1
|
Drozdov D, Kandil J, Long SE, Demorest CV, Cao Q, Lund TC, Gupta AO, Boelens JJ, Orchard PJ. Bodyweight and Absolute Lymphocyte Count-Based Dosing of Rabbit Anti-thymocyte Globulin Results in Early CD4 + Immune Reconstitution in Patients with Inborn Errors of Metabolism Undergoing Umbilical Cord Blood Transplantation. Transplant Cell Ther 2025; 31:263.e1-263.e7. [PMID: 39914492 DOI: 10.1016/j.jtct.2025.01.893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/14/2025] [Accepted: 01/31/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Rabbit anti-thymocyte globulin (rATG) decreases the risk of graft failure and graft-versus-host disease (GVHD) in a setting of allogenic hematopoietic cell transplantation (HCT) but has highly variable pharmacokinetics. Recently, it was shown that a dosing nomogram based on recipient bodyweight and absolute lymphocyte count reduced rATG overexposure, which led to faster immune reconstitution. The aim of this study is to evaluate the feasibility and benefits of using an rATG dosing nomogram to achieve early CD4+ immune reconstitution in pediatric patients with inborn errors of metabolism (IEM) undergoing umbilical cord blood transplantation. METHODS The rATG dosing nomogram in pediatric patients with IEM receiving an umbilical cord blood transplant with busulfan-based myeloablative conditioning at the University of Minnesota Masonic Children's Hospital was used prospectively since 2017. The primary endpoint was CD4+ immune reconstitution (>50 CD4+ T-cells/mL) within 100 days after HCT. Secondary endpoints included overall survival, graft failure, acute and chronic GVHD, and viral reactivations. RESULTS A total of 27 patients were included in the study. Median follow-up time was 31 months (interquartile range [IQR], 22-38) and median age was 1.5 years (IQR, 0.7-3.9). The underlying disease was Hurler syndrome in 17 (63%), Hunter syndrome in 4 (15%), and cerebral adrenoleukodystrophy in 4 (15%) patients; 2 patients were transplanted for other IEM. The CD4+ recovery (>50 CD4+ T cells/mL) at 100 days post-HCT was reached in 22 (85%) of 26 patients. Overall survival was 83% (95% confidence interval [CI], 67%-100%). No graft failure was observed. Two (7%) patients developed acute GVHD grade II to IV and no patients had chronic GVHD. Six patients (22%) had cytomegalovirus (CMV) viremia. One patient had Epstein-Barr virus reactivation requiring treatment. CONCLUSION In patients with IEM, individualized dosing of rATG was associated with a robust and early CD4+ immune reconstitution, with no graft failures and low GVHD incidence.
Collapse
Affiliation(s)
- Daniel Drozdov
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota; Division of Stem Cell Transplantation and Cellular Therapies and Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland; Division of Pediatric Hematology Oncology, Children's Hospital, Kantonsspital Aarau, Aarau, Switzerland.
| | - Jessica Kandil
- Division of Stem Cell Transplantation and Cellular Therapies and Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland; University of Zürich, Zürich, Switzerland
| | - Susie E Long
- Acute Care Pharmacy Services, University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota
| | - Connor V Demorest
- Biostatistics Core at Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Qing Cao
- Biostatistics Core at Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Troy C Lund
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Ashish O Gupta
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul J Orchard
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
2
|
Yska HAF, Engelen M, Bugiani M. The pathology of X-linked adrenoleukodystrophy: tissue specific changes as a clue to pathophysiology. Orphanet J Rare Dis 2024; 19:138. [PMID: 38549180 PMCID: PMC10976706 DOI: 10.1186/s13023-024-03105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024] Open
Abstract
Although the pathology of X-linked adrenoleukodystrophy (ALD) is well described, it represents the end-stage of neurodegeneration. It is still unclear what cell types are initially involved and what their role is in the disease process. Revisiting the seminal post-mortem studies from the 1970s can generate new hypotheses on pathophysiology. This review describes (histo)pathological changes of the brain and spinal cord in ALD. It aims at integrating older works with current insights and at providing an overarching theory on the pathophysiology of ALD. The data point to an important role for axons and glia in the pathology of both the myelopathy and leukodystrophy of ALD. In-depth pathological analyses with new techniques could help further unravel the sequence of events behind the pathology of ALD.
