1
|
Al Agrafi F, Gaballa A, Hahn P, Arruda LCM, Jaramillo AC, Witsen M, Lehmann S, Önfelt B, Uhlin M, Stikvoort A. Selective lysis of acute myeloid leukemia cells by CD34/CD3 bispecific antibody through the activation of γδ T-cells. Oncoimmunology 2024; 13:2379063. [PMID: 39076247 PMCID: PMC11285226 DOI: 10.1080/2162402x.2024.2379063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/31/2024] Open
Abstract
Despite the considerable progress in acute myeloid leukemia (AML) treatment, relapse after allogeneic hematopoietic stem cell transplantation (HSCT) is still frequent and associated with a poor prognosis. Relapse has been shown to be correlated with an incomplete eradication of CD34+ leukemic stem cells prior to HSCT. Previously, we have shown that a novel CD34-directed, bispecific T-cell engager (BTE) can efficiently redirect the T-cell effector function toward cancer cells, thus eliminating leukemic cells in vitro and in vivo. However, its impact on γδ T-cells is still unclear. In this study, we tested the efficacy of the CD34-specific BTE using in vitro expanded γδ T-cells as effectors. We showed that the BTEs bind to γδ T-cells and CD34+ leukemic cell lines and induce target cell killing in a dose-dependent manner. Additionally, γδ T-cell mediated killing was found to be superior to αβ T-cell mediated cytotoxicity. Furthermore, we observed that only in the presence of BTE the γδ T-cells induced primary AML blast killing in vitro. Importantly, our results show that γδ T-cells did not target the healthy CD34intermediate endothelial blood-brain barrier cell line (hCMEC/D3) nor lysed CD34+ HSCs from healthy bone marrow samples.
Collapse
MESH Headings
- Humans
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/therapeutic use
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Antigens, CD34/metabolism
- CD3 Complex/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Lymphocyte Activation/immunology
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
Collapse
Affiliation(s)
- Faisal Al Agrafi
- Healthy Aging Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, Kingdom of Saudi Arabia
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Ahmed Gaballa
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
- Department of Biochemistry and Molecular Biology, National Liver Institute, Menoufia University, Shebeen El-Kom, Egypt
| | - Paula Hahn
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Lucas C. M. Arruda
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Adrian C. Jaramillo
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Maartje Witsen
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Sören Lehmann
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
- Department of Medical Sciences, Hematology, Uppsala University Hospital, Uppsala, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Michael Uhlin
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
- Department of Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Arwen Stikvoort
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| |
Collapse
|
2
|
Schoenmakers DH, Mochel F, Adang LA, Boelens JJ, Calbi V, Eklund EA, Grønborg SW, Fumagalli F, Groeschel S, Lindemans C, Sevin C, Schöls L, Ram D, Zerem A, Graessner H, Wolf NI. Inventory of current practices regarding hematopoietic stem cell transplantation in metachromatic leukodystrophy in Europe and neighboring countries. Orphanet J Rare Dis 2024; 19:46. [PMID: 38326898 PMCID: PMC10848395 DOI: 10.1186/s13023-024-03075-3] [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: 11/14/2023] [Accepted: 02/03/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND For decades, early allogeneic stem cell transplantation (HSCT) has been used to slow neurological decline in metachromatic leukodystrophy (MLD). There is lack of consensus regarding who may benefit, and guidelines are lacking. Clinical practice relies on limited literature and expert opinions. The European Reference Network for Rare Neurological Diseases (ERN-RND) and the MLD initiative facilitate expert panels for treatment advice, but some countries are underrepresented. This study explores organizational and clinical HSCT practices for MLD in Europe and neighboring countries to enhance optimization and harmonization of cross-border MLD care. METHODS A web-based EUSurvey was distributed through the ERN-RND and the European Society for Blood and Marrow Transplantation Inborn Errors Working Party. Personal invitations were sent to 89 physicians (43 countries) with neurological/metabolic/hematological expertise. The results were analyzed and visualized using Microsoft Excel and IBM SPSS statistics. RESULTS Of the 30 countries represented by 42 respondents, 23 countries offer HSCT for MLD. The treatment is usually available in 1-3 centers per country (18/23, 78%). Most countries have no or very few MLD patients transplanted during the past 1-5 years. The eligibility criteria regarding MLD subtype, motor function, IQ, and MRI largely differ across countries. CONCLUSION HSCT for MLD is available in most European countries, but uncertainties exist in Eastern and South-Eastern Europe. Applied eligibility criteria and management vary and may not align with the latest scientific insights, indicating physicians' struggle in providing evidence-based care. Interaction between local physicians and international experts is crucial for adequate treatment decision-making and cross-border care in the rapidly changing MLD field.
