapse
Affiliation(s)
- Marko Barešić
- Division of Clinical Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, University of Zagreb, Zagreb, Croatia.
| | - Mirna Reihl Crnogaj
- Division of Physical Medicine, Rehabilitation and Rheumatology, National Memorial Hospital Vukovar, Vukovar, Croatia
| | - Ivana Zadro
- Department of Neurology, School of Medicine, University Hospital Center Zagreb, University of Zagreb, Zagreb, Croatia
| | - Branimir Anić
- Division of Clinical Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
22
|
Oh J, Vukusic S, Tiel-Wilck K, Inshasi JS, Rog D, Baker DP, Pyatkevich Y, Poole EM, Vermersch P. Efficacy and Safety of Teriflunomide in Multiple Sclerosis across Age Groups: Analysis from Pooled Pivotal and Real-world Studies. J Cent Nerv Syst Dis 2021; 13:11795735211028781. [PMID: 34377047 PMCID: PMC8330455 DOI: 10.1177/11795735211028781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Evidence suggests that efficacy and safety of disease-modifying treatments for multiple sclerosis may differ with age. We evaluate efficacy and safety of teriflunomide across age subgroups of patients from pooled clinical trials and real-world studies. Methods: Post hoc analyses of patients who received teriflunomide 14 mg in the pooled phase II and III TEMSO, TOWER, TENERE, and TOPIC core and extension studies (n = 1978), and the real-world Teri-PRO (n = 928) and TAURUS-MS I (n = 1126) studies were conducted. Data were stratified by age at study entry: ⩽25, >25 to ⩽35, >35 to ⩽45, and >45 years. In Teri-PRO and TAURUS-MS I, an additional group, >55 years, was assessed. Results: In the pooled core studies, teriflunomide reduced annualized relapse rate (ARR) versus placebo across all ages. Unadjusted ARRs remained low across age groups in pooled extensions (0.18-0.30), Teri-PRO (0.10-0.35), and TAURUS-MS I (0.14-0.35). Baseline Expanded Disability Status Scale scores were higher with age, but stable through core and extension studies (mean increases over 7 years: ⩽25 years, +0.59; >25 to ⩽35 years, +0.46; >35 to ⩽45 years, +0.35; >45 years, +0.81). Across age groups, adverse event (AE) incidences were 78.4% to 90.7% in pooled core and extension studies and Teri-PRO, and 29.2% to 37.7% in TAURUS-MS I; serious AE incidences were ⩽21.3% in all studies. In pooled phase III and Teri-PRO studies, lymphocyte count decreases over 1 year after initiating teriflunomide, and proportions of patients developing lymphopenia, were small across age groups. Conclusions: Teriflunomide efficacy was demonstrated regardless of age. Safety was generally consistent across age groups.
Collapse
Affiliation(s)
- Jiwon Oh
- St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Sandra Vukusic
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-inflammation et Fondation Eugène Devic EDMUS Pour la Sclérose en Plaques, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Lyon-Bron, France.,Centre des Neurosciences de Lyon, INSERM 1028 et CNRS UMR5292, Observatoire Français de la Sclérose en Plaques, Lyon, France.,Université Claude Bernard Lyon 1, Villeurbanne, France
| | | | | | - David Rog
- Manchester Centre for Clinical Neurosciences, Salford, UK
| | | | - Yelena Pyatkevich
- Sanofi, Cambridge, MA, USA.,Takeda Pharmaceutical, Cambridge, MA, USA
| | | | - Patrick Vermersch
- Universite de Lille, Inserm U1172, CHU Lille, FHU Precise, Lille, France
| |
Collapse
|
23
|
Li M, Yan Y, Zhang X, Zhang Y, Xu X, Zhang L, Lu L, Wang J, Zhang Y, Song Q, Zhao C. Scaffold compound L971 exhibits anti-inflammatory activities through inhibition of JAK/STAT and NFκB signalling pathways. J Cell Mol Med 2021; 25:6333-6347. [PMID: 34018320 PMCID: PMC8256347 DOI: 10.1111/jcmm.16609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
JAK/STAT and NFκB signalling pathways play essential roles in regulating inflammatory responses, which are important pathogenic factors of various serious immune-related diseases, and function individually or synergistically. To find prodrugs that can treat inflammation, we performed a preliminary high-throughput screening of 18 840 small molecular compounds and identified scaffold compound L971 which significantly inhibited JAK/STAT and NFκB driven luciferase activities. L971 could inhibit the constitutive and stimuli-dependent activation of STAT1, STAT3 and IκBα and could significantly down-regulate the proinflammatory gene expression in mouse peritoneal macrophages stimulated by LPS. Gene expression profiles upon L971 treatment were determined using high-throughput RNA sequencing, and significant differentially up-regulated and down-regulated genes were identified by DESeq analysis. The bioinformatic studies confirmed the anti-inflammatory effects of L971. Finally, L971 anti-inflammatory character was further verified in LPS-induced sepsis shock mouse model in vivo. Taken together, these data indicated that L971 could down-regulate both JAK/STAT and NFκB signalling activities and has the potential to treat inflammatory diseases such as sepsis shock.