Collapse
Affiliation(s)
- Hemmo A F Yska
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Marc Engelen
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Pathology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Hashemi E, Narain Srivastava I, Aguirre A, Tilahan Yoseph E, Kaushal E, Awani A, Kyu. Ryu J, Akassoglou K, Talebian S, Chu P, Pisani L, Musolino P, Steinman L, Doyle K, Robinson WH, Sharpe O, Cayrol R, Orchard P, Lund T, Vogel H, Lenail M, Han MH, Bonkowsky JL, Van Haren KP. A novel mouse model of cerebral adrenoleukodystrophy highlights NLRP3 activity in lesion pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.564025. [PMID: 37986739 PMCID: PMC10659266 DOI: 10.1101/2023.11.07.564025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Objective We sought to create and characterize a mouse model of the inflammatory, cerebral demyelinating phenotype of X-linked adrenoleukodystrophy (ALD) that would facilitate the study of disease pathogenesis and therapy development. We also sought to cross-validate potential therapeutic targets such as fibrin, oxidative stress, and the NLRP3 inflammasome, in post-mortem human and murine brain tissues. Background ALD is caused by mutations in the gene ABCD1 encoding a peroxisomal transporter. More than half of males with an ABCD1 mutation develop the cerebral phenotype (cALD). Incomplete penetrance and absence of a genotype-phenotype correlation imply a role for environmental triggers. Mechanistic studies have been limited by the absence of a cALD phenotype in the Abcd1-null mouse. Methods We generated a cALD phenotype in 8-week-old, male Abcd1-null mice by deploying a two-hit method that combines cuprizone (CPZ) and experimental autoimmune encephalomyelitis (EAE) models. We employed in vivo MRI and post-mortem immunohistochemistry to evaluate myelin loss, astrogliosis, blood-brain barrier (BBB) disruption, immune cell infiltration, fibrin deposition, oxidative stress, and Nlrp3 inflammasome activation in mice. We used bead-based immunoassay and immunohistochemistry to evaluate IL-18 in CSF and post-mortem human cALD brain tissue. Results MRI studies revealed T2 hyperintensities and post-gadolinium enhancement in the medial corpus callosum of cALD mice, similar to human cALD lesions. Both human and mouse cALD lesions shared common histologic features of myelin phagocytosis, myelin loss, abundant microglial activation, T and B-cell infiltration, and astrogliosis. Compared to wild-type controls, Abcd1-null mice had more severe cerebral inflammation, demyelination, fibrin deposition, oxidative stress, and IL-18 activation. IL-18 immunoreactivity co-localized with macrophages/microglia in the perivascular region of both human and mouse brain tissue. Interpretation This novel mouse model of cALD suggests loss of Abcd1 function predisposes to more severe cerebral inflammation, oxidative stress, fibrin deposition, and Nlrp3 pathway activation, which parallels the findings seen in humans with cALD. We expect this model to enable long-sought investigations into cALD mechanisms and accelerate development of candidate therapies for lesion prevention, cessation, and remyelination.
Collapse
Affiliation(s)
- Ezzat Hashemi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Isha Narain Srivastava
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Alejandro Aguirre
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Ezra Tilahan Yoseph
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Esha Kaushal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Avni Awani
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jae Kyu. Ryu
- Gladstone Institute for Neurological Disease; San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF; San Francisco, CA USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco; San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institute for Neurological Disease; San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF; San Francisco, CA USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco; San Francisco, CA, USA
| | - Shahrzad Talebian
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Pauline Chu
- Stanford Human Research Histology Core, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Pisani
- Department of Radiology, Stanford University School of Medicine Stanford, CA, USA
| | - Patricia Musolino
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristian Doyle
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - William H Robinson
- Department of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Orr Sharpe
- Department of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Romain Cayrol
- Department of Pathology, Clinical Department of Laboratory Medicine, University of Montreal, Quebec, Canada
| | - Paul Orchard
- Division of Pediatric Blood & Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Troy Lund
- Division of Pediatric Blood & Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Hannes Vogel
- Departments of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Max Lenail
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - May Htwe Han
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua Leith Bonkowsky
- Division of Pediatric Neurology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
- Brain and Spine Center, Primary Children’s Hospital, Salt Lake City, Utah
- Primary Children’s Center for Personalized Medicine, Salt Lake City, Utah
| | - Keith P. Van Haren
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
4
|
Lauer A, Speroni SL, Choi M, Da X, Duncan C, McCarthy S, Krishnan V, Lusk CA, Rohde D, Hansen MB, Kalpathy-Cramer J, Loes DJ, Caruso PA, Williams DA, Mouridsen K, Emblem KE, Eichler FS, Musolino PL. Hematopoietic stem-cell gene therapy is associated with restored white matter microvascular function in cerebral adrenoleukodystrophy. Nat Commun 2023; 14:1900. [PMID: 37019892 PMCID: PMC10076264 DOI: 10.1038/s41467-023-37262-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
Blood-brain barrier disruption marks the onset of cerebral adrenoleukodystrophy (CALD), a devastating cerebral demyelinating disease caused by loss of ABCD1 gene function. The underlying mechanism are not well understood, but evidence suggests that microvascular dysfunction is involved. We analyzed cerebral perfusion imaging in boys with CALD treated with autologous hematopoietic stem-cells transduced with the Lenti-D lentiviral vector that contains ABCD1 cDNA as part of a single group, open-label phase 2-3 safety and efficacy study (NCT01896102) and patients treated with allogeneic hematopoietic stem cell transplantation. We found widespread and sustained normalization of white matter permeability and microvascular flow. We demonstrate that ABCD1 functional bone marrow-derived cells can engraft in the cerebral vascular and perivascular space. Inverse correlation between gene dosage and lesion growth suggests that corrected cells contribute long-term to remodeling of brain microvascular function. Further studies are needed to explore the longevity of these effects.
Collapse
Affiliation(s)
- Arne Lauer
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neuroradiology, Heidelberg University, Heidelberg, Germany
| | - Samantha L Speroni
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Myoung Choi
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Xiao Da
- Functional Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
| | - Christine Duncan
- Dana-Farber and Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, MA, USA
| | - Siobhan McCarthy
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Vijai Krishnan
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Cole A Lusk
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - David Rohde
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Mikkel Bo Hansen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Daniel J Loes
- Suburban Radiologic Consultants, Ltd, Minneapolis, MN, USA
| | - Paul A Caruso
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - David A Williams
- Dana-Farber and Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, MA, USA
| | - Kim Mouridsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Kyrre E Emblem
- Department of Diagnostic Physics, Oslo University Hospital, Oslo, Norway
| | - Florian S Eichler
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Patricia L Musolino
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Athinoula A. Martinos Centre for Biomedical Imaging, Charlestown, MA, USA.