Collapse
Affiliation(s)
- Daphne H Schoenmakers
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma's Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cellular and Molecular Mechanisms, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Medicine for Society, Platform at Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Fanny Mochel
- Hôpital La Pitié-Salpêtrière, Assistance-Publique Hôpitaux de Paris, Inserm U1127, Paris, France
| | - Laura A Adang
- Division of Child Neurology, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Jaap-Jan Boelens
- Stem Cell Transplantation and Cellular Therapies Program, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Valeria Calbi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Pediatric Immunohematology Unit and Neurology and Neurophysiology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Erik A Eklund
- Section for Pediatric Neurology, Skåne University Hospital and Clinical Sciences, Lund, Lund University, 221 84, Lund, Sweden
| | - Sabine W Grønborg
- Center for Inherited Metabolic Diseases, Department of Pediatrics and Adolescent Medicine and Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Francesca Fumagalli
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Pediatric Immunohematology Unit and Neurology and Neurophysiology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Samuel Groeschel
- Department of Paediatric Neurology and Developmental Medicine, University Children's Hospital, Tübingen, Germany
| | - Caroline Lindemans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
- Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
| | - Caroline Sevin
- Reference Center for Leukodystrophies, Pediatric Neurology Department, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Ludger Schöls
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center of Neurodeenerative Diseases (DZNE), Tübingen, Germany
| | - Dipak Ram
- Department of Paediatric Neurology, Royal Manchester Children's Hospital, Manchester, UK
| | - Ayelet Zerem
- Pediatric Neurology Institute, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Holm Graessner
- Institute for Medical Genetics and Applied Genomics, Center for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Nicole I Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma's Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Cellular and Molecular Mechanisms, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Elnaggar M, Shenoy S. Hyperthyroidism following transplantation and SARS-CoV-2 exposure. Pediatr Blood Cancer 2023; 70:e30713. [PMID: 37792314 DOI: 10.1002/pbc.30713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Affiliation(s)
| | - Shalini Shenoy
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Loeb AM, Pattwell SS, Meshinchi S, Bedalov A, Loeb KR. Donor bone marrow-derived macrophage engraftment into the central nervous system of patients following allogeneic transplantation. Blood Adv 2023; 7:5851-5859. [PMID: 37315172 PMCID: PMC10558597 DOI: 10.1182/bloodadvances.2023010409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/16/2023] Open
Abstract
Hematopoietic stem cell transplantation is a well-known treatment for hematologic malignancies, wherein nascent stem cells provide regenerating marrow and immunotherapy against the tumor. The progeny of hematopoietic stem cells also populate a wide spectrum of tissues, including the brain, as bone marrow-derived macrophages similar to microglial cells. We developed a sensitive and novel combined immunohistochemistry (IHC) and XY fluorescence in situ hybridization assay to detect, quantify, and characterize donor cells in the cerebral cortices of 19 female patients who underwent allogeneic stem cell transplantation. We showed that the number of male donor cells ranged from 0.14% to 3.0% of the total cells or from 1.2% to 25% of microglial cells. Using tyramide-based fluorescent IHC, we found that at least 80% of the donor cells expressed the microglial marker ionized calcium-binding adapter molecule-1, consistent with bone marrow-derived macrophages. The percentage of donor cells was related to pretransplantation conditioning; donor cells from radiation-based myeloablative cases averaged 8.1% of microglial cells, whereas those from nonmyeloablative cases averaged only 1.3%. The number of donor cells in patients conditioned with busulfan- or treosulfan-based myeloablation was similar to that in total body irradiation-based conditioning; donor cells averaged 6.8% of the microglial cells. Notably, patients who received multiple transplantations and those with the longest posttransplantation survival had the highest level of donor engraftment, with donor cells averaging 16.3% of the microglial cells. Our work represents the largest study characterizing bone marrow-derived macrophages in patients after transplantation. The efficiency of engraftment observed in our study warrants future research on microglial replacement as a therapeutic option for disorders of the central nervous system.