Collapse
Affiliation(s)
- Mengyuan Li
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Yu Yan
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Xinxin Zhang
- Innovation Platform of Marine Drug Screening & EvaluationQingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| | - Yidan Zhang
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Xiaohan Xu
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Lei Zhang
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Liangliang Lu
- School of Life ScienceLanzhou UniversityLanzhouChina
| | - Jie Wang
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Yazhuo Zhang
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Qiaoling Song
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
- Innovation Platform of Marine Drug Screening & EvaluationQingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| | - Chenyang Zhao
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
- Innovation Platform of Marine Drug Screening & EvaluationQingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| |
Collapse
|
24
|
Shields MD, Skelton WP, Laber DA, Verbosky M, Ashraf N. A Novel Case of Leflunomide-Induced Thrombotic Thrombocytopenic Purpura. J Hematol 2021; 10:139-142. [PMID: 34267852 PMCID: PMC8256916 DOI: 10.14740/jh837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 04/23/2021] [Indexed: 02/01/2023] Open
Abstract
Leflunomide has not been previously associated with thrombotic thrombocytopenic purpura (TTP), a rare life-threatening clinical syndrome characterized by thrombotic microangiopathy (TMA) due to inability to cleave ADAMTS13. Here, we present the first case of leflunomide-induced TTP. Our patient developed encephalopathy, thrombocytopenia, anemia and hyperbilirubinemia 2 months after starting leflunomide. Schistocytes were noted on peripheral smear and ADAMTS13 activity was low (< 5%), consistent with acquired TTP. He received therapeutic plasma exchange, corticosteroids, rituximab and caplacizumab with normalization of hemolysis labs and ADAMTS13 activity. However, pancytopenia persisted, raising the suspicion for leflunomide toxicity. Oral cholestyramine treatment was empirically started before teriflunomide (a leflunomide metabolite) level was found to be elevated. Blood counts normalized after cholestyramine and have remained normal at last follow-up over a year later. This is the first reported case of TTP precipitated by leflunomide. Our case highlights the importance of recognizing drugs as an etiology of TMA and adds leflunomide to this list.
Collapse
Affiliation(s)
- Misty Dawn Shields
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Morsani School of Medicine, University of South Florida, Tampa, FL, USA
| | - William Paul Skelton
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Morsani School of Medicine, University of South Florida, Tampa, FL, USA
| | - Damian A Laber
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Morsani School of Medicine, University of South Florida, Tampa, FL, USA
| | - Michael Verbosky
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Noman Ashraf
- Morsani School of Medicine, University of South Florida, Tampa, FL, USA.,Department of Hematology Oncology, James A. Haley Veterans Hospital, Tampa, FL, USA
| |
Collapse
|
25
|
Microwave-assisted synthesis of (3,5-disubstituted isoxazole)-linked benzimidazolone derivatives: DFT calculations and biological activities. MONATSHEFTE FUR CHEMIE 2021. [DOI: 10.1007/s00706-021-02764-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
26
|
Ariav Y, Ch'ng JH, Christofk HR, Ron-Harel N, Erez A. Targeting nucleotide metabolism as the nexus of viral infections, cancer, and the immune response. SCIENCE ADVANCES 2021; 7:eabg6165. [PMID: 34138729 PMCID: PMC8133749 DOI: 10.1126/sciadv.abg6165] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/29/2021] [Indexed: 05/11/2023]
Abstract
Virus-infected cells and cancers share metabolic commonalities that stem from their insatiable need to replicate while evading the host immune system. These similarities include hijacking signaling mechanisms that induce metabolic rewiring in the host to up-regulate nucleotide metabolism and, in parallel, suppress the immune response. In both cancer and viral infections, the host immune cells and, specifically, lymphocytes augment nucleotide synthesis to support their own proliferation and effector functions. Consequently, established treatment modalities targeting nucleotide metabolism against cancers and virally infected cells may result in restricted immune response. Encouragingly, following the introduction of immunotherapy against cancers, multiple studies improved our understanding for improving antigen presentation to the immune system. We propose here that understanding the immune consequences of targeting nucleotide metabolism against cancers may be harnessed to optimize therapy against viral infections.