| |
Collapse
|
5
|
Al-Ahmad AJ. Human-Induced Pluripotent Stem Cell-Based Model of the Blood-Brain at 10 Years: A Retrospective on Past and Current Disease Models. Handb Exp Pharmacol 2023; 281:141-156. [PMID: 36943490 DOI: 10.1007/164_2023_645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
The initial discovery and derivation of induced pluripotent stem cells (iPSCs) by Yamanaka and colleagues in 2006 revolutionized the field of personalized medicine, as it opened the possibility to model diseases using patient-derived stem cells. A decade of adoption of iPSCs within the community of the blood-brain barrier (BBB) significantly opened the door for modeling diseases at the BBB, a task until then considered challenging, if not impossible.In this book chapter, we provided an extensive review of the literature on the use of iPSC-based models of the human BBB to model neurological diseases including infectious diseases (COVID-19, Streptococcus, Neisseria) neurodevelopmental diseases (adrenoleukodystrophy, Allan-Herndon-Dudley Syndrome, Batten's disease, GLUT1 deficiency syndrome), and neurodegenerative diseases (Alzheimer's disease, the current findings and observations, but also the challenges and limitations inherent to the use of iPSC-based models in reproducing the human BBB during health and diseases in a Petri dish.
Collapse
Affiliation(s)
- Abraham J Al-Ahmad
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
| |
Collapse
|
6
|
Cebeci H, Gencturk M, Koksel Y, Nascene D. Contrast enhancement in cerebral adrenoleukodystrophy: a comparison of T1 TSE and MPRAGE sequences. Jpn J Radiol 2022; 40:1241-1245. [PMID: 35821375 DOI: 10.1007/s11604-022-01309-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/15/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE To compare conventional T1 TSE with MPRAGE for enhancement detection in cerebral adrenoleukodystrophy (CALD). MATERIALS AND METHODS Contrast-enhanced T1 TSE and MPRAGE sequences of 34 CALD patients demonstrating enhancement were evaluated. Contrast ratios were calculated by drawing ROIs to the most enhancing part of demyelination and normal-appearing deep white matter on both T1 TSE and MPRAGE. A comparison was performed between ratios using paired T test. RESULTS Mean age of 34 included male children was 8 (5-11 years). There was no statistically significant difference between T1 TSE and MPRAGE ratios. However, in 4 out of 34 examinations, minimal contrast enhancement was noted only in T1 TSE sequence. CONCLUSION Our data indicate that both T1 TSE and MPRAGE sequences are valuable in determining contrast enhancement in CALD. Although there is not a statistically significant difference between the two techniques, T1 TSE sequence appears to be more sensitive for low degree of enhancement.
Collapse
Affiliation(s)
- Hakan Cebeci
- Division of Neuroradiology, Department of Radiology, University of Minnesota Medical Center, 420 Delaware Street SE, Minneapolis, USA.,Department of Radiology, Faculty of Medicine, Selçuk University, Konya, Turkey
| | - Mehmet Gencturk
- Division of Neuroradiology, Department of Radiology, University of Minnesota Medical Center, 420 Delaware Street SE, Minneapolis, USA.
| | - Yasemin Koksel
- Division of Neuroradiology, Department of Radiology, University of Minnesota Medical Center, 420 Delaware Street SE, Minneapolis, USA
| | - David Nascene
- Division of Neuroradiology, Department of Radiology, University of Minnesota Medical Center, 420 Delaware Street SE, Minneapolis, USA
| |
Collapse
|
7
|
Alarcan H, Al Ojaimi Y, Lanznaster D, Escoffre JM, Corcia P, Vourc'h P, Andres CR, Veyrat-Durebex C, Blasco H. Taking Advantages of Blood–Brain or Spinal Cord Barrier Alterations or Restoring Them to Optimize Therapy in ALS? J Pers Med 2022; 12:jpm12071071. [PMID: 35887567 PMCID: PMC9319288 DOI: 10.3390/jpm12071071] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder that still lacks an efficient therapy. The barriers between the central nervous system (CNS) and the blood represent a major limiting factor to the development of drugs for CNS diseases, including ALS. Alterations of the blood–brain barrier (BBB) or blood–spinal cord barrier (BSCB) have been reported in this disease but still require further investigations. Interestingly, these alterations might be involved in the complex etiology and pathogenesis of ALS. Moreover, they can have potential consequences on the diffusion of candidate drugs across the brain. The development of techniques to bypass these barriers is continuously evolving and might open the door for personalized medical approaches. Therefore, identifying robust and non-invasive markers of BBB and BSCB alterations can help distinguish different subgroups of patients, such as those in whom barrier disruption can negatively affect the delivery of drugs to their CNS targets. The restoration of CNS barriers using innovative therapies could consequently present the advantage of both alleviating the disease progression and optimizing the safety and efficiency of ALS-specific therapies.