Collapse
Affiliation(s)
| | - Siobhan S. Pattwell
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Soheil Meshinchi
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Antonio Bedalov
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Keith R. Loeb
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| |
Collapse
|
5
|
Sheth J, Nair A, Jee B. Lysosomal storage disorders: from biology to the clinic with reference to India. THE LANCET REGIONAL HEALTH. SOUTHEAST ASIA 2023; 9:100108. [PMID: 37383036 PMCID: PMC10305895 DOI: 10.1016/j.lansea.2022.100108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/20/2022] [Accepted: 10/27/2022] [Indexed: 06/30/2023]
Abstract
Lysosomal storage disorders (LSDs) are a group of seventy different metabolic storage diseases due to accumulation of substrate mainly in the form of carbohydrate, lipids, proteins, and cellular debris. They occur due to variant in different genes that regulate lysosomal enzymes synthesis, transport, and secretion. In recent years, due to an increased availability of various therapies to treat these disorders, and increased diagnostic tools, there has been an escalated awareness of LSDs. Due to heterogeneous population and various social reasons, India is likely to have a high frequency of LSDs. Therefore, to understand the burden of various LSDs, its molecular spectrum, and understanding the phenotype-genotype correlation, Indian Council of Medical Research (ICMR) and Department of Health Research (DHR), Government of India had set up a task force in the year 2015. It has resulted in identifying common LSDs, and founder variant for some of the storage disorders and molecular spectrum of various LSDs across the country. This review describes in detail the spectrum of LSDs, its molecular epidemiology and prevention in context to Indian population.
Collapse
Affiliation(s)
- Jayesh Sheth
- FRIGE's Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad 380015, India
| | - Aadhira Nair
- FRIGE's Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad 380015, India
| | - Babban Jee
- Department of Health Research, Ministry of Health and Family Welfare, Government of India, 2nd Floor, IRCS Building, Red Cross Road, New Delhi 110001, India
| |
Collapse
|
6
|
Cohen DL, Salman NA, Conklin HM, Ehrhardt MJ, Potter BS. Case series: Neurocognitive assessment of three siblings with SAMD9-associated monosomy 7/myelodysplastic syndrome. BRAIN DISORDERS 2022. [DOI: 10.1016/j.dscb.2022.100045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
7
|
Sala D, Ornaghi F, Morena F, Argentati C, Valsecchi M, Alberizzi V, Di Guardo R, Bolino A, Aureli M, Martino S, Gritti A. Therapeutic advantages of combined gene/cell therapy strategies in a murine model of GM2 gangliosidosis. Mol Ther Methods Clin Dev 2022; 25:170-189. [PMID: 35434178 PMCID: PMC8983315 DOI: 10.1016/j.omtm.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/13/2022] [Indexed: 11/28/2022]
Abstract
Genetic deficiency of β-N-acetylhexosaminidase (Hex) functionality leads to accumulation of GM2 ganglioside in Tay-Sachs disease and Sandhoff disease (SD), which presently lack approved therapies. Current experimental gene therapy (GT) approaches with adeno-associated viral vectors (AAVs) still pose safety and efficacy issues, supporting the search for alternative therapeutic strategies. Here we leveraged the lentiviral vector (LV)-mediated intracerebral (IC) GT platform to deliver Hex genes to the CNS and combined this strategy with bone marrow transplantation (BMT) to provide a timely, pervasive, and long-lasting source of the Hex enzyme in the CNS and periphery of SD mice. Combined therapy outperformed individual treatments in terms of lifespan extension and normalization of the neuroinflammatory/neurodegenerative phenotypes of SD mice. These benefits correlated with a time-dependent increase in Hex activity and a remarkable reduction in GM2 storage in brain tissues that single treatments failed to achieve. Our results highlight the synergic mode of action of LV-mediated IC GT and BMT, clarify the contribution of treatments to the therapeutic outcome, and inform on the realistic threshold of corrective enzymatic activity. These results have important implications for interpretation of ongoing experimental therapies and for design of more effective treatment strategies for GM2 gangliosidosis.
