1
|
Pavan C, Davidson KC, Payne N, Frausin S, Hunt CPJ, Moriarty N, Berrocal Rubio MÁ, Elahi Z, Quattrocchi AT, Abu-Bonsrah KD, Wang L, Clow W, Yang H, Pellegrini M, Wells CA, Thompson LH, Nagy A, Parish CL. A cloaked human stem-cell-derived neural graft capable of functional integration and immune evasion in rodent models. Cell Stem Cell 2025; 32:710-726.e8. [PMID: 40209717 DOI: 10.1016/j.stem.2025.03.008] [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: 04/05/2024] [Revised: 11/13/2024] [Accepted: 03/13/2025] [Indexed: 04/12/2025]
Abstract
Human pluripotent stem cell (hPSC)-derived therapies are a realistic possibility for numerous disorders, including Parkinson's disease. While generating replacement neurons is achievable, immunosuppressive drug challenges, to prevent rejection, remain. Here we adopted a hPSC line (termed H1-FS-8IM), engineered to overexpress 8 immunomodulatory transgenes, to enable transplant immune evasion. In co-cultures, H1-FS-8IM PSC-derived midbrain neurons evaded rejection by T lymphocytes, natural killer cells, macrophages, and dendritic cells. In humanized mice, allogeneic H1-FS-8IM neural grafts evaded rejection, while control hPSC-derived neural grafts evoked activation of human immune cells, elevated inflammatory cytokines in blood and cerebrospinal fluid, and caused spleen and lymph node enlargement. H1-FS-8IM neural grafts retained functionality, reversing motor deficits in Parkinsonian rats. Additional incorporation of a suicide gene into the H1-FS-8IM hPSC line enabled proliferative cell elimination within grafts. Findings demonstrate feasibility of generating a population-wide applicable, safe, off-the-shelf cell product, suitable for treating diseases for which cell-based therapies are a viable option.
Collapse
Affiliation(s)
- Chiara Pavan
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
| | - Kathryn C Davidson
- Infectious Diseases & Immune Defence Division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Natalie Payne
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Stefano Frausin
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Cameron P J Hunt
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Niamh Moriarty
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | | | - Zahra Elahi
- Department of Anatomy & Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Andrew T Quattrocchi
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | | | - Le Wang
- Infectious Diseases & Immune Defence Division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - William Clow
- Infectious Diseases & Immune Defence Division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Huijuan Yang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Marc Pellegrini
- Centenary Institute of Cancer Medicine and Cell Biology, Sydney, NSW, Australia
| | - Christine A Wells
- Department of Anatomy & Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Lachlan H Thompson
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; Faculty of Medicine and Health, School of Medical Sciences & Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Andras Nagy
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; Department of Anatomy & Physiology, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
2
|
Izadifar M, Massumi M, Prentice KJ, Oussenko T, Li B, Elbaz J, Puri M, Wheeler MB, Nagy A. Microfluidic chip systems for characterizing glucose-responsive insulin-secreting cells equipped with FailSafe kill-switch. Stem Cell Res Ther 2024; 15:486. [PMID: 39696686 DOI: 10.1186/s13287-024-04059-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/10/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Pluripotent cell-derived islet replacement therapy offers promise for treating Type 1 diabetes (T1D), but concerns about uncontrolled cell proliferation and tumorigenicity present significant safety challenges. To address the safety concern, this study aims to establish a proof-of-concept for a glucose-responsive, insulin-secreting cell line integrated with a built-in FailSafe kill-switch. METHOD We generated β cell-induced progenitor-like cells (βiPLCs) from primary mouse pancreatic β cells through interrupted reprogramming. Then, we transcriptionally linked our FailSafe (FS) kill-switch, HSV-thymidine kinase (TK), to Cdk1 gene using a CRISPR/Cas9 knock-in strategy, resulting in a FailSafe βiPLC line, designated as FSβiPLCs. Subsequently we evaluated and confirmed the functionality of the drug-inducible kill-switch in FSβiPLCs at different ganciclovir (GCV) concentrations using our PDMS-based transcapillary microfluidic system. Finally, we assessed the functionality of FSβiPLCs by characterizing the dynamics of insulin secretion in response to changes in glucose concentration using our microfluidic perfusion glucose-stimulated insulin secretion (GSIS) assay-on- chip. RESULTS The βiPLCs exhibited Ins1, Pdx1 and Nkx6.1 expression, and glucose responsive insulin secretion, the essential properties of pancreatic beta cells. The βiPLCs were amenable to genome editing which allowed for the insertion of the kill-switch into the 3'UTR of Cdk1, confirmed by PCR genotyping. Our transcapillary microfluidic system confirmed the functionality of the drug-inducible kill-switch in FSβiPLCs, showing an effective cell ablation of dividing cells from a heterogeneous cell population at different ganciclovir (GCV) concentrations. The Ki67 expression assessment further confirmed that slow- or non-dividing cells in the FSβiPLC population were resistant to GCV. Our perfusion glucose-stimulated insulin secretion (GSIS) assay-on-chip revealed that the resistant non-dividing FSβiPLCs exhibited higher levels of insulin secretion and glucose responsiveness compared to their proliferating counterparts. CONCLUSIONS This study establishes a proof-of-concept for the integration of a FailSafe kill-switch system into a glucose-responsive, insulin-secreting cell line to address the safety concerns in stem cell-derived cell replacement treatment for T1D. The microfluidic systems provided valuable insights into the functionality and safety of these engineered cells, demonstrating the potential of the kill-switch to reduce the risk of tumorigenicity in pluripotent cell-derived insulin-secreting cells.
Collapse
Affiliation(s)
- Mohammad Izadifar
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Mohammad Massumi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Kacey J Prentice
- Departments of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tatiana Oussenko
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Biao Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Judith Elbaz
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Mira Puri
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Michael B Wheeler
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Chan F, Hardy TA, Malik S, Ramanathan S, Riminton DS, Reddel SW. Induction cyclophosphamide with maintenance immunosuppression in high-risk myasthenia gravis: long-term follow-up and safety profile. J Neurol Neurosurg Psychiatry 2024; 95:1096-1101. [PMID: 38964847 DOI: 10.1136/jnnp-2023-333189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/10/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Patients with refractory or high-risk myasthenia gravis (MG) respond poorly to conventional immunosuppressive therapy, requiring rescue therapies and often experiencing treatment toxicity. Rescue and injectable therapies do not induce remission and require repetitive administration leading to significant constraints on patients and the healthcare system. This long-term follow-up study demonstrates cyclophosphamide as a rapidly effective and safe treatment in patients with refractory or high-risk MG. METHODS Retrospective cohort study of MG patients treated with cyclophosphamide between January 2000 and June 2022 conducted at a quaternary neuroimmunology clinic in New South Wales, Australia. RESULTS 31 patients were treated: mean age of 64 years; median follow-up 3.6 years (5 months to 11 years); 94% seropositive to acetylcholine receptor (AChR) antibodies and 45% had thymoma. A reduced intensity cyclophosphamide induction protocol followed by oral antiproliferative maintenance is described.Median myasthenia gravis composite scores reduced by >50% after the third cycle of cyclophosphamide. Complete cessation of prednisolone was possible in 11 patients while 20 remained on prednisolone with a median daily dose of 5 mg. Plasma exchange was ceased in 62% of patients and intravenous immunoglobulin ceased in 55%. Cyclophosphamide was generally well tolerated with mild cytopenias. There were no malignancies or cases of haemorrhagic cystitis. CONCLUSION We describe a large cohort of high-risk MG patients treated with cyclophosphamide in a retrospective single-clinic cohort. We suggest cyclophosphamide should be considered for rapid remission induction, corticosteroid reduction and long-term freedom from recurrent injectable therapies in selected patients, typically those with AChR antibodies.