Collapse
Affiliation(s)
- Yarden Ariav
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - James H Ch'ng
- Department of Pediatrics, Division of Hematology/Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Heather R Christofk
- Department of Biological Chemistry, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Noga Ron-Harel
- Department of Biology, Technion, Israel Institute of Technology, Haifa, Israel.
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
27
|
Alamri RD, Elmeligy MA, Albalawi GA, Alquayr SM, Alsubhi SS, El-Ghaiesh SH. Leflunomide an immunomodulator with antineoplastic and antiviral potentials but drug-induced liver injury: A comprehensive review. Int Immunopharmacol 2021; 93:107398. [PMID: 33571819 PMCID: PMC7869628 DOI: 10.1016/j.intimp.2021.107398] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Abstract
Leflunomide (LF) represents the prototype member of dihydroorotate dehydrogenase (DHODH) enzyme inhibitors. DHODH is a mitochondrial inner membrane enzyme responsible for catalytic conversion of dihydroorotate into orotate, a rate-limiting step in the de novo synthesis of the pyrimidine nucleotides. LF produces cellular depletion of pyrimidine nucleotides required for cell growth and proliferation. Based on the affected cells the outcome can be attainable as immunosuppression, antiproliferative, and/or the recently gained attention of the antiviral potentials of LF and its new congeners. Also, protein tyrosine kinase inhibition is an additional mechanistic benefit of LF, which inhibits immunological events such as cellular expansion and immunoglobulin production with an enhanced release of immunosuppressant cytokines. LF is approved for the treatment of autoimmune arthritis of rheumatoid and psoriatic pathogenesis. Also, LF has been used off-label for the treatment of relapsing-remitting multiple sclerosis. However, LF antiviral activity is repurposed and under investigation with related compounds under a phase-I trial as a SARS CoV-2 antiviral in cases with COVID-19. Despite success in improving patients' mobility and reducing joint destruction, reported events of LF-induced liver injury necessitated regulatory precautions. LF should not be used in patients with hepatic impairment or in combination with drugs elaborating a burden on the liver without regular monitoring of liver enzymes and serum bilirubin as safety biomarkers. This study aims to review the pharmacological and safety profile of LF with a focus on the LF-induced hepatic injury from the perspective of pathophysiology and possible protective agents.
Collapse
Affiliation(s)
- Raghad D Alamri
- Faculty of Medicine, University of Tabuk, Tabuk 47713, Saudi Arabia
| | | | | | - Sarah M Alquayr
- Faculty of Medicine, University of Tabuk, Tabuk 47713, Saudi Arabia
| | | | - Sabah H El-Ghaiesh
- Deaprtment of Pharmacology, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk 47713, Saudi Arabia.