Collapse
Affiliation(s)
- Davide Sala
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Francesca Ornaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Francesco Morena
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Chiara Argentati
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Manuela Valsecchi
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Via Fratelli Cervi 93, 20090 Segrate, MI, Italy
| | - Valeria Alberizzi
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, 20132 Milan, Italy
| | - Roberta Di Guardo
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, 20132 Milan, Italy
| | - Alessandra Bolino
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, 20132 Milan, Italy
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Via Fratelli Cervi 93, 20090 Segrate, MI, Italy
| | - Sabata Martino
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| |
Collapse
|
8
|
Shibuya Y, Kumar KK, Mader MMD, Yoo Y, Ayala LA, Zhou M, Mohr MA, Neumayer G, Kumar I, Yamamoto R, Marcoux P, Liou B, Bennett FC, Nakauchi H, Sun Y, Chen X, Heppner FL, Wyss-Coray T, Südhof TC, Wernig M. Treatment of a genetic brain disease by CNS-wide microglia replacement. Sci Transl Med 2022; 14:eabl9945. [PMID: 35294256 PMCID: PMC9618306 DOI: 10.1126/scitranslmed.abl9945] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Hematopoietic cell transplantation after myeloablative conditioning has been used to treat various genetic metabolic syndromes but is largely ineffective in diseases affecting the brain presumably due to poor and variable myeloid cell incorporation into the central nervous system. Here, we developed and characterized a near-complete and homogeneous replacement of microglia with bone marrow cells in mice without the need for genetic manipulation of donor or host. The high chimerism resulted from a competitive advantage of scarce donor cells during microglia repopulation rather than enhanced recruitment from the periphery. Hematopoietic stem cells, but not immediate myeloid or monocyte progenitor cells, contained full microglia replacement potency equivalent to whole bone marrow. To explore its therapeutic potential, we applied microglia replacement to a mouse model for Prosaposin deficiency, which is characterized by a progressive neurodegeneration phenotype. We found a reduction of cerebellar neurodegeneration and gliosis in treated brains, improvement of motor and balance impairment, and life span extension even with treatment started in young adulthood. This proof-of-concept study suggests that efficient microglia replacement may have therapeutic efficacy for a variety of neurological diseases.