Collapse
Affiliation(s)
- Fiona Chan
- Queen Elizabeth Hospital, Birmingham, UK
| | - Todd A Hardy
- Department of Neurology, Concord Hospital, Concord, New South Wales, Australia
| | - Sameer Malik
- Department of Immunology, Concord Hospital, Concord, New South Wales, Australia
| | - Sudarshini Ramanathan
- Department of Neurology, Concord Hospital, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - D Sean Riminton
- Department of Immunology, Concord Hospital, Concord, New South Wales, Australia
| | | |
Collapse
|
4
|
Mahadeen AZ, Carlson AK, Cohen JA, Galioto R, Abbatemarco JR, Kunchok A. Review of the Longitudinal Management of Autoimmune Encephalitis, Potential Biomarkers, and Novel Therapeutics. Neurol Clin Pract 2024; 14:e200306. [PMID: 38831758 PMCID: PMC11145747 DOI: 10.1212/cpj.0000000000200306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 02/08/2024] [Indexed: 06/05/2024]
Abstract
Purpose of Review Increasing awareness and earlier diagnosis of autoimmune encephalitis (AE) have led to a greater number of patients being cared for longitudinally by neurologists. Although many neurologists are now familiar with the general approach to diagnosis and acute immunosuppression, this review aims to provide neurologists with guidance related to management beyond the acute phase of disease, including long-term immunosuppression, monitoring, potential biomarkers of disease activity, outcome measures, and symptom management. Recent Findings Observational studies in AE have demonstrated that early diagnosis and treatment is associated with improved neurologic outcomes, particularly in AE with antibodies targeting neuronal cell surface/synaptic proteins. The literature regarding long-term management is evolving. In addition to traditional immunosuppressive approaches, there is emerging use of novel immunosuppressive therapies (ISTs) in case series, and several randomized controlled trials are planned. Novel biomarkers of disease activity and methods to measure outcomes and response to treatment are being explored. Furthermore, it is increasingly recognized that many individuals have chronic symptoms affecting quality of life including seizures, cognitive impairment, fatigue, sleep disorders, and mood disorders, and there are emerging data supporting the use of patient centered outcome measures and multidisciplinary symptom-based care. Summary This review aims to summarize recent literature and offer a practical approach to long-term management of adult patients with AE through a multidisciplinary approach. We summarize current knowledge on ISTs, potential biomarkers of disease activity, outcome measures, and long-term sequelae. Further research is needed to answer questions regarding optimal IST, biomarker validity, and sequelae of disease.
Collapse
Affiliation(s)
- Ahmad Z Mahadeen
- Department of Neurology (AZM), University of Mississippi Medical Center, Jackson; and Cleveland Clinic Mellen Center (AKC, JAC, RG, JRA, AK), OH
| | - Alise K Carlson
- Department of Neurology (AZM), University of Mississippi Medical Center, Jackson; and Cleveland Clinic Mellen Center (AKC, JAC, RG, JRA, AK), OH
| | - Jeffrey A Cohen
- Department of Neurology (AZM), University of Mississippi Medical Center, Jackson; and Cleveland Clinic Mellen Center (AKC, JAC, RG, JRA, AK), OH
| | - Rachel Galioto
- Department of Neurology (AZM), University of Mississippi Medical Center, Jackson; and Cleveland Clinic Mellen Center (AKC, JAC, RG, JRA, AK), OH
| | - Justin R Abbatemarco
- Department of Neurology (AZM), University of Mississippi Medical Center, Jackson; and Cleveland Clinic Mellen Center (AKC, JAC, RG, JRA, AK), OH
| | - Amy Kunchok
- Department of Neurology (AZM), University of Mississippi Medical Center, Jackson; and Cleveland Clinic Mellen Center (AKC, JAC, RG, JRA, AK), OH
| |
Collapse
|
5
|
Harding J, Vintersten-Nagy K, Yang H, Tang JK, Shutova M, Jong ED, Lee JH, Massumi M, Oussenko T, Izadifar Z, Zhang P, Rogers IM, Wheeler MB, Lye SJ, Sung HK, Li C, Izadifar M, Nagy A. Immune-privileged tissues formed from immunologically cloaked mouse embryonic stem cells survive long term in allogeneic hosts. Nat Biomed Eng 2024; 8:427-442. [PMID: 37996616 PMCID: PMC11087263 DOI: 10.1038/s41551-023-01133-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/30/2023] [Indexed: 11/25/2023]
Abstract
The immunogenicity of transplanted allogeneic cells and tissues is a major hurdle to the advancement of cell therapies. Here we show that the overexpression of eight immunomodulatory transgenes (Pdl1, Cd200, Cd47, H2-M3, Fasl, Serpinb9, Ccl21 and Mfge8) in mouse embryonic stem cells (mESCs) is sufficient to immunologically 'cloak' the cells as well as tissues derived from them, allowing their survival for months in outbred and allogeneic inbred recipients. Overexpression of the human orthologues of these genes in human ESCs abolished the activation of allogeneic human peripheral blood mononuclear cells and their inflammatory responses. Moreover, by using the previously reported FailSafe transgene system, which transcriptionally links a gene essential for cell division with an inducible and cell-proliferation-dependent kill switch, we generated cloaked tissues from mESCs that served as immune-privileged subcutaneous sites that protected uncloaked allogeneic and xenogeneic cells from rejection in immune-competent hosts. The combination of cloaking and FailSafe technologies may allow for the generation of safe and allogeneically accepted cell lines and off-the-shelf cell products.
Collapse
Affiliation(s)
- Jeffrey Harding
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Kristina Vintersten-Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Huijuan Yang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jean Kit Tang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Maria Shutova
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Eric D Jong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Ju Hee Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mohammad Massumi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Tatiana Oussenko
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Zohreh Izadifar
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Puzheng Zhang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Ian M Rogers
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | - Michael B Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Stephen J Lye
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - ChengJin Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Mohammad Izadifar
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Increasingly, therapeutic strategy in multiple sclerosis (MS) is informed by imaging and laboratory biomarkers, in addition to traditional clinical factors. Here, we review aspects of monitoring the efficacy and risks of disease-modifying therapy (DMT) with both conventional and emerging MRI and laboratory measures. RECENT FINDINGS The adoption of consensus-driven, stable MRI acquisition protocols and artificial intelligence-based, quantitative image analysis is heralding an era of precision monitoring of DMT efficacy. New MRI measures of compartmentalized inflammation, neuro-degeneration and repair complement traditional metrics but require validation before use in individual patients. Laboratory markers of brain cellular injury, such as neurofilament light, are robust outcomes in DMT efficacy trials; their use in clinical practice is being refined. DMT-specific laboratory monitoring for safety is critical and may include lymphocytes, immunoglobulins, autoimmunity surveillance, John Cunningham virus serology and COVID-19 vaccination seroresponse. SUMMARY A biomarker-enhanced monitoring strategy has immediate clinical application, with growing evidence of long-term reductions in disability accrual when both clinically symptomatic and asymptomatic inflammatory activity is fully suppressed; and amelioration of the risks associated with therapy. Emerging MRI and blood-based measures will also become important tools for monitoring agents that target the innate immune system and promote neuro-repair.
Collapse
|
7
|
Fričová D, Korchak JA, Zubair AC. Challenges and translational considerations of mesenchymal stem/stromal cell therapy for Parkinson's disease. NPJ Regen Med 2020; 5:20. [PMID: 33298940 PMCID: PMC7641157 DOI: 10.1038/s41536-020-00106-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta and the presence of Lewy bodies, which gives rise to motor and non-motor symptoms. Unfortunately, current therapeutic strategies for PD merely treat the symptoms of the disease, only temporarily improve the patients' quality of life, and are not sufficient for completely alleviating the symptoms. Therefore, cell-based therapies have emerged as a novel promising therapeutic approach in PD treatment. Mesenchymal stem/stromal cells (MSCs) have arisen as a leading contender for cell sources due to their regenerative and immunomodulatory capabilities, limited ethical concerns, and low risk of tumor formation. Although several studies have shown that MSCs have the potential to mitigate the neurodegenerative pathology of PD, variabilities in preclinical and clinical trials have resulted in inconsistent therapeutic outcomes. In this review, we strive to highlight the sources of variability in studies using MSCs in PD therapy, including MSC sources, the use of autologous or allogenic MSCs, dose, delivery methods, patient factors, and measures of clinical outcome. Available evidence indicates that while the use of MSCs in PD has largely been promising, conditions need to be standardized so that studies can be effectively compared with one another and experimental designs can be improved upon, such that this body of science can continue to move forward.
Collapse
Affiliation(s)
- Dominika Fričová
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Jennifer A Korchak
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Abba C Zubair
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
8
|
Heckmann JM, Marais S. Management Issues in Myasthenia Gravis Patients Living With HIV: A Case Series and Literature Review. Front Neurol 2020; 11:775. [PMID: 32973647 PMCID: PMC7472955 DOI: 10.3389/fneur.2020.00775] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022] Open
Abstract
South Africa is home to more than seven million people living with human immunodeficiency virus (HIV) and a high prevalence of tuberculosis. Human immunodeficiency virus–infected individuals may develop myasthenia gravis (MG), which raises questions regarding their management. An MG database, with 24 years of observational data, was audited for HIV-infected persons. Case reports of MG in HIV-infected persons were reviewed. We identified 17 persons with MG and HIV infection. All had generalized MG with a mean age at onset of 37.8 years. Eleven had acetylcholine receptor antibody–positive MG; one had antibodies against muscle-specific kinase. Six developed MG prior to HIV infection (mean CD4+ 361 cells/mm3); four worsened <6 months of starting antiretrovirals. Eleven developed MG while HIV-infected (mean CD4+ 423 cells/mm3); five presented with mild MG; three in MG crisis requiring rescue therapies (intravenous immune globulin or plasma exchange and/or intravenous cyclophosphamide). Two were diagnosed with HIV infection and MG at the same time. Fifteen required maintenance steroid-sparing immune therapies, predominantly azathioprine, or methotrexate. Plasma HIV viral loads remained below detectable levels on antiretrovirals during immunosuppressant treatment. Over the average follow-up of 6 years, 10 achieved minimal manifestation status, and the remainder improved to mild symptoms. Three cases had tuberculosis before MG, but none developed tuberculosis reactivation on immunosuppressive therapy; one used isoniazid prophylaxis. Herpes zoster reactivation during treatment occurred in one. Conclusions include the following: MG in HIV-infected patients should be managed similarly to individuals without HIV infection; half develop moderate–severe MG; MG symptoms may worsen within 6 months of antiretroviral initiation; safety monitoring must include plasma HIV viral load estimation. Isoniazid prophylaxis may not be indicated in all cases.