| |
Collapse
|
28
|
Liu Q, Gupta A, Okesli-Armlovich A, Qiao W, Fischer CR, Smith M, Carette JE, Bassik MC, Khosla C. Enhancing the Antiviral Efficacy of RNA-Dependent RNA Polymerase Inhibition by Combination with Modulators of Pyrimidine Metabolism. Cell Chem Biol 2020; 27:668-677.e9. [PMID: 32442424 PMCID: PMC7241336 DOI: 10.1016/j.chembiol.2020.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/12/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022]
Abstract
Genome-wide analysis of the mode of action of GSK983, a potent antiviral agent, led to the identification of dihydroorotate dehydrogenase as its target along with the discovery that genetic knockdown of pyrimidine salvage sensitized cells to GSK983. Because GSK983 is an ineffective antiviral in the presence of physiological uridine concentrations, we explored combining GSK983 with pyrimidine salvage inhibitors. We synthesized and evaluated analogs of cyclopentenyl uracil (CPU), an inhibitor of uridine salvage. We found that CPU was converted into its triphosphate in cells. When combined with GSK983, CPU resulted in large drops in cellular UTP and CTP pools. Consequently, CPU-GSK983 suppressed dengue virus replication in the presence of physiological concentrations of uridine. In addition, the CPU-GSK983 combination markedly enhanced the effect of RNA-dependent RNA polymerase (RdRp) inhibition on viral infection. Our findings highlight a new host-targeting strategy for potentiating the antiviral activity of RdRp inhibitors.
Collapse
Affiliation(s)
- Qi Liu
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Amita Gupta
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Ayse Okesli-Armlovich
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Wenjie Qiao
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Curt R Fischer
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Mark Smith
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Jan E Carette
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Michael C Bassik
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
29
|
Muehler A, Peelen E, Kohlhof H, Gröppel M, Vitt D. Vidofludimus calcium, a next generation DHODH inhibitor for the Treatment of relapsing-remitting multiple sclerosis. Mult Scler Relat Disord 2020; 43:102129. [PMID: 32428844 DOI: 10.1016/j.msard.2020.102129] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/10/2020] [Accepted: 04/12/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND Inhibition of dihydroorotate dehydrogenase (DHODH) is an established mechanism for the treatment of relapsing-remitting multiple sclerosis (RRMS). Currently approved treatments have several shortcomings. Consequently, new and effective treatments with improved safety and convenience profiles are sought after by patients. OBJECTIVE To explore the overall profile of vidofludimus for the treatment of RRMS. METHODS Preclinical investigations were done exploring the species-dependency of DHODH inhibition of vidofludimus. In addition, the preclinical efficacy in a rat experimental autoimmune encephalomyelitis (EAE) model and the inhibition of cytokine release from activated PBMC were investigated. Pharmacokinetic data were also obtained in a Phase 1 multiple ascending dose trial of the formulation IMU-838 (vidofludimus calcium). RESULTS It was shown that vidofludimus is 2.6 times more potent in inhibiting DHO oxidation by human DHODH compared to teriflunomide. Although both compounds increased cell apoptosis, vidofludimus was more efficacious in the inhibition of T-lymphocyte proliferation compared to teriflunomide. The same was also observed for the secretion of IL-17 and IFN-γ. Interestingly, the potency or vidofludimus to inhibit rat or mouse DHODH is 7.5 and 64.4 time lower than the for the human DHODH, respectively. The rat EAE study clearly exhibited a dose-dependent inhibition of cumulative disease scores by vidofludimus. In the multiple ascending dose Phase 1 clinical trial, the serum half-life of about 30 h provides a favorable profile for once daily dosing of IMU-838, with quick dosing to steady state through levels within 5 days and the ability to wash out drug quickly, if required. CONCLUSIONS The investigations highlighted that the desired selective immunomodulatory properties can be separated from general antiproliferative effects seen and related adverse events in first-generation DHODH inhibitors. Based on data obtained from a series of pre-clinical as well as phase 1 and phase 2 studies, IMU-838 is a promising next-generation candidate for the oral treatment of RRMS. However, this will need to be confirmed in the currently ongoing Phase 2 study in RRMS patients.
Collapse
Affiliation(s)
- Andreas Muehler
- Immunic AG, Am Klopferspitz 19, 82152 Planegg-Martinsried, Germany.
| | - Evelyn Peelen
- Immunic AG, Am Klopferspitz 19, 82152 Planegg-Martinsried, Germany
| | - Hella Kohlhof
- Immunic AG, Am Klopferspitz 19, 82152 Planegg-Martinsried, Germany
| | - Manfred Gröppel
- Immunic AG, Am Klopferspitz 19, 82152 Planegg-Martinsried, Germany
| | - Daniel Vitt
- Immunic AG, Am Klopferspitz 19, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
30
|
Serra MC, Accardi CJ, Ma C, Park Y, Tran V, Jones DP, Hafer-Macko CE, Ryan AS. Metabolomics of Aerobic Exercise in Chronic Stroke Survivors: A Pilot Study. J Stroke Cerebrovasc Dis 2019; 28:104453. [PMID: 31668688 DOI: 10.1016/j.jstrokecerebrovasdis.2019.104453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/09/2019] [Accepted: 09/27/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Understanding the metabolic response to exercise may aid in optimizing stroke management. Therefore, the purpose of this pilot study was to evaluate plasma metabolomic profiles in chronic stroke survivors following aerobic exercise training. METHODS Participants (age: 62 ± 1 years, body mass index: 31 ± 1 kg/m2, mean ± standard error of the mean) were randomized to 6 months of treadmill exercise (N = 17) or whole-body stretching (N = 8) with preintervention and postintervention measurement of aerobic capacity (VO2peak). Linear models for microarray data expression analysis was performed to determine metabolic changes over time, and Mummichog was used for pathway enrichment analysis following analysis of plasma samples by high-performance liquid chromatography coupled to ultrahigh resolution mass spectrometry. RESULTS VO2peak change was greater following exercise than stretching (18.9% versus -.2%; P < .01). Pathway enrichment analysis of differentially expressed metabolites results showed significant enrichment in 4 pathways following treadmill exercise, 3 of which (heparan-, chondroitin-, keratan-sulfate degradation) involved connective tissue metabolism and the fourth involve lipid signaling (linoleate metabolism). More pathways were altered in pre and post comparisons of stretching, including branched-chain amino acid, tryptophan, tyrosine, and urea cycle, which could indicate loss of lean body mass. CONCLUSIONS These preliminary data show different metabolic changes due to treadmill training and stretching in chronic stroke survivors and suggest that in addition to improved aerobic capacity, weight-bearing activity, like walking, could protect against loss of lean body mass. Future studies are needed to examine the relationship between changes in metabolomic profiles to reductions in cardiometabolic risk after treadmill rehabilitation.
Collapse
Affiliation(s)
- Monica C Serra
- San Antonio GRECC, South Texas VA and the Division of Geriatrics, Gerontology & Palliative Medicine and the Sam & Ann Barshop Institute for Longevity & Aging Studies, UT Health San Antonio, San Antonio, Texas.
| | - Carolyn J Accardi
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Chunyu Ma
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Younja Park
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, Atlanta, Georgia; College of Pharmacy, Korea University, Sejong City, Korea
| | - ViLinh Tran
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Charlene E Hafer-Macko
- Baltimore VA Research Service and GRECC and the Division of Gerontology and Geriatric Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Alice S Ryan
- Baltimore VA Research Service and GRECC and the Division of Gerontology and Geriatric Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
31
|
Chisari CG, Toscano S, D’Amico E, Lo Fermo S, Zanghì A, Arena S, Zappia M, Patti F. An update on the safety of treating relapsing-remitting multiple sclerosis. Expert Opin Drug Saf 2019; 18:925-948. [DOI: 10.1080/14740338.2019.1658741] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Clara G. Chisari
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Simona Toscano
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Emanuele D’Amico
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Salvatore Lo Fermo
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Aurora Zanghì
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Sebastiano Arena