Collapse
Affiliation(s)
- Yohei Shibuya
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kevin K Kumar
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA,These authors contributed equally
| | - Marius Marc-Daniel Mader
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,These authors contributed equally
| | - Yongjin Yoo
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,These authors contributed equally
| | - Luis Angel Ayala
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mu Zhou
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Gernot Neumayer
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ishan Kumar
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ryo Yamamoto
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paul Marcoux
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Benjamin Liou
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA,Division of Stem Cell Therapy, Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Ying Sun
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Xiaoke Chen
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Frank L. Heppner
- Department of Neuropathology, Cluster of Excellence, NeuroCure, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany,Department of Neuropathology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany,Cluster of Excellence, NeuroCure, Charitéplatz 1, 10117 Berlin, Germany,Berlin Institute of Health (BIH), 10117 Berlin, Germany,German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA,Veterans Administration Palo Alto Healthcare System, Palo Alto, CA 94304, USA
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Lead Contact,Correspondence:
| |
Collapse
|
9
|
Gold NB, Harrison SM, Rowe JH, Gold J, Furutani E, Biffi A, Duncan CN, Shimamura A, Lehmann LE, Green RC. Low frequency of treatable pediatric disease alleles in gnomAD: An opportunity for future genomic screening of newborns. HGG ADVANCES 2022; 3:100059. [PMID: 35047849 PMCID: PMC8756496 DOI: 10.1016/j.xhgg.2021.100059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/20/2021] [Indexed: 01/18/2023] Open
Abstract
Hematopoietic stem cell transplant (HSCT) can prevent progression of several genetic disorders. Although a subset of these disorders are identified on newborn screening panels, others are not identified until irreversible symptoms develop. Genetic testing is an efficient methodology to ascertain pre-symptomatic children, but the penetrance of risk-associated variants in the general population is not well understood. We developed a list of 127 genes associated with disorders treatable with HSCT. We identified likely pathogenic or pathogenic (LP/P) and loss-of-function (LoF) variants in these genes in the Genome Aggregation Database (gnomAD), a dataset containing exome and genome sequencing data from 141,456 healthy adults. Within gnomAD, we identified 59 individuals with a LP/P or LoF variant in 15 genes. Genes were associated with bone marrow failure syndromes, bleeding disorders, primary immunodeficiencies, osteopetrosis, metabolic disorders, and epidermolysis bullosa. In conclusion, few ostensibly healthy adults had genotypes associated with pediatric disorders treatable with HSCTs. Given that most of these disorders do not have biomarkers that could be cheaply and universally assessed on a standard newborn screen, our data suggest that genetic testing may be a complementary approach to traditional newborn screening methodology that has the potential to improve mortality and is not expected to lead to a high burden of false-positive results.
Collapse
Affiliation(s)
- Nina B. Gold
- Massachusetts General Hospital for Children, Division of Medical Genetics and Metabolism, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Jared H. Rowe
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Jessica Gold
- Department of Pediatrics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elissa Furutani
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
| | - Alessandra Biffi
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Christine N. Duncan
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Akiko Shimamura
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Leslie E. Lehmann
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Robert C. Green
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital, Boston, MA, USA
- Ariadne Labs, Boston, MA, USA
| |
Collapse
|
10
|
Selvanathan A, Kinsella J, Moore F, Wynn R, Jones S, Shaw PJ, Wilcken B, Bhattacharya K. Effectiveness of early hematopoietic stem cell transplantation in preventing neurocognitive decline in aspartylglucosaminuria: A case series. JIMD Rep 2021; 61:3-11. [PMID: 34485011 PMCID: PMC8411101 DOI: 10.1002/jmd2.12222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/28/2021] [Accepted: 04/06/2021] [Indexed: 11/06/2022] Open
Abstract
Aspartylglucosaminuria (AGU) (OMIM #208400) is a recessively inherited disorder of glycoprotein catabolism, a subset of the lysosomal storage disorders (LSDs). Deficiency of the enzyme glycosylasparaginase (E.C. 3.5.1.26) leads to accumulation of aspartylglucosamine in various organs and its excretion in the urine. The disease is characterized by an initial period of normal development in infancy, a plateau in childhood, and subsequent regression in adolescence and adulthood. No curative treatments are currently available, leading to a protracted period of significant disability prior to early death. Hematopoietic stem cell transplantation (HSCT) has demonstrated efficacy in other LSDs, by providing enzyme replacement therapy in somatic viscera and decreasing substrate accumulation. Moreover, donor-derived monocytes cross the blood-brain barrier, differentiate into microglia, and secrete enzyme in the central nervous system (CNS). This has been shown to improve neurocognitive outcomes in other LSDs. The evidence to date for HSCT in AGU is varied, with marked improvement in glycosylasparaginase enzyme activity in the CNS in mice models, but varying neurocognitive outcomes in humans. We present a case series of four children with AGU who underwent HSCT at different ages (9 years, 5 years, 5 months, and 7 months of age), with long-term follow-up post-transplant (over 10 years). These cases demonstrate similar neurodevelopmental heterogeneity based on formal developmental assessments. The third case, transplanted prior to the onset of neurocognitive involvement, is developing normally despite a severe phenotype in other family members. This suggests that further research should examine the role of early HSCT in management of AGU.