Collapse
Affiliation(s)
- Jeannine M Heckmann
- Neurology, Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa.,Neurology Research Group, UCT Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Suzaan Marais
- Neurology, Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa.,Neurology Research Group, UCT Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
9
|
Griffin JD, Song JY, Sestak JO, DeKosky BJ, Berkland CJ. Linking autoantigen properties to mechanisms of immunity. Adv Drug Deliv Rev 2020; 165-166:105-116. [PMID: 32325104 PMCID: PMC7572523 DOI: 10.1016/j.addr.2020.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023]
Abstract
Antigen-specific immunotherapies (ASIT) present compelling potential for introducing precision to the treatment of autoimmune diseases where nonspecific, global immunosuppression is currently the only treatment option. Central to ASIT design is the delivery of autoantigen, which parallels allergy desensitization approaches. Clinical success in tolerizing allergen-specific responses spans longer than a century, but autoimmune ASITs have yet to see an FDA-approved breakthrough. Allergens and autoantigens differ substantially in physicochemical properties, and these discrepancies influence the nature of their interactions with the immune system. Approved allergen-specific immunotherapies are typically administered as water soluble, neutrally charged protein fractions from 10 to 70 kDa. Conversely, autoantigens are native proteins that exhibit wide-ranging sizes, solubilities, and charges that render them susceptible to immunogenicity. To translate the success of allergen hyposensitization to ASIT, delivery strategies may be necessary to effectively format autoantigens, guide biodistribution, and engage appropriate immune mechanisms.
Collapse
Affiliation(s)
- J Daniel Griffin
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, United States of America; Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Jimmy Y Song
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America; Bioengineering Graduate Program, University of Kansas, Lawrence, KS, United States of America
| | - Joshua O Sestak
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America; Orion BioScience, Inc, Omaha, NE, United States of America
| | - Brandon J DeKosky
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, United States of America; Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, United States of America
| | - Cory J Berkland
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, United States of America; Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, United States of America.
| |
Collapse
|
10
|
Feng X, Xu W, Li Z, Song W, Ding J, Chen X. Immunomodulatory Nanosystems. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900101. [PMID: 31508270 PMCID: PMC6724480 DOI: 10.1002/advs.201900101] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/21/2019] [Indexed: 05/15/2023]
Abstract
Immunotherapy has emerged as an effective strategy for the prevention and treatment of a variety of diseases, including cancer, infectious diseases, inflammatory diseases, and autoimmune diseases. Immunomodulatory nanosystems can readily improve the therapeutic effects and simultaneously overcome many obstacles facing the treatment method, such as inadequate immune stimulation, off-target side effects, and bioactivity loss of immune agents during circulation. In recent years, researchers have continuously developed nanomaterials with new structures, properties, and functions. This Review provides the most recent advances of nanotechnology for immunostimulation and immunosuppression. In cancer immunotherapy, nanosystems play an essential role in immune cell activation and tumor microenvironment modulation, as well as combination with other antitumor approaches. In infectious diseases, many encouraging outcomes from using nanomaterial vaccines against viral and bacterial infections have been reported. In addition, nanoparticles also potentiate the effects of immunosuppressive immune cells for the treatment of inflammatory and autoimmune diseases. Finally, the challenges and prospects of applying nanotechnology to modulate immunotherapy are discussed.
Collapse
Affiliation(s)
- Xiangru Feng
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
- University of Science and Technology of ChinaHefei230026P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Zhongmin Li
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
- Department of Gastrointestinal Colorectal and Anal SurgeryChina–Japan Union Hospital of Jilin UniversityChangchun130033P. R. China
| | - Wantong Song
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| |
Collapse
|
11
|
Saito E, Kuo R, Kramer KR, Gohel N, Giles DA, Moore BB, Miller SD, Shea LD. Design of biodegradable nanoparticles to modulate phenotypes of antigen-presenting cells for antigen-specific treatment of autoimmune disease. Biomaterials 2019; 222:119432. [PMID: 31480002 DOI: 10.1016/j.biomaterials.2019.119432] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/18/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Current therapeutic options for autoimmune diseases, such as multiple sclerosis (MS), often require lifelong treatment with immunosuppressive drugs, yet strategies for antigen-specific immunomodulation are emerging. Biodegradable particles loaded with disease-specific antigen, either alone or with immunomodulators, have been reported to ameliorate disease. Herein, we hypothesized that the carrier could impact polarization of the immune cells that associate with particles and the subsequent disease progression. Single injection of three polymeric carriers, 50:50 poly (DL-lactide-co-glycolide) (PLG) with two molecular weights (Low, High) and poly (DL-lactide) (PLA), loaded with the disease-specific antigen, proteolipid protein (PLP139-151), were investigated for the ability to attenuate clinical scores in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. At a low particle dose, mice treated with PLA-based particles had significantly lower clinical scores at the chronic stage of the disease over 200 days post immunization, while neither PLG-based particles nor OVA control particles reduced the clinical scores. Compared to PLG-based particles, PLA-based particles were largely associated with Kupffer cells and liver sinusoidal endothelial cells, which had a reduced co-stimulatory molecule expression that correlated with a reduction of CD4+ T-cell populations in the central nervous system. Delivery of PLA-based particles encapsulated with higher levels of PLP139-151 at a reduced dose were able to completely ameliorate EAE over 200 days along with inhibition of Th1 and Th17 polarization. Collectively, our study demonstrates that the carrier properties and antigen loading determine phenotypes of immune cells in the peripheral organs, influencing the amelioration of both acute and chronic stages of autoimmunity.
Collapse
Affiliation(s)
- Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert Kuo
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kevin R Kramer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nishant Gohel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David A Giles
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Bethany B Moore
- Department of Immunology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, 60208, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, 60611, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
12
|
Klotz L, Havla J, Schwab N, Hohlfeld R, Barnett M, Reddel S, Wiendl H. Risks and risk management in modern multiple sclerosis immunotherapeutic treatment. Ther Adv Neurol Disord 2019; 12:1756286419836571. [PMID: 30967901 PMCID: PMC6444778 DOI: 10.1177/1756286419836571] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/14/2019] [Indexed: 12/13/2022] Open
Abstract
In recent years, there has been a paradigm shift in the treatment of multiple
sclerosis (MS) owing to the approval of a number of new drugs with very distinct
mechanisms of action. All approved disease-modifying drugs primarily work
directly on the immune system. However, the identification of an ‘optimal
choice’ for individual patients with regard to treatment efficacy, treatment
adherence and side-effect profile has become increasingly complex including
conceptual as well as practical considerations. Similarly, there are
peculiarities and specific requirements with regard to treatment monitoring,
especially in relation to immunosuppression, the development of secondary
immune-related complications, as well as the existence of drug-specific on- and
off-target effects. Both classical immunosuppression and selective immune
interventions generate a spectrum of potential therapy-related complications.
This article provides a comprehensive overview of available immunotherapeutics
for MS and their risks, detailing individual mechanisms of action and
side-effect profiles. Furthermore, practical recommendations for patients
treated with modern MS immunotherapeutics are provided.
Collapse
Affiliation(s)
- Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University of Münster, Building A1, Albert Schweitzer Campus 1, 48149 Münster, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, University Hospital; Data Integration for Future Medicine consortium (DIFUTURE), Ludwig-Maximilians University, Munich, Germany
| | - Nicholas Schwab
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University, Munich, Germany Munich Cluster for Systems Neurology, Ludwig-Maximilians University, Munich, Germany
| | | | - Stephen Reddel
- Brain and Mind Centre, University of Sydney, NSW, Australia
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Münster, Building A1, Albert Schweitzer Campus 1, 48149 Münster, Germany
| |
Collapse
|
13
|
Thompson JA, Schneider BJ, Brahmer J, Andrews S, Armand P, Bhatia S, Budde LE, Costa L, Davies M, Dunnington D, Ernstoff MS, Frigault M, Hoffner B, Hoimes CJ, Lacouture M, Locke F, Lunning M, Mohindra NA, Naidoo J, Olszanski AJ, Oluwole O, Patel SP, Reddy S, Ryder M, Santomasso B, Shofer S, Sosman JA, Wahidi M, Wang Y, Johnson-Chilla A, Scavone JL. Management of Immunotherapy-Related Toxicities, Version 1.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2019; 17:255-289. [DOI: 10.6004/jnccn.2019.0013] [Citation(s) in RCA: 288] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The aim of the NCCN Guidelines for Management of Immunotherapy-Related Toxicities is to provide guidance on the management of immune-related adverse events resulting from cancer immunotherapy. The NCCN Management of Immunotherapy-Related Toxicities Panel is an interdisciplinary group of representatives from NCCN Member Institutions and ASCO, consisting of medical and hematologic oncologists with expertise in a wide array of disease sites, and experts from the fields of dermatology, gastroenterology, neuro-oncology, nephrology, emergency medicine, cardiology, oncology nursing, and patient advocacy. Several panel representatives are members of the Society for Immunotherapy of Cancer (SITC). The initial version of the NCCN Guidelines was designed in general alignment with recommendations published by ASCO and SITC. The content featured in this issue is an excerpt of the recommendations for managing toxicity related to immune checkpoint blockade and a review of existing evidence. For the full version of the NCCN Guidelines, including recommendations for managing toxicities related to chimeric antigen receptor T-cell therapy, visitNCCN.org.
Collapse
Affiliation(s)
- John A. Thompson
- 1Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Julie Brahmer
- 3The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | - Shailender Bhatia
- 1Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Luciano Costa
- 7University of Alabama at Birmingham Comprehensive Cancer Center
| | | | | | | | | | | | - Christopher J. Hoimes
- 13Case Comprehensive Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | - Nisha A. Mohindra
- 16Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Jarushka Naidoo
- 3The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | | | | | | | - Jeffrey A. Sosman
- 16Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | - Yinghong Wang
- 23The University of Texas MD Anderson Cancer Center; and
| | | | | |
Collapse
|
14
|
Li B, Yuan Z, McMullen P, Xie J, Jain P, Hung HC, Xu S, Zhang P, Lin X, Wu K, Jiang S. A Chromatin-Mimetic Nanomedicine for Therapeutic Tolerance Induction. ACS NANO 2018; 12:12004-12014. [PMID: 30412375 DOI: 10.1021/acsnano.8b04314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The undesirable immune response poses a life-threatening challenge to human health. It not only deteriorates the therapeutic performance of biologic drugs but also contributes to various diseases such as allergies and autoimmune diseases. Inspired by the role of chromatin in the maintenance of natural immune tolerance, here we report a DNA-protein polymeric nanocomplex that can mimic the tolerogenic function of chromatin and induce an immune tolerance to its protein cargos. We first proved that the chromatin-mimetic nanomedicine loaded with keyhole limpet hemocyanin (KLH), a highly immunogenic model protein, could elicit a durable antigen-specific immune tolerance to KLH lasting for at least five weeks in mice. Following the proof-of-concept study, we demonstrated that this nanomedicine could be applied to improve the safety and efficacy of a biologic drug, PEGylated uricase, by attenuating the relevant antibody (Ab) responses. Moreover, we also demonstrated that prophylactic treatments with this nanomedicine could tolerize the immune system with the allergen of ovalbumin (OVA) and thus inhibit the occurrence of airway inflammation in an OVA-induced allergic asthma murine model. Collectively, our work illustrates a nature-inspired concept of immune tolerance induction and establishes a useful tool to specifically suppress unwanted immune responses for therapeutic purposes.
Collapse
Affiliation(s)
- Bowen Li
- Department of Bioengineering , University of Washington , Seattle , Washington 98195 , United States
| | - Zhefan Yuan
- Department of Chemical Engineering , University of Washington , Seattle , Washington 98195 , United States
| | - Patrick McMullen
- Department of Chemical Engineering , University of Washington , Seattle , Washington 98195 , United States
| | - Jingyi Xie
- Department of Bioengineering , University of Washington , Seattle , Washington 98195 , United States
| | - Priyesh Jain
- Department of Chemical Engineering , University of Washington , Seattle , Washington 98195 , United States
| | - Hsiang-Chieh Hung
- Department of Chemical Engineering , University of Washington , Seattle , Washington 98195 , United States
| | - Shihan Xu
- Department of Bioengineering , University of Washington , Seattle , Washington 98195 , United States
| | - Peng Zhang
- Department of Chemical Engineering , University of Washington , Seattle , Washington 98195 , United States
| | - Xiaojie Lin
- Department of Chemical Engineering , University of Washington , Seattle , Washington 98195 , United States
| | - Kan Wu
- Department of Chemical Engineering , University of Washington , Seattle , Washington 98195 , United States
| | - Shaoyi Jiang
- Department of Bioengineering , University of Washington , Seattle , Washington 98195 , United States
- Department of Chemical Engineering , University of Washington , Seattle , Washington 98195 , United States
| |
Collapse
|
15
|
Abstract
BACKGROUND AND AIM Alagille syndrome (ALGS) is an inherited multisystem disorder typically manifesting as cholestasis, and potentially leading to end-stage liver disease and death. The aim of the study was to perform the first systematic review of the epidemiology, natural history, and burden of ALGS with a focus on the liver component. METHODS Electronic databases and proceedings from key congresses were searched in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2009 guidelines. This analysis included publications reporting epidemiology, natural history, economic burden or health-related quality of life (HRQoL) outcomes in patients with ALGS. RESULTS Of 525 screened publications, 20 met the inclusion criteria. Liver-related features included cholestasis (87%-100% of patients), jaundice (66%-85%), and cirrhosis (44%-95%). Between 15% and 47% of patients underwent liver transplantation and 4% to 14% received partial biliary diversion. Pruritus affected the majority of patients (59%-88%, of whom up to 45% had severe pruritus) and manifested during the first 10 years of life. Children with ALGS had significantly impaired HRQoL compared with healthy controls and those with other diseases. Itching was the symptom that most affected children with ALGS. No study assessed the economic burden of ALGS. CONCLUSIONS Our findings consolidate information on the clinical course of ALGS, and highlight gaps in knowledge, most notably the absence of any research on the economic consequences of the disease. Further research is needed to establish the incidence of genetically confirmed ALGS. Disease-specific tools are also needed to improve the measurement of symptoms, such as itching, and better understand the impact of ALGS on HRQoL.
Collapse
|
16
|
Reddel SW, Barnett MH, Riminton S, Dugal T, Buzzard K, Wang CT, Fitzgerald F, Beadnall HN, Erickson D, Gahan D, Wang D, Ackland T, Thompson R. Successful implementation of an automated electronic support system for patient safety monitoring: The alemtuzumab in multiple sclerosis safety systems (AMS3) study. Mult Scler 2018; 25:1124-1131. [PMID: 29911471 DOI: 10.1177/1352458518783673] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alemtuzumab is a highly effective treatment for relapsing-remitting multiple sclerosis (MS) but requires ongoing pathology monitoring for autoimmune adverse effects. The Alemtuzumab in MS Safety Systems (AMS3) study evaluated the implementation of an automated pathology-monitoring system. OBJECTIVES To develop an efficient automated clinical decision support system (CDSS) to electronically prompt and track pathology collection and to provide prescribers and patients with customised alerts of abnormal results for identified risks. METHODS A total of 10 patients with relapsing-remitting MS treated with alemtuzumab were enrolled to test the system. Standard care laboratory monitoring was performed and compared to the performance of the CDSS. RESULTS The automated CDSS, an integrated patient smartphone application and an additional pre-screening tool were all successfully developed. Compliance with pathology monitoring was 96.7%. The automated analysis of pathology results was significantly faster than standard care neurologist review (p < 0.001). The system correctly identified and alerted abnormalities, including one case of immune thrombocytopenia (ITP) while the treating neurologist was on leave, enabling prompt treatment of serious adverse events. During the course of the study, the CDSS was deployed throughout Australia. CONCLUSION We successfully developed automated pathology monitoring with a CDSS, demonstrating real-world benefits of high compliance and timely alerting of important results.
Collapse
Affiliation(s)
- Stephen W Reddel
- Department of Neurology, Concord Hospital, The University of Sydney, Concord, NSW, Australia
| | - Michael H Barnett
- Sydney Neuroimaging Analysis Centre, Brain and Mind Centre, Sydney, NSW, Australia/Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Sean Riminton
- Department of Immunology, Concord Hospital, The University of Sydney, Sydney, NSW, Australia
| | - Tej Dugal
- Norwest Medical Imaging, Sydney, NSW, Australia
| | - Katherine Buzzard
- Department of Neurology, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Chenu Tim Wang
- Sydney Neuroimaging Analysis Centre, Brain and Mind Centre, Sydney, NSW, Australia
| | | | | | | | - David Gahan
- Medical Safety Systems, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
17
|
Xu X, Bian L, Shen M, Li X, Zhu J, Chen S, Xiao L, Zhang Q, Chen H, Xu K, Yang T. Multipeptide-coupled nanoparticles induce tolerance in 'humanised' HLA-transgenic mice and inhibit diabetogenic CD8 + T cell responses in type 1 diabetes. Diabetologia 2017; 60:2418-2431. [PMID: 28887632 DOI: 10.1007/s00125-017-4419-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/12/2017] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS Induction of antigen-specific immunological tolerance may provide an attractive immunotherapy in the NOD mouse model but the conditions that lead to the successful translation to human type 1 diabetes are limited. In this study, we covalently linked 500 nm carboxylated polystyrene beads (PSB) with a mixture of immunodominant HLA-A*02:01-restricted epitopes (peptides-PSB) that may have high clinical relevance in humans as they promote immune tolerance; we then investigated the effect of the nanoparticle-peptide complexes on T cell tolerance. METHODS PSB-coupled mixtures of HLA-A*02:01-restricted epitopes were administered to HHD II mice via intravenous injection. The effects on delaying the course of the disease were verified in NOD.β2m null HHD mice. The diabetogenic HLA-A*02:01-restricted cytotoxic lymphocyte (CTL) responses to treatment with peptides-PSB were validated in individuals with type 1 diabetes. RESULTS We showed that peptides-PSB could induce antigen-specific tolerance in HHD II mice. The protective immunological mechanisms were mediated through the function of CD4+CD25+ regulatory T cells, suppressive T cell activation and T cell anergy. Furthermore, the peptides-PSB induced an activation and accumulation of regulatory T cells and CD11c+ dendritic cells through a rapid production of CD169+ macrophage-derived C-C motif chemokine 22 (CCL22). Peptides-PSB also prevented diabetes in 'humanised' NOD.β2m null HHD mice and suppressed pathogenic CTL responses in people with type 1 diabetes. CONCLUSIONS/INTERPRETATION Our findings demonstrate for the first time the potential for using multipeptide-PSB complexes to induce T cell tolerance and halt the autoimmune process. These findings represent a promising platform for an antigen-specific tolerance strategy in type 1 diabetes and highlight a mechanism through which metallophilic macrophages mediate the early cell-cell interactions required for peptides-PSB-induced immune tolerance.
Collapse
Affiliation(s)
- Xinyu Xu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Lingling Bian
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
- Department of Endocrinology, Yancheng City No.1 People's Hospital, Yancheng, Jiangsu Province, People's Republic of China
| | - Min Shen
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Xin Li
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Jing Zhu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Shuang Chen
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Lei Xiao
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Qingqing Zhang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Heng Chen
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Kuanfeng Xu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| |
Collapse
|
18
|
Mori AM, Agarwal S, Lee MW, Rafferty M, Hardy TA, Coles A, Reddel SW, Riminton DS. A systematic checklist approach to immunosuppression risk management: An audit of practice at two clinical neuroimmunology centers. J Neuroimmunol 2017; 312:4-7. [DOI: 10.1016/j.jneuroim.2017.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/04/2017] [Accepted: 08/18/2017] [Indexed: 10/19/2022]
|
19
|
Pang L, Macauley MS, Arlian BM, Nycholat CM, Paulson JC. Encapsulating an Immunosuppressant Enhances Tolerance Induction by Siglec-Engaging Tolerogenic Liposomes. Chembiochem 2017; 18:1226-1233. [PMID: 28231415 DOI: 10.1002/cbic.201600702] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Indexed: 12/29/2022]
Abstract
Unwanted antibody responses significantly impact human health, and current options for treating deleterious antibody responses largely rely on broad immunosuppressants that can compromise overall immunity. A desirable alternative is to induce antigen-specific immune tolerance. We have shown that co-presentation of antigen and ligands of B cell sialic acid-binding immunoglobulin-like lectins (Siglecs) on a liposomal nanoparticle induces antigen-specific tolerance. Although Siglec-engaging tolerance-inducing antigenic liposomes (STALs) induce robust B cell tolerance in naïve mice, the full potential of STALs requires long-term tolerance induction and suppression of an ongoing immune response. We hypothesized that STALs encapsulated with rapamycin (RAPA), an immunomodulator, could improve the efficacy of STALs and potentially enable their use in the context of immunological memory. Here, we showed that formulation of STALs with RAPA produced enhanced tolerance induction in naïve mice compared to STALs without RAPA but had minimal impact on inducing tolerance in previously sensitized mice. These findings indicate that the addition of immunomodulators to STALs could be beneficial in tolerance induction and support future development of STALs for the treatment of allergy and autoimmune diseases.
Collapse
Affiliation(s)
- Lijuan Pang
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Matthew S Macauley
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Britni M Arlian
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Corwin M Nycholat
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
20
|
Grigoriadis N, Linnebank M, Alexandri N, Muehl S, Hofbauer GFL. Considerations on long-term immuno-intervention in the treatment of multiple sclerosis: an expert opinion. Expert Opin Pharmacother 2016; 17:2085-95. [PMID: 27594523 DOI: 10.1080/14656566.2016.1232712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION As management of multiple sclerosis (MS) requires life-long treatment with disease-modifying agents, any risks associated with long-term use should be considered when evaluating therapeutic options. AREAS COVERED Immune cells of the innate and adaptive immune systems play various roles in the pathogenesis of MS. MS therapies affect the immune system, each with a unique mode of action, and consequently possess different long-term safety profiles. Rare, but serious safety concerns, including an increased risk of infection and cancer, have been associated with immunosuppressant use. The risks associated with newer immunosuppressive agents, which target specific elements of MS disease pathophysiology, are not yet fully established as the duration of clinical trials is relatively short and post-marketing experience is limited. Non-immunosuppressants used to treat MS have well-defined safety profiles established over a large number of patient-years demonstrating them to be well-tolerated long-term treatment options. When considering the long-term use of disease-modifying agents for treating MS, classification as immunosuppressants or non-immunosuppressants can be useful when evaluating potential risks associated with chronic use. EXPERT OPINION A successful therapeutic strategy for any serious, chronic disease such as MS should weigh effectiveness versus long-term safety of available treatments.
Collapse
Affiliation(s)
- Nikolaos Grigoriadis
- a B' Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology , AHEPA University Hospital, Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Michael Linnebank
- b Klinik für Neurologie Universitätsspital Zürich , Zürich , Switzerland.,c Department of Neurology , Helios-Klinik, Hagen-Ambrock , Hagen , Germany
| | | | - Sarah Muehl
- e Merck (Schweiz) AG, Zug, Switzerland, a subsidiary of Merck KGaA Darmstadt , Germany
| | | |
Collapse
|
21
|
[Infection risks in multiple sclerosis therapy by infusion of disease modifying drugs]. DER NERVENARZT 2016; 86:971-7. [PMID: 26187545 DOI: 10.1007/s00115-015-4388-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The increased risk of developing infections when using disease-modifying drugs for treatment of multiple sclerosis (MS) is a major challenge in the daily clinical routine. In the growing field of treatment options specific knowledge of treatment-related risks of infections and appropriate preventive and countermeasures is mandatory. Current clinical experience shows that an individual risk stratification is necessary when choosing treatment options and while monitoring during and after treatment administration. The determination of the individual risk of infection in the context of serial use of disease-modifying drugs remains a challenging issue. In addition to the mechanisms of action, the warning notices and current recommendations on infection prophylaxis when using intravenous disease-modifying drugs, such as alemtuzumab, natalizumab and mitoxantron, are presented in detail.
Collapse
|
22
|
Abstract
Immunomodulatory and immunosuppressive treatments for multiple sclerosis (MS) are associated with an increased risk of infection, which makes treatment of this condition challenging in daily clinical practice. Use of the expanding range of available drugs to treat MS requires extensive knowledge of treatment-associated infections, risk-minimizing strategies and approaches to monitoring and treatment of such adverse events. An interdisciplinary approach to evaluate the infectious events associated with available MS treatments has become increasingly relevant. In addition, individual stratification of treatment-related infectious risks is necessary when choosing therapies for patients with MS, as well as during and after therapy. Determination of the individual risk of infection following serial administration of different immunotherapies is also crucial. Here, we review the modes of action of the available MS drugs, and relate this information to the current knowledge of drug-specific infectious risks and risk-minimizing strategies.
Collapse
|
23
|
Winkelmann A, Löbermann M, Reisinger EC, Hartung HP, Zettl UK. [Immunotherapy and infectious issues in multiple sclerosis. Self-injectable and oral drugs for immunotherapy]. DER NERVENARZT 2015; 86:960-970. [PMID: 26187544 DOI: 10.1007/s00115-015-4369-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Immunotherapy is generally associated with an increased risk for the development of infections. Due to the continuously expanding spectrum of new and potent immunotherapy treatment options for multiple sclerosis (MS), this article describes the currently known risks for treatment-related infections and the current recommendations for prevention of corresponding problems with drugs used in treatment strategies for MS and their mechanisms of action. The new treatment options in particular are linked to specific and severe infections; therefore, intensive and long-lasting monitoring is required before, during and after treatment and multidisciplinary surveillance of patients is needed. This article gives a detailed review of drug-specific red flags and current recommendations for the prophylaxis of infections associated with treatment of relapsing-remitting MS and when using self-injectable and oral disease-modifying immunotherapeutic drugs.
Collapse
Affiliation(s)
- A Winkelmann
- Klinik und Poliklinik für Neurologie, Universitätsmedizin Rostock, Gehlsheimer Str. 20, 18147, Rostock, Deutschland,
| | | | | | | | | |
Collapse
|
24
|
Buzzard KA, Meyer NJ, Hardy TA, Riminton DS, Reddel SW. Induction intravenous cyclophosphamide followed by maintenance oral immunosuppression in refractory myasthenia gravis. Muscle Nerve 2015; 52:204-10. [PMID: 25487528 DOI: 10.1002/mus.24536] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2014] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Myasthenia gravis (MG) can be refractory to conventional immunotherapy. We report on the efficacy and durability of intravenous (IV) remission-induction cyclophosphamide (CYC) followed by oral immunosuppression in refractory MG. METHODS We identified 8 patients from our medical records with moderate or severe refractory MG who were treated with 6 cycles of IV CYC (0.75 g/m(2) ) every 4 weeks followed by oral immunosuppression. RESULTS Six patients improved within 3 months of treatment. Four patients remained in clinical remission (mean follow-up 31 months). Two patients responded partially, and 1 patient relapsed after 11 months. Two patients were non-responders. CYC was well tolerated. Acetylcholine receptor antibody levels remained below pretreatment levels in patients in clinical remission. The leukocyte nadir was lower in CYC responders. CONCLUSIONS Remission-induction IV CYC followed by oral immunosuppression is a rapid, effective, and durable treatment for refractory MG. Adding a post-CYC immunosuppressant may account for low relapse rates compared with other published series.
Collapse
Affiliation(s)
- Katherine A Buzzard
- Department of Neurology, Concord Repatriation General Hospital, Concord West, New South Wales, 2139, Australia
| | - Nicholas J Meyer
- Department of Immunology, Concord Repatriation General Hospital, Concord West, New South Wales, Australia
| | - Todd A Hardy
- Department of Neurology, Concord Repatriation General Hospital, Concord West, New South Wales, 2139, Australia
| | - D Sean Riminton
- Department of Immunology, Concord Repatriation General Hospital, Concord West, New South Wales, Australia
| | - Stephen W Reddel
- Department of Neurology, Concord Repatriation General Hospital, Concord West, New South Wales, 2139, Australia
| |
Collapse
|
25
|
Curtin F, Perron H, Kromminga A, Porchet H, Lang AB. Preclinical and early clinical development of GNbAC1, a humanized IgG4 monoclonal antibody targeting endogenous retroviral MSRV-Env protein. MAbs 2015; 7:265-75. [PMID: 25427053 PMCID: PMC4623301 DOI: 10.4161/19420862.2014.985021] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/15/2014] [Accepted: 10/31/2014] [Indexed: 11/19/2022] Open
Abstract
Monoclonal antibodies (mAbs) play an increasing important role in the therapeutic armamentarium against multiple sclerosis (MS), an inflammatory and degenerative disorder of the central nervous system. Most of the mAbs currently developed for MS are immunomodulators blocking the inflammatory immune process. In contrast with mAbs targeting immune function, GNbAC1, a humanized IgG4 mAb, targets the multiple sclerosis associated retrovirus envelope (MSRV-Env) protein, an upstream factor in the pathophysiology of MS. MSRV-Env protein is of endogenous retroviral origin, expressed in MS brain lesions, and it is pro-inflammatory and toxic to the remyelination process, by preventing the differentiation of oligodendrocyte precursor cells. We present the preclinical and early clinical development results of GNbAC1. The specificity of GNbAC1 for its endogenous retroviral target is described. Efficacy of different mAb versions of GNbAC1 were assessed in MSRV-Env induced experimental allergic encephalitis (EAE), an animal model of MS. Because the target MSRV-Env is not expressed in animals, no relevant animal model exists for a proper in vivo toxicological program. An off-target 2-week toxicity study in mice was thus performed, and it showed an absence of safety risk. Additional in vitro analyses showed an absence of complement or antibody-dependent cytotoxicity as well as a low level of cross-reactivity to human tissues. The first-in-man clinical study in 33 healthy subjects and a long-term clinical study in 10 MS patients showed that GNbAC1 is well tolerated in humans without induction of immunogenicity and that it induces a pharmacodynamic response on MSRV biomarkers. These initial results suggest that the mAb GNbAC1 could be a safe long-term treatment for patients with MS with a unique therapeutic mechanism of action.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- AE, adverse events
- AUC, area under the curve
- BLAST, Basic Local Alignment Search Tool
- CDC, complement-dependent cytotoxicity
- CDR, complementarity-determining regions
- Cmax, maximal concentration
- Cmin, minimal concentration
- HERV-W
- HERV-W, human endogenous retrovirus type W
- HLA, human leukocyte antigen
- MOG, myelin oligodendrocyte glycoprotein
- MS, multiple sclerosis
- MSRV
- MSRV, multiple sclerosis associated retrovirus
- MSRV-Env, multiple sclerosis associated retrovirus envelope protein
- PBMC, peripheral blood mononuclear cell
- SAE, serious adverse event
- SU, surface domain
- Syncytin
- TLR4, Toll-like receptor 4
- ch-GNbAC1, chimeric version of mAb GNbAC1
- drug safety
- human endogenous retrovirus
- mAb, monoclonal antibody
- monoclonal antibody
- mu-GNbAC1, murine version of mAb GNbAC1
- multiple sclerosis
- neurotoxicity
- toxicology
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/immunology
- Antibody Specificity
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Endogenous Retroviruses/immunology
- Female
- Gene Products, env/immunology
- HEK293 Cells
- Humans
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/adverse effects
- Immunoglobulin G/immunology
- Male
- Mice
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
Collapse
Affiliation(s)
- François Curtin
- GeNeuro SA; Plan-les-Ouates/Geneva, Switzerland
- Division of Clinical Pharmacology and Toxicology; University of Geneva; Geneva, Switzerland
| | | | - Arno Kromminga
- Institute of Immunology; University Kiel; Kiel, Germany
- IPM Biotech; Hamburg, Germany
| | - Hervé Porchet
- GeNeuro SA; Plan-les-Ouates/Geneva, Switzerland
- Department of Pharmacology; University of Pretoria; Pretoria, South Africa
| | | |
Collapse
|
26
|
Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc Natl Acad Sci U S A 2014; 112:E156-65. [PMID: 25548186 DOI: 10.1073/pnas.1408686111] [Citation(s) in RCA: 343] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Current treatments to control pathological or unwanted immune responses often use broadly immunosuppressive drugs. New approaches to induce antigen-specific immunological tolerance that control both cellular and humoral immune responses are desirable. Here we describe the use of synthetic, biodegradable nanoparticles carrying either protein or peptide antigens and a tolerogenic immunomodulator, rapamycin, to induce durable and antigen-specific immune tolerance, even in the presence of potent Toll-like receptor agonists. Treatment with tolerogenic nanoparticles results in the inhibition of CD4+ and CD8+ T-cell activation, an increase in regulatory cells, durable B-cell tolerance resistant to multiple immunogenic challenges, and the inhibition of antigen-specific hypersensitivity reactions, relapsing experimental autoimmune encephalomyelitis, and antibody responses against coagulation factor VIII in hemophilia A mice, even in animals previously sensitized to antigen. Only encapsulated rapamycin, not the free form, could induce immunological tolerance. Tolerogenic nanoparticle therapy represents a potential novel approach for the treatment of allergies, autoimmune diseases, and prevention of antidrug antibodies against biologic therapies.
Collapse
|
27
|
Mangin M, Sinha R, Fincher K. Inflammation and vitamin D: the infection connection. Inflamm Res 2014; 63:803-19. [PMID: 25048990 PMCID: PMC4160567 DOI: 10.1007/s00011-014-0755-z] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 06/04/2014] [Accepted: 06/23/2014] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Inflammation is believed to be a contributing factor to many chronic diseases. The influence of vitamin D deficiency on inflammation is being explored but studies have not demonstrated a causative effect. METHODS Low serum 25(OH)D is also found in healthy persons exposed to adequate sunlight. Despite increased vitamin D supplementation inflammatory diseases are increasing. The current method of determining vitamin D status may be at fault. The level of 25(OH)D does not always reflect the level of 1,25(OH)2D. Assessment of both metabolites often reveals elevated 1,25(OH)2D, indicating abnormal vitamin D endocrine function. FINDINGS This article reviews vitamin D's influence on the immune system, examines the myths regarding vitamin D photosynthesis, discusses ways to accurately assess vitamin D status, describes the risks of supplementation, explains the effect of persistent infection on vitamin D metabolism and presents a novel immunotherapy which provides evidence of an infection connection to inflammation. CONCLUSION Some authorities now believe that low 25(OH)D is a consequence of chronic inflammation rather than the cause. Research points to a bacterial etiology pathogenesis for an inflammatory disease process which results in high 1,25(OH)2D and low 25(OH)D. Immunotherapy, directed at eradicating persistent intracellular pathogens, corrects dysregulated vitamin D metabolism and resolves inflammatory symptoms.
Collapse
Affiliation(s)
- Meg Mangin
- Chronic Illness Recovery, Fort Worth, Texas, USA,
| | | | | |
Collapse
|
28
|
Clinical and scientific aspects of muscle-specific tyrosine kinase-related myasthenia gravis. Curr Opin Neurol 2014; 27:558-65. [DOI: 10.1097/wco.0000000000000136] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Clanet MC, Wolinsky JS, Ashton RJ, Hartung HP, Reingold SC. Risk evaluation and monitoring in multiple sclerosis therapeutics. Mult Scler 2014; 20:1306-11. [PMID: 24293456 PMCID: PMC4232326 DOI: 10.1177/1352458513513207] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 10/23/2013] [Accepted: 10/25/2013] [Indexed: 11/21/2022]
Abstract
BACKGROUND Risk for multiple sclerosis (MS) disease-modifying therapies (DMT) must be assessed on an ongoing basis. Early concerns regarding the first-approved DMTs for MS have been mitigated, but recently licensed therapies have been linked to possibly greater risks. OBJECTIVES The objective of this review is to discuss risk assessment in MS therapeutics based on an international workshop and comprehensive literature search and recommend strategies for risk assessment/monitoring. RESULTS Assessment and perception of therapeutic risks vary between patients, doctors and regulators. Acceptability of risk depends on the magnitude of risk and the demonstrated clinical benefits of any agent. Safety signals must be distinguishable from chance occurrences in a clinical trial and in long-term use of medications. Post-marketing research is crucial for assessing longer-term safety in large patient cohorts. Reporting of adverse events is becoming more proactive, allowing more rapid identification of risks. Communication about therapeutic risks and their relationship to clinical benefit must involve patients in shared decision making. CONCLUSIONS It is difficult to produce a general risk-assessment algorithm for all MS therapies. Specific algorithms are required for each DMT in every treated-patient population. New and evolving risks must be evaluated and communicated rapidly to allow patients and physicians to be well informed and able to share treatment decisions.
Collapse
Affiliation(s)
- Michel C Clanet
- Department of Neurosciences University Hospital Toulouse, France
| | - Jerry S Wolinsky
- Department of Neurology, The University of Texas Health Science Center at Houston, USA
| | | | - Hans-Peter Hartung
- Department of Neurology and Center for Neuropsychiatry, Medical Faculty, Heinrich-Heine-Universität im UKD, Germany
| | | |
Collapse
|
30
|
Winkelmann A, Loebermann M, Reisinger EC, Zettl UK. Multiple sclerosis treatment and infectious issues: update 2013. Clin Exp Immunol 2014; 175:425-38. [PMID: 24134716 DOI: 10.1111/cei.12226] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2013] [Indexed: 01/13/2023] Open
Abstract
Immunomodulation and immunosuppression are generally linked to an increased risk of infection. In the growing field of new and potent drugs for multiple sclerosis (MS), we review the current data concerning infections and prevention of infectious diseases. This is of importance for recently licensed and future MS treatment options, but also for long-term established therapies for MS. Some of the disease-modifying therapies (DMT) go along with threats of specific severe infections or complications, which require a more intensive long-term monitoring and multi-disciplinary surveillance. We update the existing warning notices and infectious issues which have to be considered using drugs for multiple sclerosis.
Collapse
Affiliation(s)
- A Winkelmann
- Department of Neurology, University of Rostock, Rostock, Germany
| | | | | | | |
Collapse
|
31
|
Ghazanfari N, Morsch M, Tse N, Reddel SW, Phillips WD. Effects of the ß2-adrenoceptor agonist, albuterol, in a mouse model of anti-MuSK myasthenia gravis. PLoS One 2014; 9:e87840. [PMID: 24505322 PMCID: PMC3914858 DOI: 10.1371/journal.pone.0087840] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 12/31/2013] [Indexed: 11/19/2022] Open
Abstract
The β2-adrenergic receptor agonist, albuterol, has been reported beneficial in treating several forms of congenital myasthenia. Here, for the first time, we examined the potential benefit of albuterol in a mouse model of anti-Muscle Specific Kinase (MuSK) myasthenia gravis. Mice received 15 daily injections of IgG from anti-MuSK positive patients, which resulted in whole-body weakness. At neuromuscular junctions in the tibialis anterior and diaphragm muscles the autoantibodies caused loss of postsynaptic acetylcholine receptors, and reduced the amplitudes of the endplate potential and spontaneous miniature endplate potential in the diaphragm muscle. Treatment with albuterol (8 mg/kg/day) during the two-week anti-MuSK injection series reduced the degree of weakness and weight loss, compared to vehicle-treated mice. However, the compound muscle action potential recorded from the gastrocnemius muscle displayed a decremental response in anti-MuSK-injected mice whether treated with albuterol or vehicle. Ongoing albuterol treatment did not increase endplate potential amplitudes compared to vehicle-treated mice nor did it prevent the loss of acetylcholine receptors from motor endplates. On the other hand, albuterol treatment significantly reduced the degree of fragmentation of endplate acetylcholine receptor clusters and increased the extent to which the remaining receptor clusters were covered by synaptophysin-stained nerve terminals. The results provide the first evidence that short-term albuterol treatment can ameliorate weakness in a robust mouse model of anti-MuSK myasthenia gravis. The results also demonstrate that it is possible for albuterol treatment to reduce whole-body weakness without necessarily reversing myasthenic impairment to the structure and function of the neuromuscular junction.
Collapse
MESH Headings
- Adrenergic beta-2 Receptor Agonists/pharmacology
- Albuterol/pharmacology
- Animals
- Autoantibodies/immunology
- Autoantibodies/toxicity
- Female
- Humans
- Mice
- Muscle, Skeletal/immunology
- Muscle, Skeletal/pathology
- Myasthenia Gravis, Autoimmune, Experimental/chemically induced
- Myasthenia Gravis, Autoimmune, Experimental/drug therapy
- Myasthenia Gravis, Autoimmune, Experimental/immunology
- Myasthenia Gravis, Autoimmune, Experimental/pathology
- Neuromuscular Junction/immunology
- Neuromuscular Junction/pathology
- Receptor Protein-Tyrosine Kinases/immunology
Collapse
Affiliation(s)
- Nazanin Ghazanfari
- Physiology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Marco Morsch
- Physiology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Nigel Tse
- Physiology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Stephen W. Reddel
- Department of Molecular Medicine, Concord Hospital, Concord, New South Wales, Australia
| | - William D. Phillips
- Physiology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
32
|
Abstract
Multiple sclerosis (MS) is considered as an autoimmune disorder of the CNS with neuro-inflammatory and neurodegenerative components. We review here the innovative drugs recently registered and those in clinical development for MS. Immunomodulation has been the preferred therapeutic approach for MS since the first IFN-β was registered in the 1990s. Several immunomodulators are now available, which show a high efficacy in reducing the number of relapses in patients with the relapsing-remitting form of MS (RRMS). The high efficacy of most immunomodulators is, however, associated with substantial safety risks, notably concerning infections or cancers. Recently oral drugs have been approved for RRMS; however, biologics, and notably, monoclonal antibodies are still well represented in the development pipelines. An unmet medical need remains the treatment of the primary and secondary forms of MS or chronic progressive MS (CPMS). Half a dozen immunomodulators with proven efficacy in RRMS are now undergoing evaluation in Phase III trials in the CPMS indication. Neuroprotective drugs that prevent demyelination and/or improve remyelination would be interesting for CPMS, but these drugs are currently in the early development phase and their efficacy has not been demonstrated yet.