Collapse
Affiliation(s)
- Hugo Alarcan
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Yara Al Ojaimi
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Debora Lanznaster
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Jean-Michel Escoffre
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Philippe Corcia
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
- Service de Neurologie, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
| | - Patrick Vourc'h
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Christian R Andres
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Charlotte Veyrat-Durebex
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Hélène Blasco
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| |
Collapse
|
8
|
de Vasconcelos P, Lacerda JF. Hematopoietic Stem Cell Transplantation for Neurological Disorders: A Focus on Inborn Errors of Metabolism. Front Cell Neurosci 2022; 16:895511. [PMID: 35693884 PMCID: PMC9178264 DOI: 10.3389/fncel.2022.895511] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022] Open
Abstract
Hematopoietic stem cells have been investigated and applied for the treatment of certain neurological disorders for a long time. Currently, their therapeutic potential is harnessed in autologous and allogeneic hematopoietic stem cell transplantation (HSCT). Autologous HSCT is helpful in immune-mediated neurological diseases such as Multiple Sclerosis. However, clinical benefits derive more from the immunosuppressive conditioning regimen than the interaction between stem cells and the nervous system. Mainly used for hematologic malignancies, allogeneic HSCT explores the therapeutic potential of donor-derived hematopoietic stem cells. In the neurological setting, it has proven to be most valuable in Inborn Errors of Metabolism, a large spectrum of multisystem disorders characterized by congenital deficiencies in enzymes involved in metabolic pathways. Inborn Errors of Metabolism such as X-linked Adrenoleukodystrophy present with brain accumulation of enzymatic substrates that result in progressive inflammatory demyelination. Allogeneic HSCT can halt ongoing inflammatory neural destruction by replacing hematopoietic-originated microglia with donor-derived myeloid precursors. Microglia, the only neural cells successfully transplanted thus far, are the most valuable source of central nervous system metabolic correction and play a significant role in the crosstalk between the brain and hematopoietic stem cells. After transplantation, engrafted donor-derived myeloid cells modulate the neural microenvironment by recapitulating microglial functions and enhancing repair mechanisms such as remyelination. In some disorders, additional benefits result from the donor hematopoietic stem cell secretome that cross-corrects neighboring neural cells via mannose-6-phosphatase paracrine pathways. The limitations of allogeneic HSCT in this setting relate to the slow turnover of microglia and complications such as graft-vs.-host disease. These restraints have accelerated the development of hematopoietic stem cell gene therapy, where autologous hematopoietic stem cells are collected, manipulated ex vivo to overexpress the missing enzyme, and infused back into the patient. With this cellular drug vehicle strategy, the brain is populated by improved cells and exposed to supraphysiological levels of the flawed protein, resulting in metabolic correction. This review focuses on the mechanisms of brain repair resulting from HSCT and gene therapy in Inborn Errors of Metabolism. A brief mention will also be made on immune-mediated nervous system diseases that are treated with this approach.
Collapse
Affiliation(s)
- Pedro de Vasconcelos
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - João F. Lacerda
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
9
|
Umeda K. Unresolved issues in allogeneic hematopoietic cell transplantation for non-malignant diseases. Int J Hematol 2022; 116:41-47. [PMID: 35568772 DOI: 10.1007/s12185-022-03361-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/27/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) can be curative for a variety of non-malignant diseases (NMDs) as well as hematological malignancies. However, there are several fundamental differences between HCT for NMDs and hematological malignancies, which may necessitate the use of alternative HCT strategies. For example, these diseases differ in the intensity of conditioning regimen sufficient to improve disease. In addition, patients with NMDs are at higher risk of graft failure or mixed chimerism following HCT, and gain no or little survival benefit from graft-versus-host disease. Because more than 80% of patients with NMDs become long-term survivors, greater attention has been paid to late adverse effects and decreased of quality of life after HCT. This review addresses several unresolved issues in allogeneic HCT for patients with NMDs, such as (1) stem cell source, (2) conditioning regimen, (3) use of serotherapy or low-dose irradiation, and (4) therapeutic intervention for mixed chimerism. Resolving these issues may improve transplant outcomes in patients with NMDs.
Collapse
Affiliation(s)
- Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
10
|
Wang J, Xu J, Zang G, Zhang T, Wu Q, Zhang H, Chen Y, Wang Y, Qin W, Zhao S, Qin E, Qiu J, Zhang X, Wen L, Wang Y, Wang G. trans-2-Enoyl-CoA Reductase Tecr-Driven Lipid Metabolism in Endothelial Cells Protects against Transcytosis to Maintain Blood-Brain Barrier Homeostasis. RESEARCH 2022; 2022:9839368. [PMID: 35465346 PMCID: PMC9006154 DOI: 10.34133/2022/9839368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/02/2022] [Indexed: 12/29/2022]
Abstract
The transport and metabolism of lipids in cerebrovascular endothelial cells (ECs) have been hypothesized to regulate blood-brain barrier (BBB) maturation and homeostasis. Long-chain polyunsaturated fatty acids (LCPUFAs) as the important lipids components of cell membranes are essential for the development and function of BBB, but the direct links of lipid metabolism and ECs barrier function remain to be established. Here, we comprehensively characterize the transcriptomic phenotype of developmental cerebrovascular ECs in single-cell resolution and firstly find that trans-2-enoyl-CoA reductase (Tecr), a very-long-chain fatty acid synthesis, is highly expressed during barriergenesis and decreased after BBB maturation. EC-specific knockout of Tecr compromises angiogenesis due to delayed vascular sprouting. Importantly, EC-specific deletion of Tecr loss restrictive quality of vascular permeability from neonatal stages to adulthood, with high levels of transcytosis, but maintains the vascular tight junctions. Moreover, lipidomic analysis shows that the expression of Tecr in ECs is associated with the containing of omega-3 fatty acids, which directly suppresses caveolae vesicles formation. These results reveal a protective role for Tecr in BBB integrity and suggest that Tecr as a novel therapeutic target in the central nervous system (CNS) diseases associated with BBB dysfunction.