Collapse
Affiliation(s)
- Arthavan Selvanathan
- Genetic Metabolic Disorders ServiceThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Children's Hospital at Westmead Clinical School, the Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Jane Kinsella
- Manchester Centre for Genomic MedicineUniversity of ManchesterManchesterUK
| | - Francesca Moore
- NSW Biochemical Genetics ServiceThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Robert Wynn
- Department of Blood and Marrow TransplantRoyal Manchester Children's HospitalManchesterUK
| | - Simon Jones
- Manchester Centre for Genomic MedicineUniversity of ManchesterManchesterUK
| | - Peter J. Shaw
- Children's Hospital at Westmead Clinical School, the Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Blood and Marrow Transplant ServiceThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Bridget Wilcken
- Genetic Metabolic Disorders ServiceThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Children's Hospital at Westmead Clinical School, the Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Kaustuv Bhattacharya
- Genetic Metabolic Disorders ServiceThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Children's Hospital at Westmead Clinical School, the Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
11
|
Lee CL, Chuang CK, Hsu CH, Chiu HC, Tu RY, Lo YT, Chang YH, Lin HY, Lin SP. The first mucopolysaccharidosis type VII in a Taiwanese girl: A case report and review of the literature. J Formos Med Assoc 2021; 121:712-717. [PMID: 34420841 DOI: 10.1016/j.jfma.2021.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/24/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022] Open
Abstract
The present study included the first case of mucopolysaccharidosis (MPS) type VII in Taiwan. During pregnancy, the patient was diagnosed with hydrops fetalis and had ascites aspiration 4 times. In the following years, she presented gradually with chronic lung disease, developmental delay, short stature, dysmorphic features of coarse face, macroglossia and pigeon chest with scoliosis. Upon referral at age 4 years, she had corneal clouding, mild limitation of range of motion (ROM) and hepatosplenomegaly. X-ray showed paddle ribs and dysplastic vertebral bodies. MPS was suspected and urine glycosaminoglycans (GAGs) elevated were noted. The leukocyte enzymatic analyses for MPS I, MPS II, MPS IIIB, MPS IVA, and MPS VI were all normal. Afterward, the molecular analysis showed two heterozygous genetic variants of c.104C > A and c.1454C > T in trans in the GUSB gene (NM_000181.4) which were the causes for MPS VII. Then, we checked the leukocyte β-glucuronidase activity for MPS VII and showed extremely low, therefore confirmed the diagnosis. Clinicians should increase the awareness on the early signs of MPS to have a prompt diagnosis and offer the correct treatment like enzyme replacement therapy (ERT) as early as possible.
Collapse
Affiliation(s)
- Chung-Lin Lee
- Department of Pediatrics, MacKay Memorial Hospital, Hsinchu, Taiwan; Institute of Clinical Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan; Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan; Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan; MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Chih-Kuang Chuang
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
| | - Chyong-Hsin Hsu
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
| | - Huei-Ching Chiu
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ru-Yi Tu
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yun-Ting Lo
- Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ya-Hui Chang
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan; Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Hsiang-Yu Lin
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan; Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan; MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.
| | - Shuan-Pei Lin
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan; Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.