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Mario Zappia
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Francesco Patti
- Department “GF Ingrassia”, Section of Neurosciences, Multiple Sclerosis Center, University of Catania, Catania, Italy
| |
Collapse
|
32
|
Turki T, Taguchi YH. Machine learning algorithms for predicting drugs–tissues relationships. EXPERT SYSTEMS WITH APPLICATIONS 2019. [DOI: 10.1016/j.eswa.2019.02.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
33
|
Kim YJ, Cubitt B, Chen E, Hull MV, Chatterjee AK, Cai Y, Kuhn JH, de la Torre JC. The ReFRAME library as a comprehensive drug repurposing library to identify mammarenavirus inhibitors. Antiviral Res 2019; 169:104558. [PMID: 31302150 DOI: 10.1016/j.antiviral.2019.104558] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 01/05/2023]
Abstract
Several mammarenaviruses, chiefly Lassa virus (LASV) in Western Africa and Junín virus (JUNV) in the Argentine Pampas, cause severe disease in humans and pose important public health problems in their endemic regions. Moreover, mounting evidence indicates that the worldwide-distributed mammarenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen of clinical significance. The lack of licensed mammarenavirus vaccines and partial efficacy of current anti-mammarenavirus therapy limited to an off-label use of the nucleoside analog ribavirin underscore an unmet need for novel therapeutics to combat human pathogenic mammarenavirus infections. This task can be facilitated by the implementation of "drug repurposing" strategies to reduce the time and resources required to advance identified antiviral drug candidates into the clinic. We screened a drug repurposing library of 11,968 compounds (Repurposing, Focused Rescue and Accelerated Medchem [ReFRAME]) and identified several potent inhibitors of LCMV multiplication that had also strong anti-viral activity against LASV and JUNV. Our findings indicate that enzymes of the rate-limiting steps of pyrimidine and purine biosynthesis, the pro-viral MCL1 apoptosis regulator, BCL2 family member protein and the mitochondrial electron transport complex III, play critical roles in the completion of the mammarenavirus life cycle, suggesting they represent potential druggable targets to counter human pathogenic mammarenavirus infections.
Collapse
Affiliation(s)
- Yu-Jin Kim
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Beatrice Cubitt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Emily Chen
- California Institute for Biomedical Research, La Jolla, CA, 92037, USA
| | - Mitchell V Hull
- California Institute for Biomedical Research, La Jolla, CA, 92037, USA
| | | | - Yingyun Cai
- Integrated Research Facility at Fort Detrick (IRF-Frederick), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD, 21702, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick (IRF-Frederick), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD, 21702, USA
| | - Juan C de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
34
|
Rommer PS, Milo R, Han MH, Satyanarayan S, Sellner J, Hauer L, Illes Z, Warnke C, Laurent S, Weber MS, Zhang Y, Stuve O. Immunological Aspects of Approved MS Therapeutics. Front Immunol 2019; 10:1564. [PMID: 31354720 PMCID: PMC6637731 DOI: 10.3389/fimmu.2019.01564] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 06/24/2019] [Indexed: 12/21/2022] Open
Abstract
Multiple sclerosis (MS) is the most common neurological immune-mediated disease leading to disability in young adults. The outcome of the disease is unpredictable, and over time, neurological disabilities accumulate. Interferon beta-1b was the first drug to be approved in the 1990s for relapsing-remitting MS to modulate the course of the disease. Over the past two decades, the treatment landscape has changed tremendously. Currently, more than a dozen drugs representing 1 substances with different mechanisms of action have been approved (interferon beta preparations, glatiramer acetate, fingolimod, siponimod, mitoxantrone, teriflunomide, dimethyl fumarate, cladribine, alemtuzumab, ocrelizumab, and natalizumab). Ocrelizumab was the first medication to be approved for primary progressive MS. The objective of this review is to present the modes of action of these drugs and their effects on the immunopathogenesis of MS. Each agent's clinical development and potential side effects are discussed.
Collapse
Affiliation(s)
- Paulus S. Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Ron Milo
- Department of Neurology, Barzilai University Medical Center, Ashkelon, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - May H. Han
- Neuroimmunology Division, Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Sammita Satyanarayan
- Neuroimmunology Division, Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Johann Sellner
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
- Department of Neurology, Klinikum Rechts der Isar, Technische Universität, Munich, Germany
| | - Larissa Hauer
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Clemens Warnke
- Department of Neurology, Medical Faculty, University of Köln, Cologne, Germany
| | - Sarah Laurent
- Department of Neurology, Medical Faculty, University of Köln, Cologne, Germany
| | - Martin S. Weber
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - Yinan Zhang
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, Klinikum Rechts der Isar, Technische Universität, Munich, Germany
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, VA North Texas Health Care System, Medical Service Dallas, VA Medical Center, Dallas, TX, United States
| |
Collapse
|
35
|
|
36
|
Gregson A, Thompson K, Tsirka SE, Selwood DL. Emerging small-molecule treatments for multiple sclerosis: focus on B cells. F1000Res 2019; 8:F1000 Faculty Rev-245. [PMID: 30863536 PMCID: PMC6402079 DOI: 10.12688/f1000research.16495.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2019] [Indexed: 12/27/2022] Open
Abstract
Multiple sclerosis (MS) is a major cause of disability in young adults. Following an unknown trigger (or triggers), the immune system attacks the myelin sheath surrounding axons, leading to progressive nerve cell death. Antibodies and small-molecule drugs directed against B cells have demonstrated good efficacy in slowing progression of the disease. This review focusses on small-molecule drugs that can affect B-cell biology and may have utility in disease management. The risk genes for MS are examined from the drug target perspective. Existing small-molecule therapies for MS with B-cell actions together with new drugs in development are described. The potential for experimental molecules with B-cell effects is also considered. Small molecules can have diverse actions on B cells and be cytotoxic, anti-inflammatory and anti-viral. The current B cell-directed therapies often kill B-cell subsets, which can be effective but lead to side effects and toxicity. A deeper understanding of B-cell biology and the effect on MS disease should lead to new drugs with better selectivity, efficacy, and an improved safety profile. Small-molecule drugs, once the patent term has expired, provide a uniquely sustainable form of healthcare.
Collapse
Affiliation(s)
- Aaron Gregson
- The Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| | - Kaitlyn Thompson
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, 11794, USA
| | - Stella E Tsirka
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, 11794, USA
| | - David L Selwood
- The Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
37
|
Fakan B, Szalardy L, Vecsei L. Exploiting the Therapeutic Potential of Endogenous Immunomodulatory Systems in Multiple Sclerosis-Special Focus on the Peroxisome Proliferator-Activated Receptors (PPARs) and the Kynurenines. Int J Mol Sci 2019; 20:ijms20020426. [PMID: 30669473 PMCID: PMC6358998 DOI: 10.3390/ijms20020426] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/13/2019] [Accepted: 01/15/2019] [Indexed: 01/20/2023] Open
Abstract
Multiple sclerosis (MS) is a progressive neurodegenerative disease, characterized by autoimmune central nervous system (CNS) demyelination attributable to a disturbed balance between encephalitic T helper 1 (Th1) and T helper 17 (Th17) and immunomodulatory regulatory T cell (Treg) and T helper 2 (Th2) cells, and an alternatively activated macrophage (M2) excess. Endogenous molecular systems regulating these inflammatory processes have recently been investigated to identify molecules that can potentially influence the course of the disease. These include the peroxisome proliferator-activated receptors (PPARs), PPARγ coactivator-1alpha (PGC-1α), and kynurenine pathway metabolites. Although all PPARs ameliorate experimental autoimmune encephalomyelitis (EAE), recent evidence suggests that PPARα, PPARβ/δ agonists have less pronounced immunomodulatory effects and, along with PGC-1α, are not biomarkers of neuroinflammation in contrast to PPARγ. Small clinical trials with PPARγ agonists have been published with positive results. Proposed as immunomodulatory and neuroprotective, the therapeutic use of PGC-1α activation needs to be assessed in EAE/MS. The activation of indolamine 2,3-dioxygenase (IDO), the rate-limiting step of the kynurenine pathway of tryptophan (Trp) metabolism, plays crucial immunomodulatory roles. Indeed, Trp metabolites have therapeutic relevance in EAE and drugs with structural analogy to kynurenines, such as teriflunomide, are already approved for MS. Further studies are required to gain deeper knowledge of such endogenous immunomodulatory pathways with potential therapeutic implications in MS.
Collapse
Affiliation(s)
- Bernadett Fakan
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, H-6725 Szeged, Semmelweis u. 6, Hungary.
| | - Levente Szalardy
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, H-6725 Szeged, Semmelweis u. 6, Hungary.
| | - Laszlo Vecsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, H-6725 Szeged, Semmelweis u. 6, Hungary.
- MTA-SZTE Neuroscience Research Group, H-6725 Szeged, Semmelweis u. 6, Hungary.
| |
Collapse
|
38
|
|