Collapse
Affiliation(s)
- Jinxuan Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Jianxiong Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Guangchao Zang
- Institute of Life Science, Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China
| | - Tao Zhang
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Qi Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Hongping Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yidan Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Weixi Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Shuang Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Erdai Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Xiaojuan Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Lin Wen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yeqi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| |
Collapse
|
11
|
Wang H, Davison MD, Kramer ML, Qiu W, Gladysheva T, Chiang RMS, Kayatekin C, Nascene DR, Taghizadeh LA, King CJ, Nolan EE, Gupta AO, Orchard PJ, Lund TC. Evaluation of Neurofilament Light Chain as a Biomarker of Neurodegeneration in X-Linked Childhood Cerebral Adrenoleukodystrophy. Cells 2022; 11:913. [PMID: 35269535 PMCID: PMC8909395 DOI: 10.3390/cells11050913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 01/23/2023] Open
Abstract
Cerebral adrenoleukodystrophy (CALD) is a devastating, demyelinating neuroinflammatory manifestation found in up to 40% of young males with an inherited mutation in ABCD1, the causative gene in adrenoleukodystrophy. The search for biomarkers which correlate to CALD disease burden and respond to intervention has long been sought after. We used the Olink Proximity Extension Assay (Uppsala, Sweden) to explore the cerebral spinal fluid (CSF) of young males with CALD followed by correlative analysis with plasma. Using the Target 96 Neuro Exploratory panel, we found that, of the five proteins significantly increased in CSF, only neurofilament light chain (NfL) showed a significant correlation between CSF and plasma levels. Young males with CALD had a 11.3-fold increase in plasma NfL compared with controls. Importantly, 9 of 11 young males with CALD who underwent HCT showed a mean decrease in plasma NfL of 50% at 1 year after HCT compared with pre-HCT levels. In conclusion, plasma NfL could be a great value in determining outcomes in CALD and should be scrutinized in future studies in patients prior to CALD development and after therapeutic intervention.
Collapse
Affiliation(s)
- Hongge Wang
- Translational Sciences, Sanofi Research, Sanofi, Framingham, MA 01701, USA; (H.W.); (M.D.D.); (M.L.K.)
| | - Matthew D. Davison
- Translational Sciences, Sanofi Research, Sanofi, Framingham, MA 01701, USA; (H.W.); (M.D.D.); (M.L.K.)
| | - Martin L. Kramer
- Translational Sciences, Sanofi Research, Sanofi, Framingham, MA 01701, USA; (H.W.); (M.D.D.); (M.L.K.)
| | - Weiliang Qiu
- Nonclinical Efficacy and Safety, Department of Biostatistics and Programming, Sanofi Development, Sanofi, Framingham, MA 01701, USA;
| | - Tatiana Gladysheva
- Integrated Drug Discovery, Sanofi Research, Sanofi, Waltham, MA 02451, USA;
| | - Ruby M. S. Chiang
- Rare and Neurological Diseases Research Therapeutic Area, Sanofi, 49 New York Avenue, Framingham, MA 01701, USA; (R.M.S.C.); (C.K.)
| | - Can Kayatekin
- Rare and Neurological Diseases Research Therapeutic Area, Sanofi, 49 New York Avenue, Framingham, MA 01701, USA; (R.M.S.C.); (C.K.)
| | - David R. Nascene
- Department of Diagnostic Radiology, University of Minnesota Medical Center, Minneapolis, MN 55455, USA;
| | - Leyla A. Taghizadeh
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA; (L.A.T.); (C.J.K.); (E.E.N.); (A.O.G.); (P.J.O.)
| | - Carina J. King
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA; (L.A.T.); (C.J.K.); (E.E.N.); (A.O.G.); (P.J.O.)
| | - Erin E. Nolan
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA; (L.A.T.); (C.J.K.); (E.E.N.); (A.O.G.); (P.J.O.)
| | - Ashish O. Gupta
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA; (L.A.T.); (C.J.K.); (E.E.N.); (A.O.G.); (P.J.O.)
| | - Paul J. Orchard
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA; (L.A.T.); (C.J.K.); (E.E.N.); (A.O.G.); (P.J.O.)
| | - Troy C. Lund
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA; (L.A.T.); (C.J.K.); (E.E.N.); (A.O.G.); (P.J.O.)
| |
Collapse
|
12
|
Yalcin K, Çelen SS, Daloglu H, Demir MK, Öztürkmen S, Pasayev D, Zhumatayev S, Uygun V, Hazar V, Karasu G, Yesilipek A. Allogeneic hematopoietic stem cell transplantation in patients with childhood cerebral adrenoleukodystrophy: A single-center experience "Better prognosis in earlier stage". Pediatr Transplant 2021; 25:e14015. [PMID: 33780114 DOI: 10.1111/petr.14015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/25/2021] [Accepted: 03/11/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND ALD is a rare X-linked peroxisomal metabolic disorder with many distinct phenotypes of disease that emerge on a wide scale from adrenal insufficiency to fatal cALD which progresses to a vegetative state within a few years. Currently, HSCT is the only treatment method known to stabilize disease progression in patients with cALD. In this study, we aim to report our HSCT experience in patients with cALD and the factors that determine the success of HSCT, as a single-center experience. METHODS The study cohort involves 23 boys with cALD and three patients with ALD trait and new-onset abnormal behavior who underwent allogeneic HSCT between January 2012 and September 2019 in our transplantation center. Loes scoring, NFS, scale and MFD were performed for evaluating the severity of the cerebral disease. The study cohort was divided into two groups according to baseline NFS and Loes score: early-stage (NFS ≤ 1 and Loes score <9) and advanced stage (NFS > 1 or Loes score ≥9). RESULTS The pretransplant stage of disease impacted both OS and MFD-free survival. The estimated OS and MFD-free survival at 3 years in patients with advanced disease were 46.1% (95% CI 19.0-73.2) and 23.1% (95% CI 0.2-46.0), respectively, and all patients with the early disease were alive (p: .004) and MFD-free (p < .001) at 3 years. CONCLUSION This study demonstrated that early HSCT is vital in patients with cALD. The early-stage disease had a significant survival advantage and free from disease progression after HSCT.
Collapse
Affiliation(s)
- Koray Yalcin
- Medicalpark Goztepe Hospital Pediatric Stem Cell Transplantation Unit, Istanbul, Turkey
| | - Suna S Çelen
- Medicalpark Goztepe Hospital Pediatric Stem Cell Transplantation Unit, Istanbul, Turkey
| | - Hayriye Daloglu
- Medicalpark Antalya Hospital Pediatric Stem Cell Transplantation Unit, Antalya, Turkey
| | - Mustafa Kemal Demir
- Department of Radiology, Göztepe Medical Park Training and Education Hospital, Bahçeşehir University School of Medicine, Istanbul, Turkey
| | - Seda Öztürkmen
- Medicalpark Antalya Hospital Pediatric Stem Cell Transplantation Unit, Antalya, Turkey
| | - Dayanat Pasayev
- Medicalpark Goztepe Hospital Pediatric Stem Cell Transplantation Unit, Istanbul, Turkey
| | - Suleimen Zhumatayev
- Medicalpark Goztepe Hospital Pediatric Stem Cell Transplantation Unit, Istanbul, Turkey
| | - Vedat Uygun
- Medicalpark Antalya Hospital Pediatric Stem Cell Transplantation Unit, Antalya, Turkey
| | - Volkan Hazar
- Medicalpark Goztepe Hospital Pediatric Stem Cell Transplantation Unit, Istanbul, Turkey
| | - Gulsun Karasu
- Medicalpark Goztepe Hospital Pediatric Stem Cell Transplantation Unit, Istanbul, Turkey.,Medicalpark Antalya Hospital Pediatric Stem Cell Transplantation Unit, Antalya, Turkey
| | - Akif Yesilipek
- Medicalpark Goztepe Hospital Pediatric Stem Cell Transplantation Unit, Istanbul, Turkey.,Medicalpark Antalya Hospital Pediatric Stem Cell Transplantation Unit, Antalya, Turkey
| |
Collapse
|
13
|
Morita M, Kaizawa T, Yoda T, Oyama T, Asakura R, Matsumoto S, Nagai Y, Watanabe Y, Watanabe S, Kobayashi H, Kawaguchi K, Yamamoto S, Shimozawa N, So T, Imanaka T. Bone marrow transplantation into Abcd1-deficient mice: Distribution of donor derived-cells and biological characterization of the brain of the recipient mice. J Inherit Metab Dis 2021; 44:718-727. [PMID: 33332637 DOI: 10.1002/jimd.12346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 01/18/2023]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a severe inherited metabolic disease with cerebral inflammatory demyelination and abnormal accumulation of very long chain fatty acid (VLCFA) in tissues, especially the brain. At present, bone marrow transplantation (BMT) at an early stage of the disease is the only effective treatment for halting disease progression, but the underlying mechanism of the treatment has remained unclear. Here, we transplanted GFP-expressing wild-type (WT) or Abcd1-deficient (KO) bone marrow cells into recipient KO mice, which enabled tracking of the donor GFP+ cells in the recipient mice. Both the WT and KO donor cells were equally distributed throughout the brain parenchyma, and displayed an Iba1-positive, GFAP- and Olig2-negative phenotype, indicating that most of the donor cells were engrafted as microglia-like cells. They constituted approximately 40% of the Iba1-positive cells. Unexpectedly, no decrease of VLCFA in the cerebrum was observed when WT bone marrow cells were transplanted into KO mice. Taken together, murine study suggests that bone marrow-derived microglia-like cells engrafted in the cerebrum of X-ALD patients suppress disease progression without evidently reducing the amount of VLCFA in the cerebrum.
Collapse
Affiliation(s)
- Masashi Morita
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Taro Kaizawa
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Taiki Yoda
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Takuro Oyama
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Reina Asakura
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Shun Matsumoto
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yoshinori Nagai
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, Toyama, Japan
| | - Yasuharu Watanabe
- Toyama Prefectural Institute for Pharmaceutical Research, Toyama, Japan
| | - Shiro Watanabe
- Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Hiroshi Kobayashi
- Division of Gene Therapy, Research Center of Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Kosuke Kawaguchi
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Seiji Yamamoto
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Nobuyuki Shimozawa
- Division of Genomics Research, Life Science Research Center, Gifu University, Gifu, Japan
| | - Takanori So
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Tsuneo Imanaka
- Faculty of Pharmaceutical Sciences, Hiroshima International University, Hiroshima, Japan
| |
Collapse
|
14
|
Weinhofer I, Rommer P, Zierfuss B, Altmann P, Foiani M, Heslegrave A, Zetterberg H, Gleiss A, Musolino PL, Gong Y, Forss-Petter S, Berger T, Eichler F, Aubourg P, Köhler W, Berger J. Neurofilament light chain as a potential biomarker for monitoring neurodegeneration in X-linked adrenoleukodystrophy. Nat Commun 2021; 12:1816. [PMID: 33753741 PMCID: PMC7985512 DOI: 10.1038/s41467-021-22114-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 02/19/2021] [Indexed: 01/23/2023] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD), the most frequent monogenetic disorder of brain white matter, is highly variable, ranging from slowly progressive adrenomyeloneuropathy (AMN) to life-threatening inflammatory brain demyelination (CALD). In this study involving 94 X-ALD patients and 55 controls, we tested whether plasma/serum neurofilament light chain protein (NfL) constitutes an early distinguishing biomarker. In AMN, we found moderately elevated NfL with increased levels reflecting higher grading of myelopathy-related disability. Intriguingly, NfL was a significant predictor to discriminate non-converting AMN from cohorts later developing CALD. In CALD, markedly amplified NfL levels reflected brain lesion severity. In rare cases, atypically low NfL revealed a previously unrecognized smoldering CALD disease course with slowly progressive myelin destruction. Upon halt of brain demyelination by hematopoietic stem cell transplantation, NfL gradually normalized. Together, our study reveals that blood NfL reflects inflammatory activity and progression in CALD patients, thus constituting a potential surrogate biomarker that may facilitate clinical decisions and therapeutic development.