| |
Collapse
|
12
|
Carbajal-Rodríguez LM, Pérez-García M, Rodríguez-Herrera R, Rosales HS, Olaya-Vargas A. Long-term evolution of mucopolysaccharidosis type I in twins treated with enzyme replacement therapy plus hematopoietic stem cells transplantation. Heliyon 2021; 7:e07740. [PMID: 34458603 PMCID: PMC8379671 DOI: 10.1016/j.heliyon.2021.e07740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 10/17/2019] [Accepted: 08/05/2021] [Indexed: 11/20/2022] Open
Abstract
Mucopolysaccharidoses (MPSs) are a heterogeneous group of diseases that have in common the accumulation of glycosaminoglycans (mucopolysaccharides) within the lysosome. The diseases are caused by a deficiency of the enzyme α-L-iduronidase which is responsible for the degradation of glycosaminoglycans (GAGs or mucopolysaccharides). More than 100 mutations in the gene have been reported, resulting in marked clinical/response variability. MPSs usually present as multisystem and progressive clinical disorders which affect psychomotor and cardiovascular development, the cornea and the musculoskeletal system. Seven phenotypically distinct diseases have been described, and MPS type I (MPS-I) is divided into three clinical forms: severe (Hurler syndrome), intermediate (Hurler-Scheie syndrome) or mild (Scheie syndrome). For the treatment of MPS-I, Enzyme Replacement Therapy (ERT) with α-L-iduronidase and Hematopoietic Stem Cells Transplantation (HSCT), separately or in combination, have produced clinical improvement, especially with regards cardiovascular symptoms and psychomotor development. This article presents the long-term (more than seven years) follow-up of monochorionic, diamniotic twins who were diagnosed with MPS-I at an early stage, and treated with ERT (from age 10 months) plus HSCT (from age 18 months). Overall, the treatment has facilitated stable development with an overall good response and better control of symptoms associated with MPS-I.
Collapse
Affiliation(s)
- Luis M. Carbajal-Rodríguez
- Lysosomal Storage, Rare and Degenerative Diseases Clinic, Instituto Nacional de Pediatría, Insurgentes Sur 3700 – C, Insurgentes Cuicuilco, Mexico City, 04530, Mexico
| | - Martín Pérez-García
- Bone Marrow Transplantation Unit, Instituto Nacional de Pediatría, Av Insurgentes Sur 3700 c Insurgentes Cuicuilco, Mexico City, 04530, Mexico
| | - Raymundo Rodríguez-Herrera
- Lysosomal Storage, Rare and Degenerative Diseases Clinic, Instituto Nacional de Pediatría, Insurgentes Sur 3700 – C, Insurgentes Cuicuilco, Mexico City, 04530, Mexico
| | - Haydeé Salazar Rosales
- Bone Marrow Transplantation Unit, Instituto Nacional de Pediatría, Av Insurgentes Sur 3700 c Insurgentes Cuicuilco, Mexico City, 04530, Mexico
| | - Alberto Olaya-Vargas
- Bone Marrow Transplantation Unit, Instituto Nacional de Pediatría, Av Insurgentes Sur 3700 c Insurgentes Cuicuilco, Mexico City, 04530, Mexico
| |
Collapse
|
13
|
Sajiki D, Yamashita D, Maemura R, Kitazawa H, Sakaguchi H, Yoshida N, Hama A. Acute pancreatitis following allogeneic hematopoietic stem cell transplantation in children. Int J Hematol 2021; 114:494-501. [PMID: 34291421 DOI: 10.1007/s12185-021-03195-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
Acute pancreatitis (AP) is a potential complication of hematopoietic stem cell transplantation (HSCT), but its incidence and risk factors remain unclear. Thus, we reviewed the cases of 259 consecutive children who received allogeneic HSCT at our institution between January 2000 and December 2017 to determine the incidence and risk factors of AP. Thirteen patients developed AP during a median follow-up period of 4.4 years. The median time from HSCT to AP onset was 80 days (range 29-2426 days), and cumulative incidence (CI) at 4 years was 5.0% [95% confidence interval (95% CI) 2.7-8.3%]. The CI of AP was significantly higher in patients who received bone marrow or peripheral blood stem cells than in those who received cord blood (7.2% versus 0.0% at 4 years, P = 0.02) and was higher in patients who developed grade II-IV acute graft-versus-host disease (GVHD) than in those who did not (31.4% versus 1.4% at 4 years, P < 0.001). Multivariate analysis showed that grade II-IV acute GVHD was an independent risk factor for AP [hazard ratio 15.2 (95% CI 4.1-55.8), P < 0.001] and was strongly associated with post-HSCT AP in children.
Collapse
Affiliation(s)
- Daichi Sajiki
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, 3-35 Michishita-cho, