1
|
Balic N, Nikolac Perkovic M, Milos T, Vuic B, Kurtovic Kodzoman M, Svob Strac D, Nedic Erjavec G. Extracellular vesicles as a promising tool in neuropsychiatric pharmacotherapy application and monitoring. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111393. [PMID: 40340017 DOI: 10.1016/j.pnpbp.2025.111393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
This review deals with the application of extracellular vesicles (EVs) in the treatment of various neuropsychiatric disorders, including mood disorders, neurodegeneration, psychosis, neurological insults and injuries, epilepsy and substance use disorders. The main challenges of most neuropsychiatric pharmaceuticals nowadays are how to reach the central nervous system at therapeutic concentration and maintain it long enough and how to avoid undesirable side effects caused by unsatisfying toxicity. Extracellular vesicles, as very important mediators of intercellular communication, can have a variety of therapeutic qualities. They can act neuroprotective, regenerative and anti-inflammatory, but they also have characteristics of a good drug delivery system, including their nano- scale size, biological safety and abilities to cross BBB, to pack drugs within the lipid bilayer, and not to trigger an immunological response. Besides, due to their presence in readily accessible biofluids, they are good candidates for biomarkers of the disease, its progression and therapy response monitoring. Alternations in EVs' cargo profiles can reflect the pathogenesis of neuropsychiatric disorders, but they could also affect the disease outcomes. In the future, EVs could help physicians to tailor treatment strategies for individual patients, however, more extensive studies are needed to standardize isolation, purification and production procedures, increase efficacy of drug loading and limit unwanted effects of innate EVs' content.
Collapse
Affiliation(s)
- Nikola Balic
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | | | - Tina Milos
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | - Barbara Vuic
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | | | | | | |
Collapse
|
2
|
Hajjar J, Rehman A, Hamdi A, Fuss I. Navigating the Complexities of Common Variable Immunodeficiency Enteropathy: From Established Therapies to Emerging Interventions. Immunol Allergy Clin North Am 2025; 45:267-285. [PMID: 40287172 DOI: 10.1016/j.iac.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Common Variable Immunodeficiency (CVID) is a prevalent primary immunodeficiency in adults, marked by low immunoglobulin levels and recurrent infections. This review examines the gastrointestinal complications of CVID, including both infectious and non-infectious manifestations. It highlights therapeutic strategies, from antimicrobials to novel biologics, and the role of immune modulation. The review also explores the impact of gut microbiota dysbiosis on CVID pathogenesis and emphasizes the need for personalized treatment approaches and routine cancer screening due to the elevated risk of gastrointestinal malignancy in CVID patients.
Collapse
Affiliation(s)
- Joud Hajjar
- The William T Shearer Center for Human Immunobiology at Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | - Ahmed Rehman
- Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ahmed Hamdi
- Department of Medicine, Section of Infectious Disease, Baylor College of Medicine, One Baylor Plaza, Building Tower West McNair Campus (MCHA) A10.143 MS: BCM901, Houston, TX 77030, USA
| | - Ivan Fuss
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, 31 Center Dr Ste 7A03, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Pickett JR, Wu Y, Ta HT. VCAM-1 as a common biomarker in inflammatory bowel disease and colorectal cancer: unveiling the dual anti-inflammatory and anti-cancer capacities of anti-VCAM-1 therapies. Cancer Metastasis Rev 2025; 44:40. [PMID: 40095109 PMCID: PMC11913972 DOI: 10.1007/s10555-025-10258-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Vascular cell adhesion molecule (VCAM)-1 has garnered significant research attention due to its potential as a disease biomarker and drug target across several inflammatory pathologies-including atherosclerosis, asthma, rheumatoid arthritis, and inflammatory bowel disease (IBD). The VCAM-1 protein has also been noted for its functional involvement in cancer metastasis and drug resistance to conventional chemotherapeutics. Although the anti-inflammatory and anti-cancer facets of VCAM-1 antagonisation have been examined separately, there is yet to be a review that explicitly addresses the functional interrelationship between these mechanisms. Furthermore, the pleiotropic mechanisms of anti-VCAM-1 therapies may present a useful paradigm for designing drug candidates with synergistic anti-inflammatory and anti-tumorigenic effects. The pathological overlap between inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CRC) serves as the quintessential disease model to observe this therapeutic duality. This review thereby details the adhesive mechanisms of VCAM-1 in colorectal disease-specifically, driving immune cell infiltration during IBD and tumour cell metastasis in CRC-and posits the potential of this receptor as a common drug target for both diseases. To explore this hypothesis, the current progress of novel VCAM-1-directed drug candidates in experimental models of IBD and CRC is also discussed.
Collapse
Affiliation(s)
- Jessica R Pickett
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia
| | - Yuao Wu
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia
| | - Hang Thu Ta
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia.
| |
Collapse
|
4
|
Rida Zainab S, Zeb Khan J, Khalid Tipu M, Jahan F, Irshad N. A review on multiple sclerosis: Unravelling the complexities of pathogenesis, progression, mechanisms and therapeutic innovations. Neuroscience 2025; 567:133-149. [PMID: 39709058 DOI: 10.1016/j.neuroscience.2024.12.029] [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: 10/24/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory demyelinating disorder of the central nervous system (CNS) targeting myelinated axons. Pathogenesis of MS entails an intricate genetic, environmental, and immunological interaction. Dysregulation of immune response i.e. autoreactive T & B-Cells and macrophage infiltration into the CNS leads to inflammation, demyelination, and neurodegeneration. Disease progression of MS varies among individuals transitioning from one form of relapsing-remitting to secondary progressive MS (SPMS). Research advances have unfolded various molecular targets involved in MS from oxidative stress to blood-brain barrier (BBB) disruption. Different pathways are being targeted so far such as inflammatory and cytokine signaling pathways to overcome disease progression. Therapeutic innovations have significantly transformed the management of MS, especially the use of disease-modifying therapies (DMTs) to reduce relapse rates and control disease progression. Advancements in research, neuroprotective strategies, and remyelination strategies hold promising results in reversing CNS damage. Various mice models are being adopted for testing new entities in MS research.
Collapse
Affiliation(s)
- Syeda Rida Zainab
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Jehan Zeb Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Muhammad Khalid Tipu
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Faryal Jahan
- Shifa College of Pharmaceutical Sciences, STMU, Islamabad, Pakistan.
| | - Nadeem Irshad
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
5
|
Cardle II, Raman J, Nguyen DC, Wang T, Wu AY, Sellers DL, Pichon TJ, Cheng EL, Kacherovsky N, Salipante SJ, Jensen MC, Pun SH. DNA Aptamer-Polymer Conjugates for Selective Targeting of Integrin α4β1 + T-Lineage Cancers. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4543-4561. [PMID: 39788927 PMCID: PMC11995848 DOI: 10.1021/acsami.4c17788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Selective therapeutic targeting of T-cell malignancies is difficult due to the shared lineage between healthy and malignant T cells. Current front-line chemotherapy for these cancers is largely nonspecific, resulting in frequent cases of relapsed/refractory disease. The development of targeting approaches for effectively treating T-cell leukemia and lymphoma thus remains a critical goal for the oncology field. Here, we report the discovery of a DNA aptamer, named HR7A1, that displays low nanomolar affinity for the integrin α4β1 (VLA-4), a marker associated with chemoresistance and relapse in leukemia patients. After truncation of HR7A1 to a minimal binding motif, we demonstrate elevated binding of the aptamer to T-lineage cancer cells over healthy immune cells. Using cryo-EM and competition studies, we find that HR7A1 shares an overlapping binding site on α4β1 with fibronectin and VCAM-1, which has implications for sensitizing blood cancers to chemotherapy. We last characterize barriers to in vivo aptamer translation, including serum stability, temperature-sensitive binding, and short circulation half-life, and synthesize an aptamer-polymer conjugate that addresses these challenges. Future work will seek to validate in vivo targeting of α4β1+ tumors with the conjugate, establishing an aptamer-based biomaterial that can be readily adapted for targeted treatment of T-cell malignancies.
Collapse
Affiliation(s)
- Ian I. Cardle
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Seattle Children’s Therapeutics, Seattle, WA 98101, USA
| | - Jai Raman
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Dinh Chuong Nguyen
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Tong Wang
- Nanoscience Initiative, CUNY Advanced Science Research Center, City University of New York, NY 10031, USA
| | - Abe Y. Wu
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Trey J. Pichon
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Emmeline L. Cheng
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Nataly Kacherovsky
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Michael C. Jensen
- Seattle Children’s Therapeutics, Seattle, WA 98101, USA
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
6
|
Mansouri P, Mansouri P, Behmard E, Najafipour S, Kouhpayeh A, Farjadfar A. Novel targets for mucosal healing in inflammatory bowel disease therapy. Int Immunopharmacol 2025; 144:113544. [PMID: 39571265 DOI: 10.1016/j.intimp.2024.113544] [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: 06/01/2024] [Revised: 10/13/2024] [Accepted: 10/28/2024] [Indexed: 12/15/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic condition affecting the gastrointestinal tract, primarily manifesting as ulcerative colitis (UC) or Crohn's disease (CD). Both inflammation and disruption of the intestinal epithelial barrier are key factors in IBD pathogenesis. Substantial evidence has revealed a significant association between aberrant immune responses and impairment of the intestinal epithelial barrier in IBD pathogenesis. The components of the intestinal epithelium, particularly goblet cells and Paneth cells, are crucial to gut homeostasis, as they secrete mucin, antimicrobial peptides (AMPs), and cytokines. Furthermore, impairment of epithelial integrity, which is regulated by tight junctions, is a hallmark of IBD pathology. While common treatments for IBD, such as anti-inflammatory drugs, target various signaling pathways with varying efficacies, therapeutic approaches focused on mucosal and epithelial barrier healing have been largely neglected. Moreover, high costs, side effects, and insufficient or inconsistent therapeutic outcomes remain major drawbacks of conventional anti-IBD drugs. Recent studies on epithelial barrier regeneration and permeability reduction have introduced promising therapeutic targets, including farnesoid X receptor (FXR), urokinase-type plasminogen activator (uPA)-urokinase-type plasminogen activator receptor (uPAR) interaction, fecal microbiota transplantation (FMT), and insulin receptor (INSR). Notably, the simultaneous targeting of intestinal inflammation and promotion of epithelial barrier healing shows promise for efficient IBD treatment. Future research should explore targeted therapies and combination treatments, including natural remedies, microbiota colonization, stem cell approaches, and computer-aided drug design. It is also crucial to focus on accurate prognosis and developing a thorough understanding of IBD development mechanisms.