Collapse
Affiliation(s)
- François Curtin
- GeNeuro SA, Chemin des Aulx 18, CH-1228 Plan-les-Ouates/Geneva, Switzerland
| | | |
Collapse
|
33
|
Hradilek P, Zeman D, Tudik I, Zapletalova O, Ulmann V. Asymptomatic lung disease caused by Mycobacterium kansasii as an opportunistic infection in a patient treated with natalizumab for relapsing-remitting multiple sclerosis. Mult Scler 2013; 20:639-40. [PMID: 23959714 DOI: 10.1177/1352458513501572] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Warnke C, Kieseier BC, Hartung HP. Biotherapeutics for the treatment of multiple sclerosis: hopes and hazards. J Neural Transm (Vienna) 2013; 120 Suppl 1:S55-60. [DOI: 10.1007/s00702-013-1055-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/11/2013] [Indexed: 11/25/2022]
|
35
|
Martin-Blondel G, Brassat D, Dumas H, Uro-Coste E, Adoue D, Lassmann H, Clanet M. A case report of simultaneous PML-IRIS during corticosteroids tapering in a patient with an anti-synthetase syndrome. F1000Res 2013; 2:283. [PMID: 24715969 PMCID: PMC3968897 DOI: 10.12688/f1000research.2-283.v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/23/2013] [Indexed: 01/22/2023] Open
Abstract
We report a case of simultaneous progressive multifocal leukoencephalopathy-associated immune reconstitution inflammatory syndrome (PML-IRIS) during corticosteroid tapering in a patient with an anti-synthetase syndrome. We describe the challenges associated with the diagnosis and the management of this emerging inflammatory neurological condition in this immunocompromised patient with a severe rheumatic disease. We highlight that, in the setting of IRIS, the low-level of the JC virus viral load requires a sensitive PCR assay before excluding PML.
Collapse
Affiliation(s)
- Guillaume Martin-Blondel
- Department of Infectious and Tropical Diseases, Toulouse University Hospital, Toulouse, France ; INSERM UMR 1043, Centre de Physiopathologie, Toulouse-Purpan, France ; Université Toulouse III, Toulouse F-31000, France
| | - David Brassat
- INSERM UMR 1043, Centre de Physiopathologie, Toulouse-Purpan, France ; Université Toulouse III, Toulouse F-31000, France ; Department of Neurology, Toulouse University Hospital, Toulouse, France
| | - Hervé Dumas
- Department of Neuroradiology, Toulouse University Hospital, Toulouse, France
| | - Emmanuelle Uro-Coste
- Université Toulouse III, Toulouse F-31000, France ; Department of Pathology, Toulouse University Hospital, Toulouse, France
| | - Daniel Adoue
- Université Toulouse III, Toulouse F-31000, France ; Department of Internal Medicine, Toulouse University Hospital, Toulouse, France
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Michel Clanet
- INSERM UMR 1043, Centre de Physiopathologie, Toulouse-Purpan, France ; Université Toulouse III, Toulouse F-31000, France ; Department of Neurology, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Inflammation has been widely acknowledged to contribute throughout all stages of atherogenesis. However, these recent advances in our understanding have not been translated into clinical practice in which the mainstay of treatment is still lipid-targeted therapy. This review provides an overview of promising anti-inflammatory therapies in atherosclerosis, and discusses potential drawbacks and clinical benefits. RECENT FINDINGS Immunosuppressive drugs are likely to beneficially affect atherogenesis. Several novel anti-inflammatory targets have been scrutinized, of which some have reached clinical development stage, such as cytokine targets interleukin-1 and interleukin-6, CCR2 antagonist, selective phospholipase, and leukotriene inhibitors. Novel imaging modalities such as MRI and PET-computed tomography provide valuable surrogate inflammatory endpoints for risk stratification and testing anti-inflammatory agents in cardiovascular randomized trials. SUMMARY Anti-inflammatory therapies hold great promise in cardiovascular prevention regimens; however, atherosclerosis is a chronic disease, and systemic long-term use of anti-inflammatory agents carries the risk of complications arising from immunosuppression. In order to successfully add immunosuppressive drugs to our routine armament, we need to identify high-risk patients who benefit from anti-inflammatory treatment, increase our insight into the inflammatory pathogenesis of atherogenesis, and find safe and effective compounds capable of directly suppressing plaque inflammation.
Collapse
|
37
|
Chang T, Rodrigo C, Ranawaka N, Atukorala I. Multiple ring-enhancing cerebral lesions in systemic lupus erythematosis: a case report. J Med Case Rep 2012; 6:172. [PMID: 22742706 PMCID: PMC3407750 DOI: 10.1186/1752-1947-6-172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 06/28/2012] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Infectious disease in an immunosuppressed patient is a diagnostic challenge. The clinical presentation and the body's immune response may be quite different from those seen in an immunocompetent patient with the same infection. It is also a race against time to diagnose, as many of these infections can be fatal without timely intervention. CASE PRESENTATION We present the case of a 39-year-old Sri Lankan woman who was on immunosuppressive treatment for systemic lupus erythematosis and who presented with multiple ring-enhancing lesions of the brain. The most likely diagnosis, given the clinical picture, available investigation results, and characteristics of magnetic resonance imaging, was central nervous system tuberculosis. Owing to the small size of the lesions, a tissue biopsy could not be performed. Our patient responded well to a trial of anti-tuberculosis therapy, and there was clinical and radiological evidence of recovery. A paradoxical reaction with the initiation of anti-tuberculosis therapy was observed and this had to be countered with a prolonged course of steroids. CONCLUSIONS Our experience and previous evidence from case reports suggest that high-dose steroids for a prolonged period (up to eight weeks) should be administered to counter the initial deterioration after starting anti-tuberculous chemotherapy for central nervous system tuberculomas.
Collapse
Affiliation(s)
- Thashi Chang
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Chaturaka Rodrigo
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka,National Hospital of Sri Lanka, University Medical Unit, Colombo 10, Sri Lanka
| | - Nuwan Ranawaka
- National Hospital of Sri Lanka, University Medical Unit, Colombo 10, Sri Lanka
| | - Inoshi Atukorala
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| |
Collapse
|
38
|
Alcon A, Cagavi Bozkulak E, Qyang Y. Regenerating functional heart tissue for myocardial repair. Cell Mol Life Sci 2012; 69:2635-56. [PMID: 22388688 DOI: 10.1007/s00018-012-0942-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 01/21/2012] [Accepted: 02/13/2012] [Indexed: 12/20/2022]
Abstract
Heart disease is one of the leading causes of death worldwide and the number of patients with the disease is likely to grow with the continual decline in health for most of the developed world. Heart transplantation is one of the only treatment options for heart failure due to an acute myocardial infarction, but limited donor supply and organ rejection limit its widespread use. Cellular cardiomyoplasty, or cellular implantation, combined with various tissue-engineering methods aims to regenerate functional heart tissue. This review highlights the numerous cell sources that have been used to regenerate the heart as well as cover the wide range of tissue-engineering strategies that have been devised to optimize the delivery of these cells. It will probably be a long time before an effective regenerative therapy can make a serious impact at the bedside.
Collapse
Affiliation(s)
- Andre Alcon
- Yale University School of Medicine, Yale University, New Haven, CT, USA
| | | | | |
Collapse
|
39
|
Loebermann M, Winkelmann A, Hartung HP, Hengel H, Reisinger EC, Zettl UK. Vaccination against infection in patients with multiple sclerosis. Nat Rev Neurol 2012; 8:143-51. [PMID: 22270022 DOI: 10.1038/nrneurol.2012.8] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bacterial and viral infections have been shown to induce relapses and accelerate the progression of multiple sclerosis (MS). Vaccination to prevent communicable disease in such patients is, therefore, of key importance. Reports of potentially detrimental effects of immunization on the course of MS, however, have prompted patients and physicians to adopt a cautious attitude towards the use of vaccines. The risks associated with a number of vaccines have been investigated in patients with MS. Vaccines against some diseases, such as tetanus and hepatitis B, are not associated with an elevated risk of MS exacerbation, whereas vaccines against other diseases, such as yellow fever, are contraindicated in patients with MS. Many patients with MS receive immunosuppressive or immunomodulatory therapy, which could make them more susceptible to infectious diseases and might also affect their ability to respond to immunization. Here, we review the indications for and possible adverse effects of vaccines in patients with MS, and address issues of vaccination in the context of immunomodulatory therapy for MS.
Collapse
Affiliation(s)
- Micha Loebermann
- Department of Tropical Medicine and Infectious Diseases, University of Rostock, Schillingallee 35, Rostock, Germany
| | | | | | | | | | | |
Collapse
|