Collapse
Affiliation(s)
- Isabelle Weinhofer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Paulus Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Bettina Zierfuss
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Patrick Altmann
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Martha Foiani
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, University College London, London, UK
| | - Amanda Heslegrave
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, University College London, London, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Andreas Gleiss
- Section for Clinical Biometrics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Patricia L Musolino
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Yi Gong
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Florian Eichler
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick Aubourg
- Kremlin-Bicêtre Hospital, University Paris-Saclay, Paris, France
| | - Wolfgang Köhler
- Department of Neurology, Leukodystrophy Clinic, University of Leipzig Medical Center, Leipzig, Germany
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
15
|
Hayashi RJ. Considerations in Preparative Regimen Selection to Minimize Rejection in Pediatric Hematopoietic Transplantation in Non-Malignant Diseases. Front Immunol 2020; 11:567423. [PMID: 33193340 PMCID: PMC7604384 DOI: 10.3389/fimmu.2020.567423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/25/2020] [Indexed: 01/19/2023] Open
Abstract
The variables that influence the selection of a preparative regimen for a pediatric hematopoietic stem cell transplant procedure encompasses many issues. When one considers this procedure for non-malignant diseases, components in a preparative regimen that were historically developed to reduce malignant tumor burden may be unnecessary. The primary goal of the procedure in this instance becomes engraftment with the establishment of normal hematopoiesis and a normal immune system. Overcoming rejection becomes the primary priority, but pursuit of this goal cannot neglect organ toxicity, or post-transplant morbidity such as graft-versus-host disease or life threatening infections. With the improvements in supportive care, newborn screening techniques for early disease detection, and the expansion of viable donor sources, we have reached a stage where hematopoietic stem cell transplantation can be considered for virtually any patient with a hematopoietic based disease. Advancing preparative regiments that minimize rejection and transplant related toxicity will thus dictate to what extent this medical technology is fully utilized. This mini-review will provide an overview of the origins of conditioning regimens for transplantation and how agents and techniques have evolved to make hematopoietic stem cell transplantation a viable option for children with non-malignant diseases of the hematopoietic system. We will summarize the current state of this facet of the transplant procedure and describe the considerations that come into play in selecting a particular preparative regimen. Decisions within this realm must tailor the treatment to the primary disease condition to ideally achieve an optimal outcome. Finally, we will project forward where advances are needed to overcome the persistent engraftment obstacles that currently limit the utilization of transplantation for haematopoietically based diseases in children.
Collapse
Affiliation(s)
- Robert J Hayashi
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
16
|
Zimmerman C, Shenoy S. Chimerism in the Realm of Hematopoietic Stem Cell Transplantation for Non-malignant Disorders-A Perspective. Front Immunol 2020; 11:1791. [PMID: 32903736 PMCID: PMC7438804 DOI: 10.3389/fimmu.2020.01791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/06/2020] [Indexed: 11/21/2022] Open
Abstract
Hematopoietic stem cell transplantation (HCT) is a curative intervention in non-malignant disorders (NMD) that benefit from donor-derived hematopoiesis, immunity, and establishment of vital cells or enzyme systems. Stability or reversal of disease symptoms depends on adequacy and long-term stability of donor cell engraftment in the compartment of interest. Unlike hematologic malignancies where complete replacement with donor derived hematopoiesis is desirable for a cure, NMD manifestations can often be controlled in the presence of mixed chimerism. This allows for exploration of reduced intensity conditioning regimens that can limit organ toxicity, late effects, and increase tolerability especially in young recipients or those with a large burden of disease related morbidity. However, the levels of donor chimerism conducive to disease control vary between NMD, need to focus on the hematopoietic lineage necessary to correct individual disorders, and need to be assessed for stability over time, i.e., a whole lifespan. An enhanced ability to reject grafts due to recipient immune competence, alloimmunization, and autoimmunity add to the complexity of this balance making NMD a highly diverse group of unrelated disorders. The addition of donor factors such as stem cell source and Human-Leukocyte-Antigen match extend the complexity such that 'one size does not fit all'. In this perspective, we will discuss current knowledge of the role of chimerism and goals, approach to HCT, and emerging methods of boosting engraftment and graft function, and monitoring recommendations. We draw attention to knowledge gaps and areas of necessity for further research and research support.
Collapse
|
17
|
Pierpont EI, Nascene DR, Shanley R, Kenney-Jung DL, Ziegler RS, Miller WP, Gupta AO, Lund TC, Orchard PJ, Eisengart JB. Neurocognitive benchmarks following transplant for emerging cerebral adrenoleukodystrophy. Neurology 2020; 95:e591-e600. [PMID: 32616675 PMCID: PMC7455349 DOI: 10.1212/wnl.0000000000009929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/10/2020] [Indexed: 01/16/2023] Open
Abstract
Objective To quantify benchmark treatment outcomes that may be enabled by newborn screening surveillance for X-linked adrenoleukodystrophy (ALD), we report neurocognitive, neuropsychiatric, and MRI change for boys who underwent hematopoietic stem cell transplant (HSCT) at initial stages of demyelination, prior to neurocognitive signs of disease. Methods Retrospective chart review identified 36 patients whose cerebral ALD was detected and treated early, with lesion severity less than 5 on the ALD-specific MRI scoring system. Median age at transplant was 7.3 years (range, 4.0–16.1). Progression of radiologic disease on MRI in the 2 years following HSCT was examined relative to the severity of the initial lesion for 33 patients, and longitudinal neurocognitive and neuropsychiatric outcomes were studied for 30 patients. Results Patients whose pretransplant lesion extended beyond the splenium of the corpus callosum and adjacent periventricular white matter (MRI severity score >2) demonstrated lower posttransplant neurocognitive scores, more neuropsychiatric symptoms, and more disease progression on MRI than patients with a less severe lesion. Changes from baseline neurocognitive functioning were greater at 2 years posttransplant as compared to 1 year. There was greater variance and risk of lesion progression as pretransplant MRI severity increased. Conclusion To realize the full benefits of newborn screening, clinicians must detect very small demyelinating lesions during surveillance and intervene quickly. Novel interventions that reduce risks inherent in allogeneic transplantation are needed. Trial endpoints should include direct neurocognitive assessment and extend at least 2 years posttreatment to provide the greatest sensitivity to detect neurocognitive morbidity.
Collapse
Affiliation(s)
- Elizabeth I Pierpont
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA.
| | - David R Nascene
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA
| | - Ryan Shanley
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA
| | - Daniel L Kenney-Jung
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA
| | - Richard S Ziegler
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA
| | - Weston P Miller
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA
| | - Ashish O Gupta
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA
| | - Troy C Lund
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA
| | - Paul J Orchard
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA
| | - Julie B Eisengart
- From the Departments of Pediatrics (E.I.P., R.S.Z., W.P.M., A.O.G., T.C.L., P.J.O., J.B.E.), Radiology (D.R.N.), Bioinformatics and Biostatistics Core (R.S.), and Neurology (D.L.K.-J.), University of Minnesota, Minneapolis; and Sangamo Therapeutics (W.P.M.), Richmond, CA
| |
Collapse
|
18
|
Lund TC, Ng M, Orchard PJ, Loes DJ, Raymond GV, Gupta A, Kenny-Jung D, Nascene DR. Volume of Gadolinium Enhancement and Successful Repair of the Blood-Brain Barrier in Cerebral Adrenoleukodystrophy. Biol Blood Marrow Transplant 2020; 26:1894-1899. [PMID: 32599216 DOI: 10.1016/j.bbmt.2020.06.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/09/2020] [Accepted: 06/21/2020] [Indexed: 11/25/2022]
Abstract
Up to 40% of boys with adrenoleukodystrophy develop a severe central nervous system demyelinating form (cALD) characterized by white matter changes and gadolinium enhancement on magnetic resonance imaging (MRI). Hematopoietic cell transplant (HCT) is the only proven means to attenuate cALD progression. The elimination of active neuroinflammation is indicated radiographically by the resolution of gadolinium (Gd) enhancement and correlates to speed of donor neutrophil recovery. We analyzed 66 boys with cALD undergoing HCT for biomarkers correlating with early (30 days post-HCT) Gd signal resolution. We found that log Gd volume (cm3) on pre-HCT MRI strongly positively correlated to day 30 Gd resolution (P = .0003) with smaller volume correlating to higher proportion resolved, as was the baseline gadolinium intensity score (P = .04), plasma chitotriosidase activity (P = .04), and faster absolute neutrophil count recovery (P = .03). In multivariate analysis, log Gd volume remained superior in determining which patients would have Gd signal resolution by 30 days post-HCT (P = .016). A final analysis indicated that early Gd resolution also correlated with less neurologic progression from baseline to 1 year following HCT (P = .006). MRI Gd volume may serve as a contributing biomarker to better delineate outcomes and an important metric in comparing therapies in the treatment of cALD.
Collapse
Affiliation(s)
- Troy C Lund
- Division of Pediatric Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota.
| | - Michelle Ng
- Division of Pediatric Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Daniel J Loes
- Department of Diagnostic Radiology, University of Minnesota Medical Center, Minneapolis, Minnesota
| | - Gerald V Raymond
- Department of Genetic Medicine, Johns Hopkins, Baltimore, Maryland
| | - Ashish Gupta
- Division of Pediatric Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Dan Kenny-Jung
- Department of Neurology, University of Minnesota Medical Center, Minneapolis, Minnesota
| | - David R Nascene
- Department of Diagnostic Radiology, University of Minnesota Medical Center, Minneapolis, Minnesota
| |
Collapse
|
19
|
Oxygen-Glucose Deprivation/Reoxygenation-Induced Barrier Disruption at the Human Blood–Brain Barrier is Partially Mediated Through the HIF-1 Pathway. Neuromolecular Med 2019; 21:414-431. [DOI: 10.1007/s12017-019-08531-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
|
20
|
In ALD, I feel the need for speed. Blood 2019; 133:1275-1277. [DOI: 10.1182/blood-2019-02-897629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|