Collapse
Affiliation(s)
- Pardis Mansouri
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran; Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Pegah Mansouri
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran; Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Esmaeil Behmard
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran
| | - Sohrab Najafipour
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran
| | - Amin Kouhpayeh
- Department of Pharmacology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran.
| | - Akbar Farjadfar
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran.
| |
Collapse
|
7
|
Clarelli F, Corona A, Pääkkönen K, Sorosina M, Zollo A, Piehl F, Olsson T, Stridh P, Jagodic M, Hemmer B, Gasperi C, Harroud A, Shchetynsky K, Mingione A, Mascia E, Misra K, Giordano A, Mazzieri MLT, Priori A, Saarela J, Kockum I, Filippi M, Esposito F, Boneschi FGM. Pharmacogenomics of clinical response to Natalizumab in multiple sclerosis: a genome-wide multi-centric association study. J Neurol 2024; 271:7250-7263. [PMID: 39264442 PMCID: PMC11561017 DOI: 10.1007/s00415-024-12608-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Inter-individual differences in treatment response are marked in multiple sclerosis (MS). This is true for Natalizumab (NTZ), to which a subset of patients displays sub-optimal treatment response. We conducted a multi-centric genome-wide association study (GWAS), with additional pathway and network analysis to identify genetic predictors of response to NTZ. METHODS MS patients from three different centers were included. Response to NTZ was dichotomized, nominating responders (R) relapse-free patients and non-responders (NR) all the others, over a follow-up of 4 years. Association analysis on ~ 4.7 M imputed autosomal common single-nucleotide polymorphisms (SNPs) was performed fitting logistic regression models, adjusted for baseline covariates, followed by meta-analysis at SNP and gene level. Finally, these signals were projected onto STRING interactome, to elicit modules and hub genes linked to response. RESULTS Overall, 1834 patients were included: 119 from Italy (R = 94, NR = 25), 81 from Germany (R = 61, NR = 20), and 1634 from Sweden (R = 1349, NR = 285). The top-associated variant was rs11132400T (p = 1.33 × 10-6, OR = 0.58), affecting expression of several genes in the locus, like KLKB1. The interactome analysis implicated a module of 135 genes, with over-representation of terms like canonical WNT signaling pathway (padjust = 7.08 × 10-6). Response-associated genes like GRB2 and LRP6, already implicated in MS pathogenesis, were topologically prioritized within the module. CONCLUSION This GWAS, the largest pharmacogenomic study of response to NTZ, suggested MS-implicated genes and Wnt/β-catenin signaling pathway, an essential component for blood-brain barrier formation and maintenance, to be related to treatment response.
Collapse
Affiliation(s)
- Ferdinando Clarelli
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
| | - Andrea Corona
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
| | - Kimmo Pääkkönen
- Institute for Molecular Medicine Finland (FIMM), University of FI Helsinki, Helsinki, Finland
| | - Melissa Sorosina
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
| | - Alen Zollo
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
| | - Fredrik Piehl
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Tomas Olsson
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Pernilla Stridh
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Maja Jagodic
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Bernhard Hemmer
- Department of Neurology, School of Medicine, Technical University of Munich, Klinikum Rechts Der Isar, Ismaninger Str. 22, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christiane Gasperi
- Department of Neurology, School of Medicine, Technical University of Munich, Klinikum Rechts Der Isar, Ismaninger Str. 22, Munich, Germany
| | - Adil Harroud
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Klementy Shchetynsky
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Alessandra Mingione
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
| | - Elisabetta Mascia
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
| | - Kaalindi Misra
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
| | - Antonino Giordano
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Maria Laura Terzi Mazzieri
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
| | - Alberto Priori
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy
- Clinical Neurology Unit, Azienda Socio-Sanitaria Territoriale Santi Paolo E Carlo and Department of Health Sciences, University of Milan, Milan, Italy
| | - Janna Saarela
- Institute for Molecular Medicine Finland (FIMM), University of FI Helsinki, Helsinki, Finland
| | - Ingrid Kockum
- The Karolinska Neuroimmunology & Multiple Sclerosis Centre, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Visionsgatan 18, 171 76, Stockholm, Sweden
| | - Massimo Filippi
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 48, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy
- Vita-Salute San Raffaele University, Via Olgettina, 60, Milan, Italy
| | - Federica Esposito
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan, Italy.
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Filippo Giovanni Martinelli Boneschi
- Laboratory of Precision Medicine of Neurological Diseases, Department of Health Science, University of Milan, Milan, Italy.
- CRC "Aldo Ravelli" for Experimental Brain Therapeutics, Department of Health Science, University of Milan, Milan, Italy.
- Clinical Neurology Unit, Azienda Socio-Sanitaria Territoriale Santi Paolo E Carlo and Department of Health Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
8
|
Sinha A, Roy S. Prospective therapeutic targets and recent advancements in the treatment of inflammatory bowel disease. Immunopharmacol Immunotoxicol 2024:1-14. [PMID: 39013809 DOI: 10.1080/08923973.2024.2381756] [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: 02/16/2024] [Accepted: 07/14/2024] [Indexed: 07/18/2024]
Abstract
OBJECTIVE Inflammatory Bowel Disease (IBD) poses a persistent challenge in the realm of gastroenterology, necessitating continual exploration of innovative treatment strategies. The limited efficacy and potential side effects associated with existing therapeutic modalities underscore the urgent need for novel approaches in IBD management. This study aims to examine potential therapeutic targets and recent advancements in understanding the disease's intricate pathogenesis, with a spotlight on the gut microbiome, immune dysregulation, and genetic predispositions. METHODS A comprehensive review was conducted to delve into the pressing demand for new avenues in IBD treatment. The study examined potential therapeutic targets such as phosphodiesterase 4 (PDE4) inhibitors, immune system modulators, Tyrosine kinase receptors (TYK), Toll-like receptors (TLRs), modulation of the gut microbiota, stem cell therapy, fibrosis management, interleukins (ILs) regulation, and oxidative stress mitigation. Additionally, advances in precision medicine, biologics, small molecule inhibitors, and microbiome modulation techniques were explored. RESULTS The investigation unveiled promising therapeutic targets and provided insights into recent breakthroughs that herald a transformative era in the therapeutic landscape for IBD. Advances in precision medicine, biologics, small molecule inhibitors, and the exploration of microbiome modulation techniques stood out as pivotal milestones in the field of gastroenterology. CONCLUSIONS The findings offer renewed hope for enhanced efficacy, reduced side effects, and improved patient outcomes in the treatment of IBD. These innovative approaches necessitate continual exploration and underscore the urgent need for novel strategies in IBD management, potentially revolutionizing the realm of gastroenterology.
Collapse
Affiliation(s)
- Akshit Sinha
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, Noida, India
| | - Supriya Roy
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
9
|
Ou Yong BM, Awuah WA, Shah MH, Sanker V, Huk JKS, Venkata SY, Patel DH, Tan JK, Khan NA, Kulasekaran A, Sarkar M, Abdul-Rahman T, Atallah O. Intracerebral haemorrhage in multiple sclerosis: assessing the impact of disease-modifying medications. Eur J Med Res 2024; 29:344. [PMID: 38918831 PMCID: PMC11197372 DOI: 10.1186/s40001-024-01945-x] [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: 02/13/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
Multiple Sclerosis (MS) is a complex autoimmune disorder that significantly impacts the central nervous system, leading to a range of complications. While intracranial haemorrhage (ICH) is a rare but highly morbid complication, more common CNS complications include progressive multifocal leukoencephalopathy (PML) and other CNS infections. This severe form of stroke, known for its high morbidity and mortality rates, presents a critical challenge in the management of MS. The use of disease-modifying drugs (DMDs) in treating MS introduces a nuanced aspect to patient care, with certain medications like Dimethyl Fumarate and Fingolimod showing potential in reducing the risk of ICH, while others such as Alemtuzumab and Mitoxantrone are associated with an increased risk. Understanding the intricate relationship between these DMDs, the pathophysiological mechanisms of ICH, and the individualised aspects of each patient's condition is paramount. Factors such as genetic predispositions, existing comorbidities, and lifestyle choices play a crucial role in tailoring treatment approaches, emphasising the importance of a personalised, vigilant therapeutic strategy. The necessity for ongoing and detailed research cannot be overstated. It is crucial to explore the long-term effects of DMDs on ICH occurrence and prognosis in MS patients, aiming to refine clinical practices and promote patient-centric, informed therapeutic decisions. This approach ensures that the management of MS is not only comprehensive but also adaptable to the evolving understanding of the disease and its treatments.
Collapse
Affiliation(s)
| | | | | | - Vivek Sanker
- Department of Neurosurgery, Trivandrum Medical College, Thiruvananthapuram, India
| | | | | | - Diti H Patel
- Nova Southeastern University Dr. Kiran C Patel College of Allopathic Medicine, Davie, FL, USA
| | | | - Noor Ayman Khan
- DOW Medical College, DOW University of Health Sciences (DUHS), Baba-E-Urdu Road, Karachi, Pakistan
| | | | - Manali Sarkar
- MGM Medical College Navi, Mumbai, Maharashtra, India
| | | | - Oday Atallah
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| |
Collapse
|
10
|
Aliyu M, Zohora FT, Ceylan A, Hossain F, Yazdani R, Azizi G. Immunopathogenesis of multiple sclerosis: molecular and cellular mechanisms and new immunotherapeutic approaches. Immunopharmacol Immunotoxicol 2024; 46:355-377. [PMID: 38634438 DOI: 10.1080/08923973.2024.2330642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/09/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a central nervous system (CNS) demyelinating autoimmune disease with increasing global prevalence. It predominantly affects females, especially those of European descent. The interplay between environmental factors and genetic predisposition plays a crucial role in MS etiopathogenesis. METHODS We searched recent relevant literature on reputable databases, which include, PubMed, Embase, Web of Science, Scopus, and ScienceDirect using the following keywords: multiple sclerosis, pathogenesis, autoimmunity, demyelination, therapy, and immunotherapy. RESULTS Various animal models have been employed to investigate the MS etiopathogenesis and therapeutics. Autoreactive T cells within the CNS recruit myeloid cells through chemokine expression, leading to the secretion of inflammatory cytokines driving the MS pathogenesis, resulting in demyelination, gliosis, and axonal loss. Key players include T cell lymphocytes (CD4+ and CD8+), B cells, and neutrophils. Signaling dysregulation in inflammatory pathways and the immunogenetic basis of MS are essential considerations for any successful therapy to MS. Data indicates that B cells and neutrophils also have significant roles in MS, despite the common belief that T cells are essential. High neutrophil-to-lymphocyte ratios correlate with MS severity, indicating their contribution to disease progression. Dysregulated signaling pathways further exacerbate MS progression. CONCLUSION MS remains incurable, but disease-modifying therapies, monoclonal antibodies, and immunomodulatory drugs offer hope for patients. Research on the immunogenetics and immunoregulatory functions of gut microbiota is continuing to provide light on possible treatment avenues. Understanding the complex interplay between genetic predisposition, environmental factors, and immune dysregulation is critical for developing effective treatments for MS.
Collapse
Affiliation(s)
- Mansur Aliyu
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, International Campus, TUMS-IC, Tehran, Iran
- Department of Medical Microbiology, Faculty of Clinical Science, College of Health Sciences, Bayero University, Kano, Nigeria
| | - Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ayca Ceylan
- Medical Faculty, Department of Pediatrics, Division of Immunology and Allergy, Selcuk University, Konya, Turkey
| | - Fariha Hossain
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Reza Yazdani
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gholamreza Azizi
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
11
|
Brown JD, Muston BT, Massey J. Switching from natalizumab to an anti-CD20 monoclonal antibody in relapsing remitting multiple sclerosis: A systematic review. Mult Scler Relat Disord 2024; 86:105605. [PMID: 38640586 DOI: 10.1016/j.msard.2024.105605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Use of natalizumab (NTZ) is precluded in many Multiple Sclerosis (MS) patients by the risk of progressive multifocal leukoencephalopathy (PML). Regardless, some patients may commence natalizumab for short term disease control in spite of being seropositive, and others may seroconvert whilst on treatment. In these circumstances, discontinuation of NTZ should not occur until a clear exit strategy is established to prevent post-NTZ disease reactivation, which often exceeds the severity of disease activity prior to NTZ treatment. The objective of this systematic review was to summarise the available evidence for CD20-monoclonal antibodies (CD20mAb) as a suitable NTZ exit strategy, and to identify whether a superior switch protocol can be established. METHODS In accordance with PRISMA guidelines, a total of 2393 references were extracted from a search of three online databases (PubMed, Scopus, MEDLINE). Following the application of inclusion/exclusion criteria, a total of 5 studies representing 331 patients were included. RESULTS The overall incidence of clinical relapse during washout periods ranging from 4.4-10.7 weeks was 0 %. The incidence of clinical relapse during two-year follow-up ranged from 1.8 % to 10 % for switches to all types of CD20 monoclonal antibody. The weighted mean for clinical relapse at 12 months was 8.8 %. Three studies reported an annualised relapse rate (ARR) ranging from 0.02-0.12 with a weighted mean ARR of 0.07. The overall incidence of PML during washout was 0 % and the overall incidence of PML within 6 months follow-up was 0.6 %. CONCLUSIONS This systematic review provides the first attempt at identifying a superior switch protocol in patients at risk of PML transitioning from NTZ to a CD20mAb. Our results indicate that CD20mAb's are a suitable transitional option for patients who discontinue NTZ, with our cohort demonstrating very low rates of carryover PML and low rates of clinical relapse. The most appropriate washout period is unclear due to confounding factors but is likely between 4 and 12 weeks.
Collapse
Affiliation(s)
| | - Benjamin T Muston
- Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; The Collaborative Research Group (CORE), Sydney, Australia
| | - Jennifer Massey
- Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; Neurology Department, St Vincent's Hospital Sydney, Australia
| |
Collapse
|
12
|
Angeli E, Jordan M, Otto M, Stojanović SD, Karsdal M, Bauersachs J, Thum T, Fiedler J, Genovese F. The role of fibrosis in cardiomyopathies: An opportunity to develop novel biomarkers of disease activity. Matrix Biol 2024; 128:65-78. [PMID: 38423395 DOI: 10.1016/j.matbio.2024.02.008] [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: 10/16/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Cardiomyopathies encompass a spectrum of heart disorders with diverse causes and presentations. Fibrosis stands out as a shared hallmark among various cardiomyopathies, reflecting a common thread in their pathogenesis. This prevalent fibrotic response is intricately linked to the consequences of dysregulated extracellular matrix (ECM) remodeling, emphasizing its significance in the development and progression the disease. This review explores the ECM involvement in various cardiomyopathies and its impact on myocardial stiffness and fibrosis. Additionally, we discuss the potential of ECM fragments as early diagnosis, prognosis, and risk stratification. Biomarkers deriving from turnover of collagens and other ECM proteins hold promise in clinical applications. We outline current clinical management, future directions, and the potential for personalized ECM-targeted therapies with specific focus on microRNAs. In summary, this review examines the role of the fibrosis in cardiomyopathies, highlighting the potential of ECM-derived biomarkers in improving disease management with implications for precision medicine.
Collapse
Affiliation(s)
- Elisavet Angeli
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Nordic Bioscience A/S, Herlev, Denmark.
| | - Maria Jordan
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany
| | - Mandy Otto
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany
| | - Stevan D Stojanović
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Federal Republic of Germany; Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Federal Republic of Germany
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Federal Republic of Germany
| | - Thomas Thum
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany; Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Federal Republic of Germany
| | - Jan Fiedler
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany
| | - Federica Genovese
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Muller I, Maioli S, Armenti M, Porcaro L, Currò N, Iofrida E, Pignataro L, Manso J, Mian C, Geginat J, Salvi M. Alemtuzumab-induced thyroid eye disease successfully treated with a single low dose of rituximab. Eur Thyroid J 2024; 13:e230236. [PMID: 38471303 PMCID: PMC11046353 DOI: 10.1530/etj-23-0236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/12/2024] [Indexed: 03/14/2024] Open
Abstract
Introduction Secondary thyroid autoimmunity, especially Graves' disease (GD), frequently develops in patients with multiple sclerosis (MS) following alemtuzumab treatment (ALTZ; anti-CD52). Thyroid eye disease (TED) can also develop, and rituximab (RTX; anti-CD20) is a suitable treatment. Case presentation A 37-year-old woman with MS developed steroid-resistant active moderate-to-severe TED 3 years after ALTZ, that successfully responded to a single 500 mg dose of i.v. RTX. Before RTX peripheral B-cells were low, and were totally depleted immediately after therapy. Follow-up analysis 4 years post ALTZ and 1 year post RTX showed persistent depletion of B cells, and reduction of T regulatory cells in both peripheral blood and thyroid tissue obtained at thyroidectomy. Conclusion RTX therapy successfully inactivated TED in a patient with low B-cell count derived from previous ALTZ treatment. B-cell depletion in both thyroid and peripheral blood was still present 1 year after RTX, indicating a likely cumulative effect of both treatments.
Collapse
Affiliation(s)
- Ilaria Muller
- Department of Clinical Sciences and Community Health, University of Milan, Italy
- Endocrinology Unit, Graves’ Orbitopathy Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Maioli
- Department of Clinical Sciences and Community Health, University of Milan, Italy
| | - Mirco Armenti
- Department of Clinical Sciences and Community Health, University of Milan, Italy
| | - Laura Porcaro
- Department of Clinical Sciences and Community Health, University of Milan, Italy
| | - Nicola Currò
- Endocrinology Unit, Graves’ Orbitopathy Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Ophthalmology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisabetta Iofrida
- Department of Specialistic Surgical Sciences, Otolaryngology and Head and Neck Surgery, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Pignataro
- Department of Clinical Sciences and Community Health, University of Milan, Italy
- Department of Specialistic Surgical Sciences, Otolaryngology and Head and Neck Surgery, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jacopo Manso
- Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Caterina Mian
- Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Jens Geginat
- Department of Clinical Sciences and Community Health, University of Milan, Italy
- National Institute of Molecular Genetics (INGM) “Romeo and Enrica Invernizzi”, Milan, Italy
| | - Mario Salvi
- Endocrinology Unit, Graves’ Orbitopathy Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
14
|
Inojosa H, Ziemssen T. [Current and innovative Approaches to Multiple Sclerosis Therapy]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2024; 92:41-60. [PMID: 38272020 DOI: 10.1055/a-2167-1391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
The landscape of immunotherapies in the management of Multiple Sclerosis (MS) is currently particularly dynamic. Over 21 immunotherapeutic options are approved by the European Meidcines Agency (EMA), Food and Drug Administration (FDA) and newer approaches are ongoing in clinical trials. With advancements in the understanding of MS pathophysiology and further development of diagnosis criteria, newer and more specific disease-modifying therapies (DMTs) have emerged in recent years. The selection and timing of proper therapeutic approaches is increasingly complex. We provide an overview of the available immunotherapies for a personalized MS treatment and discuss practical insights into their application. The importance of early intervention, distinction between escalation and induction approaches, and consideration of high-efficacy treatments for specific patient groups are in discussed. We emphasize the significance of a patient-centered approach, taking into account various factors such as comorbidities, family planning, administration preferences and potential side effects in treatment decision-making.
Collapse
|
15
|
Clinton JW, Cross RK. Personalized Treatment for Crohn's Disease: Current Approaches and Future Directions. Clin Exp Gastroenterol 2023; 16:249-276. [PMID: 38111516 PMCID: PMC10726957 DOI: 10.2147/ceg.s360248] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023] Open
Abstract
Crohn's disease is a complex, relapsing and remitting inflammatory disorder of the gastrointestinal tract with a variable disease course. While the treatment options for Crohn's disease have dramatically increased over the past two decades, predicting individual patient response to treatment remains a challenge. As a result, patients often cycle through multiple different therapies before finding an effective treatment which can lead to disease complications, increased costs, and decreased quality of life. Recently, there has been increased emphasis on personalized medicine in Crohn's disease to identify individual patients who require early advanced therapy to prevent complications of their disease. In this review, we summarize our current approach to management of Crohn's disease by identifying risk factors for severe or disabling disease and tailoring individual treatments to patient-specific goals. Lastly, we outline our knowledge gaps in implementing personalized Crohn's disease treatment and describe the future directions in precision medicine.
Collapse
Affiliation(s)
- Joseph William Clinton
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Raymond Keith Cross
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Schwinghamer K, Siahaan TJ. Enhancing Antibody Exposure in the Central Nervous System: Mechanisms of Uptake, Clearance, and Strategies for Improved Brain Delivery. JOURNAL OF NANOTHERANOSTICS 2023; 4:463-479. [PMID: 39897432 PMCID: PMC11784990 DOI: 10.3390/jnt4040020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
Abstract
Antibodies (mAbs) are attractive molecules for their application as a diagnostic and therapeutic agent for diseases of the central nervous system (CNS). mAbs can be generated to have high affinity and specificity to target molecules in the CNS. Unfortunately, only a very small number of mAbs have been specifically developed and approved for neurological indications. This is primarily attributed to their low exposure within the CNS, hindering their ability to reach and effectively engage their potential targets in the brain. This review discusses aspects of various barriers such as the blood-brain barrier (BBB) and blood-cerebrospinal fluid (CSF) barrier (BCSFB) that regulate the entry and clearance of mAbs into and from the brain. The roles of the glymphatic system on brain exposure and clearance are being described. We also discuss the proposed mechanisms of the uptake of mAbs into the brain and for clearance. Finally, several methods of enhancing the exposure of mAbs in the CNS were discussed, including receptor-mediated transcytosis, osmotic BBB opening, focused ultrasound (FUS), BBB-modulating peptides, and enhancement of mAb brain retention.
Collapse
Affiliation(s)
- Kelly Schwinghamer
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66046, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66046, USA
| |
Collapse
|
17
|
Sharma P, Joshi RV, Pritchard R, Xu K, Eicher MA. Therapeutic Antibodies in Medicine. Molecules 2023; 28:6438. [PMID: 37764213 PMCID: PMC10535987 DOI: 10.3390/molecules28186438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/05/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Antibody engineering has developed into a wide-reaching field, impacting a multitude of industries, most notably healthcare and diagnostics. The seminal work on developing the first monoclonal antibody four decades ago has witnessed exponential growth in the last 10-15 years, where regulators have approved monoclonal antibodies as therapeutics and for several diagnostic applications, including the remarkable attention it garnered during the pandemic. In recent years, antibodies have become the fastest-growing class of biological drugs approved for the treatment of a wide range of diseases, from cancer to autoimmune conditions. This review discusses the field of therapeutic antibodies as it stands today. It summarizes and outlines the clinical relevance and application of therapeutic antibodies in treating a landscape of diseases in different disciplines of medicine. It discusses the nomenclature, various approaches to antibody therapies, and the evolution of antibody therapeutics. It also discusses the risk profile and adverse immune reactions associated with the antibodies and sheds light on future applications and perspectives in antibody drug discovery.
Collapse
Affiliation(s)
- Prerna Sharma
- Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA
| | | | | | | | | |
Collapse
|
18
|
Troncoso MF, Díaz-Vesga MC, Sanhueza-Olivares F, Riquelme JA, Müller M, Garrido L, Gabrielli L, Chiong M, Corbalan R, Castro PF, Lavandero S. Targeting VCAM-1: a therapeutic opportunity for vascular damage. Expert Opin Ther Targets 2023; 27:207-223. [PMID: 36880349 DOI: 10.1080/14728222.2023.2187778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
INTRODUCTION The vascular cell adhesion molecule (VCAM-1) is a transmembrane sialoglycoprotein detected in activated endothelial and vascular smooth muscle cells involved in the adhesion and transmigration of inflammatory cells into damaged tissue. Widely used as a pro-inflammatory marker, its potential role as a targeting molecule has not been thoroughly explored. AREAS COVERED We discuss the current evidence supporting the potential targeting of VCAM-1 in atherosclerosis, diabetes, hypertension and ischemia/reperfusion injury. EXPERT OPINION There is emerging evidence that VCAM-1 is more than a biomarker and may be a promising therapeutic target for vascular diseases. While there are neutralizing antibodies that allow preclinical research, the development of pharmacological tools to activate or inhibit this protein are required to thoroughly assess its therapeutic potential.
Collapse
Affiliation(s)
- Mayarling F Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Magda C Díaz-Vesga
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Pontificia Universidad Javeriana de Cali, Cali, Colombia
| | - Fernanda Sanhueza-Olivares
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Jaime A Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marioly Müller
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Luis Garrido
- Division of Cardiovascular Diseases, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luigi Gabrielli
- Division Surgery, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ramon Corbalan
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo F Castro
- Division Surgery, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
19
|
Borda M, Aquino JB, Mazzone GL. Cell-based experimental strategies for myelin repair in multiple sclerosis. J Neurosci Res 2023; 101:86-111. [PMID: 36164729 DOI: 10.1002/jnr.25129] [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: 05/15/2022] [Revised: 08/21/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disorder of the central nervous system (CNS), diagnosed at a mean age of 32 years. CNS glia are crucial players in the onset of MS, primarily involving astrocytes and microglia that can cause/allow massive oligodendroglial cells death, without immune cell infiltration. Current therapeutic approaches are aimed at modulating inflammatory reactions during relapsing episodes, but lack the ability to induce very significant repair mechanisms. In this review article, different experimental approaches based mainly on the application of different cell types as therapeutic strategies applied for the induction of myelin repair and/or the amelioration of the disease are discussed. Regarding this issue, different cell sources were applied in various experimental models of MS, with different results, both in significant improvements in remyelination and the reduction of neuroinflammation and glial activation, or in neuroprotection. All cell types tested have advantages and disadvantages, which makes it difficult to choose a better option for therapeutic application in MS. New strategies combining cell-based treatment with other applications would result in further improvements and would be good candidates for MS cell therapy and myelin repair.
Collapse
Affiliation(s)
- Maximiliano Borda
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
| | - Jorge B Aquino
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina.,CONICET, Comisión Nacional de Investigaciones Científicas y Técnicas
| | - Graciela L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina.,CONICET, Comisión Nacional de Investigaciones Científicas y Técnicas
| |
Collapse
|
20
|
Chowdhury R, Kane SV. Pregnancy and Crohn's disease: concerns and assurance of medical therapy. Gastroenterol Rep (Oxf) 2022; 10:goac055. [PMID: 36225722 PMCID: PMC9550230 DOI: 10.1093/gastro/goac055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/04/2022] Open
Abstract
Approximately 50% of patients with inflammatory bowel disease including both Crohn's disease and ulcerative colitis are female with many being diagnosed and treated during their reproductive years. It is important for women to be in remission prior to and during pregnancy. There have been many advances in the treatment of inflammatory bowel disease, including new therapies. In this review, we summarize the currently approved medications for Crohn's disease and their safety in pregnancy and postpartum. The totality of evidence suggests that the majority of therapies are low-risk before and during pregnancy, and should be continued to control maternal disease.
Collapse
Affiliation(s)
- Reezwana Chowdhury
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sunanda V Kane
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
21
|
Cegarra C, Cameron B, Chaves C, Dabdoubi T, Do TM, Genêt B, Roudières V, Shi Y, Tchepikoff P, Lesuisse D. An innovative strategy to identify new targets for delivering antibodies to the brain has led to the exploration of the integrin family. PLoS One 2022; 17:e0274667. [PMID: 36108060 PMCID: PMC9477330 DOI: 10.1371/journal.pone.0274667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/01/2022] [Indexed: 11/19/2022] Open
Abstract
Background
Increasing brain exposure of biotherapeutics is key to success in central nervous system disease drug discovery. Accessing the brain parenchyma is especially difficult for large polar molecules such as biotherapeutics and antibodies because of the blood-brain barrier. We investigated a new immunization strategy to identify novel receptors mediating transcytosis across the blood-brain barrier.
Method
We immunized mice with primary non-human primate brain microvascular endothelial cells to obtain antibodies. These antibodies were screened for their capacity to bind and to be internalized by primary non-human primate brain microvascular endothelial cells and Human Cerebral Microvascular Endothelial Cell clone D3. They were further evaluated for their transcytosis capabilities in three in vitro blood-brain barrier models. In parallel, their targets were identified by two different methods and their pattern of binding to human tissue was investigated using immunohistochemistry.
Results
12 antibodies with unique sequence and internalization capacities were selected amongst more than six hundred. Aside from one antibody targeting Activated Leukocyte Cell Adhesion Molecule and one targeting Striatin3, most of the other antibodies recognized β1 integrin and its heterodimers. The antibody with the best transcytosis capabilities in all blood-brain barrier in vitro models and with the best binding capacity was an anti-αnβ1 integrin. In comparison, commercial anti-integrin antibodies performed poorly in transcytosis assays, emphasizing the originality of the antibodies derived here. Immunohistochemistry studies showed specific vascular staining on human and non-human primate tissues.
Conclusions
This transcytotic behavior has not previously been reported for anti-integrin antibodies. Further studies should be undertaken to validate this new mechanism in vivo and to evaluate its potential in brain delivery.
Collapse
Affiliation(s)
- Céline Cegarra
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
- * E-mail:
| | | | - Catarina Chaves
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | | | - Tuan-Minh Do
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | - Bruno Genêt
- Integrated Drug Discovery, Sanofi, Vitry-Sur-Seine, France
| | - Valérie Roudières
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | - Yi Shi
- Histology, Translational Sciences, Sanofi, Vitry-Sur-Seine, France
| | | | - Dominique Lesuisse
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| |
Collapse
|
22
|
Galarza-Muñoz G, Kennedy-Boone D, Schott G, Bradrick SS, Garcia-Blanco MA. Antisense modulation of IL7R splicing to control sIL7R expression in human CD4 + T cells. RNA (NEW YORK, N.Y.) 2022; 28:1058-1073. [PMID: 35613883 PMCID: PMC9297843 DOI: 10.1261/rna.079137.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
The interleukin 7 receptor (IL7R) is strongly associated with increased risk to develop multiple sclerosis (MS), an autoimmune disease of the central nervous system, and this association is likely driven by up-regulation of the soluble isoform of IL7R (sIL7R). Expression of sIL7R is determined by exclusion of the alternative exon 6 from IL7R transcripts, and our previous work revealed that the MS risk allele of the SNP rs6897932 within this exon enhances the expression of sIL7R by promoting exclusion of exon 6. sIL7R potentiates the activity of IL7, leading to enhanced expansion of T cells and increased disability in the experimental autoimmune encephalomyelitis (EAE) murine model of MS. This role in modulating T cell-driven immunity positions sIL7R as an attractive therapeutic target whose expression could be reduced for treatment of MS or increased for treatment of cancers. In this study, we identified novel antisense oligonucleotides (ASOs) that effectively control the inclusion (anti-sIL7R ASOs) or exclusion (pro-sIL7R ASOs) of this exon in a dose-dependent fashion. These ASOs provided excellent control of exon 6 splicing and sIL7R secretion in human primary CD4+ T cells. Supporting their potential for therapeutic targeting, we showed that lead anti-sIL7R ASOs correct the enhanced exon 6 exclusion imposed by the MS risk allele of rs6897932, whereas lead pro-sIL7R ASOs phenocopy it. The data presented here form the foundation for future preclinical studies that will test the therapeutic potential of these ASOs in MS and immuno-oncology.
Collapse
Affiliation(s)
- Gaddiel Galarza-Muñoz
- Autoimmunity BioSolutions, Galveston, Texas 77550, USA
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Debbie Kennedy-Boone
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Geraldine Schott
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Mariano A Garcia-Blanco
- Autoimmunity BioSolutions, Galveston, Texas 77550, USA
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas 77550, USA
| |
Collapse
|
23
|
Assessing Blood-Based Biomarkers to Define a Therapeutic Window for Natalizumab. J Pers Med 2021; 11:jpm11121347. [PMID: 34945819 PMCID: PMC8706232 DOI: 10.3390/jpm11121347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/21/2022] Open
Abstract
Natalizumab is a monoclonal antibody that binds CD49d. Although it is one of the most effective treatments for Relapsing-Remitting Multiple Sclerosis (RRMS), a dosing regimen has not been optimized for safety and efficacy in individual patients. We aimed to identify biomarkers to monitor Natalizumab treatment and to establish a personalized dose utilizing an ongoing longitudinal study in 29 RRMS patients under Natalizumab with standard interval dose (SD) of 300 mg/4wks or extended interval dose (EID) of 300 mg/6wks. Blood samples were analyzed by flow cytometry to determine CD49d saturation and expression in several T and B lymphocytes subpopulations. Each patient was analyzed at two different timepoints separated by 3 Natalizumab administrations. Natalizumab and sVCAM-1 levels in serum were also analyzed using ELISA. To determine the reproducibility of various markers, two different timepoints were compared and no significant differences were observed for CD49d expression nor for saturation; SD patients had higher saturation levels (~80%) than EID patients (~60%). A positive correlation exists between CD49d saturation and Natalizumab serum levels. CD49d expression and saturation are stable parameters that could be used as biomarkers in the immunomonitoring of Natalizumab treatment. Moreover, Natalizumab and sVCAM-1 serum levels could be used to optimize an individual's dosing schedule.
Collapse
|
24
|
Briggs FBS, Mateen FJ, Schmidt H, Currie KM, Siefers HM, Crouthamel S, Bebo BF, Fiol J, Racke MK, O'Connor KC, Kolaczkowski LG, Klein P, Loud S, McBurney RN. COVID-19 Vaccination Reactogenicity in Persons With Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 9:9/1/e1104. [PMID: 34753828 PMCID: PMC8579248 DOI: 10.1212/nxi.0000000000001104] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES There are limited data on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine reactogenicity in persons with multiple sclerosis (PwMS) and how reactogenicity is affected by disease-modifying therapies (DMTs). The objective of this retrospective cross-sectional study was to generate real-world multiple sclerosis-specific vaccine safety information, particularly in the context of specific DMTs, and provide information to mitigate specific concerns in vaccine hesitant PwMS. METHODS Between 3/2021 and 6/2021, participants in iConquerMS, an online people-powered research network, reported SARS-CoV-2 vaccines, experiences of local (itch, pain, redness, swelling, or warmth at injection site) and systemic (fever, chills, fatigue, headache, joint pain, malaise, muscle ache, nausea, allergic, and other) reactions within 24 hours (none, mild, moderate, and severe), DMT use, and other attributes. Multivariable models characterized associations between clinical factors and reactogenicity. RESULTS In 719 PwMS, 64% reported experiencing a reaction after their first vaccination shot, and 17% reported a severe reaction. The most common reactions were pain at injection site (54%), fatigue (34%), headache (28%), and malaise (21%). Younger age, being female, prior SARS-CoV-2 infection, and receiving the ChAdOx1 nCoV-19 (Oxford-AstraZeneca) vs BNT162b2 (Pfizer-BioNTech) vaccine were associated with experiencing a reaction after the first vaccine dose. Similar relationships were observed for a severe reaction, including higher odds of reactions among PwMS with more physical impairment and lower odds of reactions for PwMS on an alpha4-integrin blocker or sphingosine-1-phosphate receptor modulator. In 442 PwMS who received their second vaccination shot, 74% reported experiencing a reaction, whereas 22% reported a severe reaction. Reaction profiles after the second shot were similar to those reported after the first shot. Younger PwMS and those who received the mRNA-1273 (Moderna) vs BNT162b2 vaccine reported higher reactogenicity after the second shot, whereas those on a sphingosine-1-phosphate receptor modulator or fumarate were significantly less likely to report a reaction. DISCUSSION SARS-CoV-2 vaccine reactogenicity profiles and the associated factors in this convenience sample of PwMS appear similar to those reported in the general population. PwMS on specific DMTs were less likely to report vaccine reactions. Overall, the short-term vaccine reactions experienced in the study population were mostly self-limiting, including pain at the injection site, fatigue, headache, and fever.
Collapse
Affiliation(s)
- Farren Basil Shaw Briggs
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA.
| | - Farrah J Mateen
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Hollie Schmidt
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Keisha M Currie
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Heather M Siefers
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Slavka Crouthamel
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Bruce F Bebo
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Julie Fiol
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Michael K Racke
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Kevin C O'Connor
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Laura G Kolaczkowski
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Phyllis Klein
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Sara Loud
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Robert Nicholas McBurney
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| |
Collapse
|
25
|
Cheraqpour K, Ahmadraji A, Rashidinia A, Irannejad M, Shahriari M. Acute retinal necrosis caused by co-infection with multiple viruses in a natalizumab-treated patient: a case report and brief review of literature. BMC Ophthalmol 2021; 21:337. [PMID: 34530769 PMCID: PMC8447524 DOI: 10.1186/s12886-021-02096-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/31/2021] [Indexed: 12/22/2022] Open
Abstract
Background Acute retinal necrosis is considered a rare infectious uveitis. This condition is usually caused by varicella-zoster virus or herpes simplex virus. Acute retinal necrosis caused by co-infection with multiple viruses is extremely rare. Herein, we report a case of acute retinal necrosis caused by co-infection with herpes simplex virus (type I and II) and varicella-zoster virus (VZV) in a natalizumab-treated patient due to multiple sclerosis. Case presentation An adult man presented with a complaint of decreased vision of the right eye from 12 days ago. He was a known case of multiple sclerosis receiving natalizumab. Examination of the right eye revealed severe conjunctival injection, fine diffuse keratic precipitates, 3 + anterior chamber and vitreous cells, elevated intraocular pressure (26 mmHg), a blurred optic disk with hemorrhagic patches, and occlusive vasculitis plus confluent necrotizing patches in the peripheral retina compatible with diagnosis of acute retinal necrosis. He underwent anterior chamber and vitreous tap, and real-time PCR detected HSV I & II and VZV on the vitreous specimen. A second PCR showed the same result. After neurological consultation, natalizumab was discontinued and intravenous acyclovir was started followed by oral acyclovir and oral prednisolone to control the disease, which was successful. Conclusions Although rare, multiple-viral infection should be considered in the physiopathology of acute retinal necrosis, especially in immunosuppressed patients.
Collapse
Affiliation(s)
- Kasra Cheraqpour
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, 1336616351, Tehran, Iran
| | - Aliasghar Ahmadraji
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, 1336616351, Tehran, Iran.
| | - Ali Rashidinia
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, 1336616351, Tehran, Iran
| | - Maziyar Irannejad
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, 1336616351, Tehran, Iran
| | - Mansoor Shahriari
- Imam Hossein Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Gutiérrez-Fernández M, de la Cuesta F, Tallón A, Cuesta I, Fernández-Fournier M, Laso-García F, Gómez-de Frutos MC, Díez-Tejedor E, Otero-Ortega L. Potential Roles of Extracellular Vesicles as Biomarkers and a Novel Treatment Approach in Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms22169011. [PMID: 34445717 PMCID: PMC8396540 DOI: 10.3390/ijms22169011] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/26/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of bilayer membrane-wrapped molecules that play an important role in cell-to-cell communication, participating in many physiological processes and in the pathogenesis of several diseases, including multiple sclerosis (MS). In recent years, many studies have focused on EVs, with promising results indicating their potential role as biomarkers in MS and helping us better understand the pathogenesis of the disease. Recent evidence suggests that there are novel subpopulations of EVs according to cell origin, with those derived from cells belonging to the nervous and immune systems providing information regarding inflammation, demyelination, axonal damage, astrocyte and microglia reaction, blood–brain barrier permeability, leukocyte transendothelial migration, and ultimately synaptic loss and neuronal death in MS. These biomarkers can also provide insight into disease activity and progression and can differentiate patients’ disease phenotype. This information can enable new pathways for therapeutic target discovery, and consequently the development of novel treatments. Recent evidence also suggests that current disease modifying treatments (DMTs) for MS modify the levels and content of circulating EVs. EVs might also serve as biomarkers to help monitor the response to DMTs, which could improve medical decisions concerning DMT initiation, choice, escalation, and withdrawal. Furthermore, EVs could act not only as biomarkers but also as treatment for brain repair and immunomodulation in MS. EVs are considered excellent delivery vehicles. Studies in progress show that EVs containing myelin antigens could play a pivotal role in inducing antigen-specific tolerance of autoreactive T cells as a novel strategy for the treatment as “EV-based vaccines” for MS. This review explores the breakthrough role of nervous and immune system cell-derived EVs as markers of pathological disease mechanisms and potential biomarkers of treatment response in MS. In addition, this review explores the novel role of EVs as vehicles for antigen delivery as a therapeutic vaccine to restore immune tolerance in MS autoimmunity.
Collapse
Affiliation(s)
- María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, 28046 Madrid, Spain; (M.G.-F.); (A.T.); (I.C.); (M.F.-F.); (F.L.-G.); (M.C.G.-d.F.)
| | - Fernando de la Cuesta
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, 28046 Madrid, Spain;
| | - Antonio Tallón
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, 28046 Madrid, Spain; (M.G.-F.); (A.T.); (I.C.); (M.F.-F.); (F.L.-G.); (M.C.G.-d.F.)
| | - Inmaculada Cuesta
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, 28046 Madrid, Spain; (M.G.-F.); (A.T.); (I.C.); (M.F.-F.); (F.L.-G.); (M.C.G.-d.F.)
| | - Mireya Fernández-Fournier
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, 28046 Madrid, Spain; (M.G.-F.); (A.T.); (I.C.); (M.F.-F.); (F.L.-G.); (M.C.G.-d.F.)
| | - Fernando Laso-García
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, 28046 Madrid, Spain; (M.G.-F.); (A.T.); (I.C.); (M.F.-F.); (F.L.-G.); (M.C.G.-d.F.)
| | - Mari Carmen Gómez-de Frutos
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, 28046 Madrid, Spain; (M.G.-F.); (A.T.); (I.C.); (M.F.-F.); (F.L.-G.); (M.C.G.-d.F.)
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, 28046 Madrid, Spain; (M.G.-F.); (A.T.); (I.C.); (M.F.-F.); (F.L.-G.); (M.C.G.-d.F.)
- Correspondence: (E.D.-T.); (L.O.-O.); Tel.: +34-91-207-1028 (L.O.-O.)
| | - Laura Otero-Ortega
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, 28046 Madrid, Spain; (M.G.-F.); (A.T.); (I.C.); (M.F.-F.); (F.L.-G.); (M.C.G.-d.F.)
- Correspondence: (E.D.-T.); (L.O.-O.); Tel.: +34-91-207-1028 (L.O.-O.)
| |
Collapse
|
27
|
Mitchell D, Shireman J, Sierra Potchanant EA, Lara-Velazquez M, Dey M. Neuroinflammation in Autoimmune Disease and Primary Brain Tumors: The Quest for Striking the Right Balance. Front Cell Neurosci 2021; 15:716947. [PMID: 34483843 PMCID: PMC8414998 DOI: 10.3389/fncel.2021.716947] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
According to classical dogma, the central nervous system (CNS) is defined as an immune privileged space. The basis of this theory was rooted in an incomplete understanding of the CNS microenvironment, however, recent advances such as the identification of resident dendritic cells (DC) in the brain and the presence of CNS lymphatics have deepened our understanding of the neuro-immune axis and revolutionized the field of neuroimmunology. It is now understood that many pathological conditions induce an immune response in the CNS, and that in many ways, the CNS is an immunologically distinct organ. Hyperactivity of neuro-immune axis can lead to primary neuroinflammatory diseases such as multiple sclerosis and antibody-mediated encephalitis, whereas immunosuppressive mechanisms promote the development and survival of primary brain tumors. On the therapeutic front, attempts are being made to target CNS pathologies using various forms of immunotherapy. One of the most actively investigated areas of CNS immunotherapy is for the treatment of glioblastoma (GBM), the most common primary brain tumor in adults. In this review, we provide an up to date overview of the neuro-immune axis in steady state and discuss the mechanisms underlying neuroinflammation in autoimmune neuroinflammatory disease as well as in the development and progression of brain tumors. In addition, we detail the current understanding of the interactions that characterize the primary brain tumor microenvironment and the implications of the neuro-immune axis on the development of successful therapeutic strategies for the treatment of CNS malignancies.
Collapse
Affiliation(s)
- Dana Mitchell
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jack Shireman
- Dey Malignant Brain Tumor Laboratory, Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | | | - Montserrat Lara-Velazquez
- Dey Malignant Brain Tumor Laboratory, Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Mahua Dey
- Dey Malignant Brain Tumor Laboratory, Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
28
|
Mockus TE, Munie A, Atkinson JR, Segal BM. Encephalitogenic and Regulatory CD8 T Cells in Multiple Sclerosis and Its Animal Models. THE JOURNAL OF IMMUNOLOGY 2021; 206:3-10. [PMID: 33443060 DOI: 10.4049/jimmunol.2000797] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS), a neuroinflammatory disease that affects millions worldwide, is widely thought to be autoimmune in etiology. Historically, research into MS pathogenesis has focused on autoreactive CD4 T cells because of their critical role in the animal model, experimental autoimmune encephalomyelitis, and the association between MS susceptibility and single-nucleotide polymorphisms in the MHC class II region. However, recent studies have revealed prominent clonal expansions of CD8 T cells within the CNS during MS. In this paper, we review the literature on CD8 T cells in MS, with an emphasis on their potential effector and regulatory properties. We discuss the impact of disease modifying therapies, currently prescribed to reduce MS relapse rates, on CD8 T cell frequency and function. A deeper understanding of the role of CD8 T cells in MS may lead to the development of more effective and selective immunomodulatory drugs for particular subsets of patients.
Collapse
Affiliation(s)
- Taryn E Mockus
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Ashley Munie
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210.,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Jeffrey R Atkinson
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Benjamin M Segal
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210; .,Neuroscience Research Institute, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
29
|
Kapoor T, Mehan S. Neuroprotective Methodologies in the Treatment of Multiple Sclerosis Current Status of Clinical and Pre-clinical Findings. Curr Drug Discov Technol 2021; 18:31-46. [PMID: 32031075 DOI: 10.2174/1570163817666200207100903] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/02/2019] [Accepted: 11/26/2019] [Indexed: 11/22/2022]
Abstract
Multiple sclerosis is an idiopathic and autoimmune associated motor neuron disorder that affects myelinated neurons in specific brain regions of young people, especially females. MS is characterized by oligodendrocytes destruction further responsible for demyelination, neuroinflammation, mitochondrial abnormalities, oxidative stress and neurotransmitter deficits associated with motor and cognitive dysfunctions, vertigo and muscle weakness. The limited intervention of pharmacologically active compounds like interferon-β, mitoxantrone, fingolimod and monoclonal antibodies used clinically are majorly associated with adverse drug reactions. Pre-clinically, gliotoxin ethidium bromide mimics the behavioral and neurochemical alterations in multiple sclerosis- like in experimental animals associated with the down-regulation of adenyl cyclase/cAMP/CREB, which is further responsible for a variety of neuropathogenic factors. Despite the considerable investigation of neuroprotection in curing multiple sclerosis, some complications still remain. The available medications only provide symptomatic relief but do not stop the disease progression. In this way, the development of unused beneficial methods tends to be ignored. The limitations of the current steady treatment may be because of their activity at one of the many neurotransmitters included or their failure to up direct signaling flag bearers detailed to have a vital part in neuronal sensitivity, biosynthesis of neurotransmitters and its discharge, development, and separation of the neuron, synaptic versatility and cognitive working. Therefore, the current review strictly focused on the exploration of various clinical and pre-clinical features available for multiple sclerosis to understand the pathogenic mechanisms and to introduce pharmacological interventions associated with the upregulation of intracellular adenyl cyclase/cAMP/CREB activation to ameliorate multiple sclerosis-like features.
Collapse
Affiliation(s)
- Tarun Kapoor
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
30
|
Laube R, Paramsothy S, Leong RW. Use of medications during pregnancy and breastfeeding for Crohn's disease and ulcerative colitis. Expert Opin Drug Saf 2021; 20:275-292. [PMID: 33412078 DOI: 10.1080/14740338.2021.1873948] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: The peak age of diagnosis of inflammatory bowel disease (IBD) occurs during childbearing years, therefore management of IBD during pregnancy is a frequent occurrence. Maintenance of disease remission is crucial to optimize pregnancy outcomes, and potential maternal or fetal toxicity from medications must be balanced against the risks of untreated IBD.Areas covered: This review summarizes the literature on safety and use of medications for IBD during pregnancy and lactation.Expert opinion: 5-aminosalicylates, corticosteroids and thiopurines are safe for use during pregnancy, while methotrexate and tofacitinib should only be used with extreme caution. Anti-TNF agents (except certolizumab), vedolizumab, ustekinumab and tofacitinib readily traverse the placenta via active transport, therefore theoretically may affect fetal development. Certolizumab only undergoes passive transfer across the placenta, thus has markedly lower cord blood levels making it likely the safest biologic agent for infants. There is reasonable evidence to support the safety of anti-TNF monotherapy and combination therapy during pregnancy and lactation. Vedolizumab and ustekinumab are also thought to be safe in pregnancy and lactation, while tofacitinib is generally avoided due to teratogenic effects in animal studies.
Collapse
Affiliation(s)
- Robyn Laube
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.,Department of Gastroenterology, Macquarie University Hospital, Sydney, Australia
| | - Sudarshan Paramsothy
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.,Department of Gastroenterology, Macquarie University Hospital, Sydney, Australia.,Department of Gastroenterology and Hepatology, Concord Repatriation General Hospital, Sydney, Australia
| | - Rupert W Leong
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.,Department of Gastroenterology, Macquarie University Hospital, Sydney, Australia.,Department of Gastroenterology and Hepatology, Concord Repatriation General Hospital, Sydney, Australia
| |
Collapse
|
31
|
Su B, Wu J. Phosphorylation of RIAM Activates Its Adaptor Function in Mediating Integrin Signaling. JOURNAL OF CELLULAR SIGNALING 2021; 2:103-110. [PMID: 35128538 PMCID: PMC8813058 DOI: 10.33696/signaling.2.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Integrins are cellular receptors that regulate cell adhesion and many other cellular functions. Integrins can be activated via an "inside-out pathway" that is promoted by RAP1 GTPase. RAP1-GTP-Interacting Adaptor Molecular (RIAM) mediates integrin activation by linking RAP1 GTPase to talin, an integrin activator. RIAM's function in integrin signaling is tightly regulated. In this commentary, we review recent studies of the molecular mechanisms underlying RIAM autoinhibition via both intramolecular interaction and oligomer assembly, and the phosphorylation-dependent activation of RIAM.
Collapse
Affiliation(s)
| | - Jinhua Wu
- Correspondence should be addressed to Jinhua Wu;
| |
Collapse
|
32
|
García-Fernández FJ, García-Fernández AE, Nava E, Del Pozo JSG, Ikuta I, Jordan J, Galindo MF. A bibliometric evaluation of the top 100 cited natalizumab articles. J Neuroimmunol 2020; 349:577379. [PMID: 33007648 DOI: 10.1016/j.jneuroim.2020.577379] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/05/2020] [Accepted: 08/26/2020] [Indexed: 10/23/2022]
Abstract
Natalizumab is being used in recurrent multiple sclerosis despite its history of market withdrawal due to lethal cases. We have carried out a bibliometric analysis of this drug from 1999 to February 2020 in order to assess the real impact of the use natalizumab with the goal to identify the key articles that sustain the current knowledge on the therapeutic possibilities of this compound. We have extracted from the Web of Science the top 100 most cited records (T100) and tabulated data on the journal, authors, publication year, number of citations, countries and institutions of publication, T100-records, citation density and citations per record of the works. The 100 most cited articles were selected from a total of 32,507 citations out of 2817 publications with an h-number of 74, 11.54 citations/publication, and a density of 1544.79 citations/year. Citations ranged from 63 of the paper placed in the 100th position (T100) to 1940 of the paper in the first position (T1). T2 was cited 888 times, and the difference in the number of citations between T1 and T2 was higher than that between T2 and T10. T1, T2 and T3 are clinical trials. When articles are arranged by institution and nationality having more than 10 T100 articles, biotechnology company Biogen and the USA, respectively, lead the ranking, but we also find that 8 out of 10 are academic European institutions. A co-authorship analysis reveals an intense collaborative activity between countries and institutions. We conclude that the clinical and academic communities have shown a sustained interest in natalizumab for the therapy of recurrent multiple sclerosis over the last 20 years.
Collapse
Affiliation(s)
| | | | - Eduardo Nava
- Departamento de Ciencias Médicas, Facultad de Medicina de Albacete, Universidad Castilla La Mancha
| | | | - Ichiro Ikuta
- Departamento de Radiología e Imágenes Biomédicas, Neurorradiología, Facultad de Medicina de la Universidad de Yale, New Haven, CT, USA
| | - Joaquin Jordan
- Departamento de Ciencias Médicas, Facultad de Medicina de Albacete, Universidad Castilla La Mancha
| | - Maria F Galindo
- Área Tecnología Farmacéutica, Facultad de Farmacia, Universidad Castilla La Mancha.
| |
Collapse
|
33
|
Kemmerer CL, Pernpeintner V, Ruschil C, Abdelhak A, Scholl M, Ziemann U, Krumbholz M, Hemmer B, Kowarik MC. Differential effects of disease modifying drugs on peripheral blood B cell subsets: A cross sectional study in multiple sclerosis patients treated with interferon-β, glatiramer acetate, dimethyl fumarate, fingolimod or natalizumab. PLoS One 2020; 15:e0235449. [PMID: 32716916 PMCID: PMC7384624 DOI: 10.1371/journal.pone.0235449] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Background Several disease modifying drugs (DMDs) have been approved for the treatment of multiple sclerosis (MS), however, little is known about their differential impact on peripheral blood (PB) B cell subsets. Methods We performed a cross sectional study on PB B cells in MS patients treated with interferon-β (n = 25), glatiramer acetate (n = 19), dimethyl fumarate (n = 15), fingolimod (n = 16) or natalizumab (n = 22), untreated MS patients (n = 20), and in patients with non-inflammatory neurological diseases (n = 12). Besides analyzing routine laboratory data, flow cytometry was performed to analyze naïve B cells (CD19+CD20+CD27-IgD+), non-class switched (CD19+CD20+CD27+IgD+) and class-switched memory B cells (CD19+CD20+CD27+IgD-), double negative B cells (CD19+CD20lowCD27-IgD-) and plasmablasts (CD19+CD20lowCD27+CD38++). Results Treatment associated changes were found for the overall B cell pool as well as for all B cell subsets. Natalizumab increased absolute numbers and percentage of all B cells mainly by expanding the memory B cell pool. Fingolimod decreased absolute numbers of all B cell subsets and the percentage of total B cells. Fingolimod, dimethyl fumarate and interferon-β treatments were associated with an increase in the fraction of naïve B cells while class switched and non-class switched memory B cells showed decreased percentages. Conclusion Our results highlight differential effects of DMDs on the PB B cell compartment. Across the examined treatments, a decreased percentage of memory B cells was found in dimethyl fumarate, interferon-β and fingolimod treated patients which might contribute to the drugs’ mode of action in MS. Further studies are necessary to decipher the exact role of B cell subsets during MS pathogenesis.
Collapse
Affiliation(s)
- C. L. Kemmerer
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - V. Pernpeintner
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - C. Ruschil
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - A. Abdelhak
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - M. Scholl
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - U. Ziemann
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - M. Krumbholz
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - B. Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - M. C. Kowarik
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|
34
|
Angus HCK, Butt AG, Schultz M, Kemp RA. Intestinal Organoids as a Tool for Inflammatory Bowel Disease Research. Front Med (Lausanne) 2020; 6:334. [PMID: 32010704 PMCID: PMC6978713 DOI: 10.3389/fmed.2019.00334] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/23/2019] [Indexed: 12/26/2022] Open
Abstract
Inflammatory Bowel Diseases (IBD) are difficult to model as freshly acquired tissues are short-lived, provide data as a snapshot in time, and are not always accessible. Many patients with IBD are non-responders to first-line treatments, and responders are prone to developing resistance to treatment over time—resulting in reduced patient quality of life, increased time to remission, and potential relapse. IBD is heterogenous and we are yet to fully understand the mechanisms of disease; thus, our ability to diagnose and prescribe optimal treatment remains ineffective. Intestinal organoids are derived from patient tissues expanded in vitro. Organoids offer unique insight into individual patient disease and are a potential route to personalized treatments. However, organoid models do not contain functional microbial and immune cell components. In this review, we discuss immune cell subsets in the context of IBD, and the requirement of immune cell and microbial components in organoid models for IBD research.
Collapse
Affiliation(s)
- Hamish C K Angus
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - A Grant Butt
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Michael Schultz
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - Roslyn A Kemp
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
35
|
Grand'Maison F, Yeung M, Morrow SA, Lee L, Emond F, Ward BJ, Laneuville P, Schecter R. Sequencing of disease-modifying therapies for relapsing-remitting multiple sclerosis: a theoretical approach to optimizing treatment. Curr Med Res Opin 2018; 34:1419-1430. [PMID: 29583054 DOI: 10.1080/03007995.2018.1458023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is a chronic disease which usually begins in young adulthood and is a lifelong condition. Individuals with MS experience physical and cognitive disability resulting from inflammation and demyelination in the central nervous system. Over the past decade, several disease-modifying therapies (DMTs) have been approved for the management of relapsing-remitting MS (RRMS), which is the most prevalent phenotype. The chronic nature of the disease and the multiple treatment options make benefit-risk-based sequencing of therapy essential to ensure optimal care. The efficacy and short- and long-term risks of treatment differ for each DMT due to their different mechanism of action on the immune system. While transitioning between DMTs, in addition to immune system effects, factors such as age, disease duration and severity, disability status, monitoring requirements, preference for the route of administration, and family planning play an important role. Determining a treatment strategy is therefore challenging as it requires careful consideration of the differences in efficacy, safety and tolerability, while at the same time minimizing risks of immune modulation. In this review, we discuss a sequencing approach for treating RRMS, with importance given to the long-term risks and individual preference when devising a treatment plan. Evidence-based strategies to counter breakthrough disease are also addressed.
Collapse
Affiliation(s)
| | - Michael Yeung
- b Clinical Neurosciences, Foothills Medical Centre , Calgary , Alberta , Canada
| | - Sarah A Morrow
- c London Health Sciences Center (LHSC), Western University , London , Ontario , Canada
| | - Liesly Lee
- d Department of Neurology , Sunnybrook Health Sciences Centre , Toronto , Ontario , Canada
| | - Francois Emond
- e CHU de Québec - hôpital de l'Enfant-Jésus , Quebec City , Quebec , Canada
| | - Brian J Ward
- f Department of Microbiology & Immunology , McGill University , Montreal , Quebec , Canada
| | - Pierre Laneuville
- g Department of Medicine , McGill University , Montreal , Quebec , Canada
| | - Robyn Schecter
- h Novartis Pharmaceuticals Canada Inc. , Dorval , Quebec , Canada
| |
Collapse
|
36
|
Keane RW, Dietrich WD, de Rivero Vaccari JP. Inflammasome Proteins As Biomarkers of Multiple Sclerosis. Front Neurol 2018; 9:135. [PMID: 29615953 PMCID: PMC5868457 DOI: 10.3389/fneur.2018.00135] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/23/2018] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease that affects the brain and spinal cord. The inflammasome is a multiprotein complex that contributes to the innate immune response in animal models of MS as well as in patients with the disease. Important to the care of patients with MS is the need for biomarkers that can predict disease onset, disease exacerbation, as well as response to treatment. In this study, we analyzed serum samples from 32 patients with MS and 120 age-matched controls, and provide receiver operator characteristic (ROC) curves with associated confidence intervals following analyses of serum samples from patients with MS, most of which had the relapsing-remitting form of the disease, and from healthy unaffected donors, and determine the sensitivity and specificity of inflammasome proteins as biomarkers of MS. We report that caspase-1 (1.662 ± 0.6024 difference between means), apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) (407.5 ± 35.79), and interleukin (IL)-18 (78.53 + 17.86) were elevated in the serum of MS patients when compared to controls. Interestingly, the levels of IL-1β (−0.5961 ± 0.265) were lower in the MS cohort. Importantly, the area under the curve (AUC) for ASC and caspase-1 were 0.9448 and 0.848, respectively. Taken together, these data suggest that ASC and caspase-1 could be potential candidate biomarkers for MS onset.
Collapse
Affiliation(s)
- Robert W Keane
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL, United States.,InflamaCORE, LLC, Miami, FL, United States
| | - W Dalton Dietrich
- InflamaCORE, LLC, Miami, FL, United States.,Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Juan Pablo de Rivero Vaccari
- InflamaCORE, LLC, Miami, FL, United States.,Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
37
|
TaŞKapilioĞLu ÖZ. Recent Advances in the Treatment for Multiple Sclerosis; Current New Drugs Specific for Multiple Sclerosis. Noro Psikiyatr Ars 2018; 55:S15-S20. [PMID: 30692849 PMCID: PMC6278629 DOI: 10.29399/npa.23402] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/23/2018] [Indexed: 01/16/2023] Open
Abstract
Since the first approved parenteral drug for the treatment of multiple sclerosis (MS) in 1993 (interferon [IFN] beta, and later glatiramer acetate [GA]), today there are both parenteral and oral treatment options for MS. After IFN beta preparations, glatiramer acetate was developed; and, until the approval of natalizumab in 2006, those dominated the treatment of MS. Later on, among oral drug options, cladribine made a promising entry; however, due to safety concerns, it was withdrawn soon. Afterwards, with the understanding of the role of sphingosine-1 phosphate (S1P) receptors in the pathogenesis of MS, fingolimod was approved in 2010, which was followed by other oral agents such as teriflunomide and dimethyl fumarate. Recently newer IV treatment options such as alemtuzumab, rituximab and ocrelizumab have widened the treatment arena. Recently, after submitting new efficacy and safety data, cladribine was approved for MS by EMA, in 2017. Moreover, seven years after its rejection due to safety reasons, in August 2018 FDA accepted to re-evaluate the data of cladribine as a treatment option for relapsing remitting MS (RRMS). Another oral treatment option, Laquinimod, was not approved because it could not be shown to slow disability progression despite favourable effect in relapsing MS. Newer generation S1P receptor modulators are being investigated currently, and they are expected to come into the treatment arena soon. In this article, mechanisms of actions, clinical trial results, and side effects of the newer drugs used for MS, are reviewed. IFN beta and glatiramer acetate were not included since they have clinical experience nearing 30 years.
Collapse
Affiliation(s)
- ÖZlem TaŞKapilioĞLu
- Department of Neurology, Mehmet Ali Aydınlar Acıbadem University School of Medicine, İstanbul, Turkey
| |
Collapse
|
38
|
Sabol RA, Noxon V, Sartor O, Berger JR, Qureshi Z, Raisch DW, Norris LB, Yarnold PR, Georgantopoulos P, Hrushesky WJ, Bobolts L, Ray P, Lebby A, Kane RC, Bennett CL. Melanoma complicating treatment with natalizumab for multiple sclerosis: A report from the Southern Network on Adverse Reactions (SONAR). Cancer Med 2017. [PMID: 28635055 PMCID: PMC5504343 DOI: 10.1002/cam4.1098] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
A 43-year-old female with multiple sclerosis developed urethral melanoma. The only potential risk factor was treatment with natalizumab, a humanized monoclonal antibody against α4 integrins. To investigate the risk-exposure relationship, we reviewed this case, all other published cases, and cases of natalizumab-associated melanoma reported to regulatory agencies. Data sources included the Food and Drug Administration's (FDA) Adverse Event Reporting System (FAERS) (2004-2014), a FDA Advisory Committee Meeting Report, and peer-reviewed publications. In the United States, the manufacturer maintains an FDA-mandated Tysabri Safety Surveillance Program (part of the Tysabri Outcomes Unified Commitment to Health (TOUCH)) of natalizumab-treated patients. We statistically compared reporting completeness for natalizumab-associated melanoma cases in FAERs for which information was obtained entirely from the TOUCH program versus cases where FAERS information was supplemented by TOUCH program information. FAERS included 137 natalizumab-associated melanoma reports in patients with multiple sclerosis. Median age at melanoma diagnosis was 45 years (range: 21-74 years). Changes in preexisting nevi occurred in 16%, history of cutaneous nevi occurred in 22%, diagnosis within 2 years of beginning natalizumab occurred in 34%, and 74% had primary surgical treatment. Among seven natalizumab-treated MS patients who developed biopsy-confirmed melanoma on treatment and reported in the literature, median age at diagnosis was 41 years (range: 38-48 years); and the melanoma diagnosis occurred following a median of 12 natalizumab doses (range: 1-77 doses). A history of mole or nevi was noted in four patients and a history of prior melanoma was noted in one patient. Completeness scores for reports were significantly lower for FAERS cases reported from the TOUCH program versus FAERS cases supplemented by TOUCH information (median score of 2 vs. 4 items out of 8-possible items, P < 0.0007). Clinicians should monitor existing nevi and maintain suspicion for melanoma developing in natalizumab-treated patients. The TOUCH Safety Surveillance Program, currently focused on progressive multifocal leukoencephalopathy, should be expanded to include information on other serious complications including malignancies, particularly if they are immunologic in nature.
Collapse
Affiliation(s)
- Rachel A Sabol
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Virginia Noxon
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Oliver Sartor
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Joseph R Berger
- Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Zaina Qureshi
- The Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Dennis W Raisch
- University of New Mexico, College of Pharmacy, Albuquerque, New Mexico
| | - LeAnn B Norris
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Paul R Yarnold
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Peter Georgantopoulos
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - William J Hrushesky
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | | | - Paul Ray
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Akida Lebby
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Robert C Kane
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina
| | - Charles L Bennett
- The Southern Network on Adverse Reactions (SONAR) program, University of South Carolina College of Pharmacy, Columbia, South Carolina.,The Medical University of South Carolina Hollings Cancer Center, Charleston, South Carolina.,William Jennings Bryan Dorn Veterans Administration Medical Center, Columbia, South Carolina
| |
Collapse
|
39
|
Chen N, Peine KJ, Collier MA, Gautam S, Jablonski KA, Guerau-de-Arellano M, Ainslie KM, Bachelder EM. Co-Delivery of Disease Associated Peptide and Rapamycin via Acetalated Dextran Microparticles for Treatment of Multiple Sclerosis. ACTA ACUST UNITED AC 2017. [DOI: 10.1002/adbi.201700022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Naihan Chen
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Kevin J. Peine
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Michael A. Collier
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Shalini Gautam
- Division of Pharmaceutics; College of Pharmacy; The Ohio State University; Columbus OH 43210 USA
| | - Kyle A. Jablonski
- Medical Laboratory Science Division; School of Health and Rehabilitation Sciences; The Ohio State University; Columbus OH 43210 USA
| | - Mireia Guerau-de-Arellano
- Medical Laboratory Science Division; School of Health and Rehabilitation Sciences; The Ohio State University; Columbus OH 43210 USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| |
Collapse
|
40
|
Davydovskaya MV, Khachanova NV, Evdoshenko EP, Pronin IN, Boiko AN, Zakharova MN, Alifirova VM, Turova EA, Malkova NA, Sivertseva SA, Tsukurova LA, Skoromets AA, Solodun IY. [Recommendations on the algorithms for drug choice and risk management plan in the treatment of patients with remitting multiple sclerosis with natalizumab]. Zh Nevrol Psikhiatr Im S S Korsakova 2017; 116:79-97. [PMID: 28139616 DOI: 10.17116/jnevro201611610279-97] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- M V Davydovskaya
- Pirogov Russian National Research Medical University, Moscow, Russia; Center of Coordination and Clinical Examination of Medical Drugs, Moscow Department of Public Health, Moscow, Russia
| | - N V Khachanova
- Pirogov Russian National Research Medical University, Moscow, Russia; Center of Coordination and Clinical Examination of Medical Drugs, Moscow Department of Public Health, Moscow, Russia
| | - E P Evdoshenko
- Center of Coordination and Clinical Examination of Medical Drugs, Moscow Department of Public Health, Moscow, Russia; City Center of Multiple Sclerosis And Autoimmune Diseases at Hospital #31, Moscow, Russia
| | - I N Pronin
- Burdenko Research Institute of Neurosurgery, Moscow, Russia
| | - A N Boiko
- Pirogov Russian National Research Medical University, Moscow, Russia
| | | | | | - E A Turova
- Siberian Regional Hospital #1, Yekaterinburg, Russia
| | - N A Malkova
- Novosibirsk State Medical University, Novosibirsk, Russia
| | | | - L A Tsukurova
- Research Institute - Ochapovsky Regional Hospital #1, Krasnodar, Russia
| | - A A Skoromets
- Pavlov St. Petersburg State Medical University, St. Petersburg, Russia
| | - I Yu Solodun
- Center of Coordination and Clinical Examination of Medical Drugs, Moscow Department of Public Health, Moscow, Russia
| |
Collapse
|
41
|
Sood AB, Kumar G, Robinson J. Bilateral acute retinal necrosis in a patient with multiple sclerosis on natalizumab. J Ophthalmic Inflamm Infect 2016; 6:26. [PMID: 27439780 PMCID: PMC4954796 DOI: 10.1186/s12348-016-0095-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/11/2016] [Indexed: 12/20/2022] Open
Abstract
Purpose The purpose of this study is to report a case of bilateral acute retinal necrosis in a patient with multiple sclerosis treated with natalizumab. Methods This study is a case report and literature review. Results A 34-year-old Caucasian female with multiple sclerosis presented with 1 week of blurry vision in both eyes during treatment with natalizumab. Clinical examination revealed bilateral acute retinal necrosis. The patient was treated with systemic intravenous acyclovir and intravitreal injections foscarnet and ganciclovir. Natalizumab therapy was also discontinued. Conclusions Natalizumab is a potent immunosuppressive agent used in relapsing remitting multiple sclerosis and Crohn’s disease. The use of this medication is commonly associated with opportunistic infections in the CNS. In rare cases, ocular opportunistic infections may occur and can lead to significant visual impairment and blindness. Neurologists and ophthalmologists should be aware of this potential complication.
Collapse
Affiliation(s)
- Arjun B Sood
- Department of Ophthalmology, Emory University, Atlanta, GA, USA. .,Emory Eye Center, Atlanta, GA, USA.
| | - Gokul Kumar
- Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Emory Eye Center, Atlanta, GA, USA
| | - Joshua Robinson
- Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Emory Eye Center, Atlanta, GA, USA
| |
Collapse
|
42
|
Past, Present, and Future of Antifungal Drug Development. TOPICS IN MEDICINAL CHEMISTRY 2016. [DOI: 10.1007/7355_2016_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
43
|
Detchokul S, Frauman AG. Editorial: cell movement. Br J Pharmacol 2015; 171:5459-61. [PMID: 25442220 DOI: 10.1111/bph.12849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
UNLABELLED Cell movement is a fundamental process of normal cellular physiology and pathophysiology. Abnormal regulation of cell migration is a common denominator of many medical disorders, including cancer metastasis, autoimmune disease and inflammation. Increased interest in the targeting of cell migration and invasion, which has potential for therapeutic intervention in many diseases are behind this special themed issue. Thus, the focus of this issue is centred on the control of cellular cytoskeletal dynamics and cellular or tissue microenvironment sensors. Novel therapeutic opportunities targeting regulation of cell migration are discussed including the emerging roles of tetraspanins, phosphoinositides, transient receptor potential cation channels, stromal interaction molecules and calcium release-activated calcium modulators. Better understanding of these regulatory factors will hopefully bring greater attention to strategically targeting aberrant cell migration, which has many therapeutic implications for common human diseases. LINKED ARTICLES This article is part of a themed section on Cytoskeleton, Extracellular Matrix, Cell Migration, Wound Healing and Related Topics. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-24
Collapse
Affiliation(s)
- S Detchokul
- Clinical Pharmacology and Therapeutics Unit, Department of Medicine, The University of Melbourne, Heidelberg, VIC, Australia
| | | |
Collapse
|
44
|
Early implementation of QbD in biopharmaceutical development: a practical example. BIOMED RESEARCH INTERNATIONAL 2015; 2015:605427. [PMID: 26075248 PMCID: PMC4449898 DOI: 10.1155/2015/605427] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/13/2015] [Accepted: 02/15/2015] [Indexed: 02/08/2023]
Abstract
In drug development, the “onus” of the low R&D efficiency has been put traditionally onto the drug discovery process (i.e., finding the right target or “binding” functionality). Here, we show that manufacturing is not only a central component of product success, but also that, by integrating manufacturing and discovery activities in a “holistic” interpretation of QbD methodologies, we could expect to increase the efficiency of the drug discovery process as a whole. In this new context, early risk assessment, using developability methodologies and computational methods in particular, can assist in reducing risks during development in a cost-effective way. We define specific areas of risk and how they can impact product quality in a broad sense, including essential aspects such as product efficacy and patient safety. Emerging industry practices around developability are introduced, including some specific examples of applications to biotherapeutics. Furthermore, we suggest some potential workflows to illustrate how developability strategies can be introduced in practical terms during early drug development in order to mitigate risks, reduce drug attrition and ultimately increase the robustness of the biopharmaceutical supply chain. Finally, we also discuss how the implementation of such methodologies could accelerate the access of new therapeutic treatments to patients in the clinic.
Collapse
|
45
|
Yang W, Xiao L, Li C, Liu X, Liu M, Shao Q, Wang D, Huang A, He C. TIP30 inhibits oligodendrocyte precursor cell differentiation via cytoplasmic sequestration of Olig1. Glia 2014; 63:684-98. [PMID: 25530119 DOI: 10.1002/glia.22778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 11/27/2014] [Accepted: 12/03/2014] [Indexed: 12/24/2022]
Abstract
Differentiation of oligodendrocyte precursor cells (OPCs) is a prerequisite for both developmental myelination and adult remyelination in the central nervous system. The molecular mechanisms underlying OPC differentiation remain largely unknown. Here, we show that the thirty-kDa HIV-1 Tat interacting protein (TIP30) is a negative regulator in oligodendrocyte development. The TIP30(-/-) mice displayed an increased myelin protein level at postnatal day 14 and 21. By using a primary OPC culture system, we demonstrated that overexpression of TIP30 dramatically inhibited the stage progression of differentiating OPCs, while knockdown of TIP30 enhanced the differentiation of oligodendroglial cells remarkably. Moreover, overexpression of TIP30 was found to sequester the transcription factor Olig1 in the cytoplasm and weaken its nuclear translocation due to the interaction between TIP30 and Olig1, whereas knockdown of TIP30 led to more Olig1 localized in the nucleus in the initiation stage during OPC differentiation. In the cuprizone-induced demyelination model, there was a dramatic increase in NG2-expressing cells with nuclear location of Olig1 in the corpus callosum during remyelination. In contrast, within chronic demyelinated lesions in multiple sclerosis, TIP30 was abnormally expressed in NG2-expressing cells, and few nuclear Olig1 was observed in these cells. Taken together, our findings suggest that TIP30 plays a negative regulatory role in oligodendroglial differentiation.
Collapse
Affiliation(s)
- Wenjing Yang
- Institute of Neuroscience and MOE Key Laboratory of Molecular Neurobiology, Neuroscience Research Center of Changzheng Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sáenz-Cuesta M, Osorio-Querejeta I, Otaegui D. Extracellular Vesicles in Multiple Sclerosis: What are They Telling Us? Front Cell Neurosci 2014; 8:100. [PMID: 24734004 PMCID: PMC3975116 DOI: 10.3389/fncel.2014.00100] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/17/2014] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound particles secreted by almost all cell types. They are classified depending on their biogenesis and size into exosomes and microvesicles or according to their cell origin. EVs play a role in cell-to-cell communication, including contact-free cell synapsis, carrying active membrane proteins, lipids, and genetic material both inside the particle and on their surface. They have been related to several physiological and pathological conditions. In particular, increasing concentrations of EVs have been found in many autoimmune diseases including multiple sclerosis (MS). MS is a central nervous system (CNS) demyelinating disease characterized by relapsing of symptoms followed by periods of remission. Close interaction between endothelial cells, leukocytes, monocytes, and cells from CNS is crucial for the development of MS. This review summarizes the pathological role of EVs in MS and the relationship of EVs with clinical characteristics, therapy, and biomarkers of the disease.
Collapse
Affiliation(s)
- Matías Sáenz-Cuesta
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain ; Spanish Network on Multiple Sclerosis , Madrid , Spain
| | - Iñaki Osorio-Querejeta
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain ; Spanish Network on Multiple Sclerosis , Madrid , Spain
| | - David Otaegui
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain ; Spanish Network on Multiple Sclerosis , Madrid , Spain
| |
Collapse
|
47
|
Sternberg Z, Leung C, Sternberg D, Yu J, Hojnacki D. Disease Modifying Therapies Modulate Cardiovascular Risk Factors in Patients with Multiple Sclerosis. Cardiovasc Ther 2014; 32:33-9. [DOI: 10.1111/1755-5922.12049] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Zohara Sternberg
- Department of Neurology; Stroke Center; Buffalo Medical Center; Buffalo NY USA
| | - Christopher Leung
- Department of Neurology; Stroke Center; Buffalo Medical Center; Buffalo NY USA
| | - Daniel Sternberg
- Department of Neurology; Stroke Center; Buffalo Medical Center; Buffalo NY USA
| | - Jinhee Yu
- Department of Biostatistics; University of Buffalo; Buffalo NY USA
| | - David Hojnacki
- Department of Neurology; Stroke Center; Buffalo Medical Center; Buffalo NY USA
| |
Collapse
|
48
|
Knier B, Hemmer B, Korn T. Novel monoclonal antibodies for therapy of multiple sclerosis. Expert Opin Biol Ther 2014; 14:503-13. [DOI: 10.1517/14712598.2014.887676] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Peine KJ, Guerau-de-Arellano M, Lee P, Kanthamneni N, Severin M, Probst GD, Peng H, Yang Y, Vangundy Z, Papenfuss TL, Lovett-Racke AE, Bachelder EM, Ainslie KM. Treatment of experimental autoimmune encephalomyelitis by codelivery of disease associated Peptide and dexamethasone in acetalated dextran microparticles. Mol Pharm 2014; 11:828-35. [PMID: 24433027 PMCID: PMC3993881 DOI: 10.1021/mp4005172] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune, demyelinating disease of the central nervous system that can cause loss of motor function and is thought to result, in part, from chronic inflammation due to an antigen-specific T cell immune response. Current treatments suppress the immune system without antigen specificity, increasing the risks of cancer, chronic infection, and other long-term side effects. In this study, we show treatment of experimental autoimmune encephalomyelitis (EAE), a model of MS, by coencapsulating the immunodominant peptide of myelin oligodendrocyte glycoprotein (MOG) with dexamethasone (DXM) into acetalated dextran (Ac-DEX) microparticles (DXM/MOG/MPs) and administering the microparticles subcutaneously. The clinical score of the mice was reduced from 3.4 to 1.6 after 3 injections 3 days apart with the coencapsulated microparticulate formulation (MOG 17.6 μg and DXM 8 μg). This change in clinical score was significantly greater than observed with phosphate-buffered saline (PBS), empty MPs, free DXM and MOG, DXM/MPs, and MOG/MPs. Additionally, treatment with DXM/MOG/MPs significantly inhibited disease-associated cytokine (e.g., IL-17, GM-CSF) expression in splenocytes isolated in treated mice. Here we show a promising approach for the therapeutic treatment of MS using a polymer-based microparticle delivery platform.
Collapse
Affiliation(s)
- Kevin J Peine
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University , Columbus, Ohio, 43210, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Aung WY, Mar S, Benzinger TL. Diffusion tensor MRI as a biomarker in axonal and myelin damage. ACTA ACUST UNITED AC 2013; 5:427-440. [PMID: 24795779 DOI: 10.2217/iim.13.49] [Citation(s) in RCA: 231] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Diffusion tensor imaging has been used extensively as a research tool to understand the structural changes associated with white matter pathology. Using water diffusion as the basis to construct anatomic details, diffusion tensor imaging offers the potential to identify structural and functional adaptations before gross anatomical changes, such as lesions and tumors, become apparent on conventional MRI. Over the past 10 years, further parameters, such as axial and radial diffusivity, have been developed to characterize white matter changes specific to axons and myelin. In this paper, the potential application and outstanding issues on the use of diffusion tensor imaging directional diffusivity as a biomarker in axonal and myelin damage in neurological disorders will be reviewed.
Collapse
Affiliation(s)
- Wint Yan Aung
- Department of Radiology, Washington University, School of Medicine, 510 South Kingshighway Boulevard, St Louis, MO 63110, USA
| | - Soe Mar
- Department of Pediatric & Developmental Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Tammie Ls Benzinger
- Department of Radiology, Washington University, School of Medicine, 510 South Kingshighway Boulevard, St Louis, MO 63110, USA ; Department of Neurological Surgery, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|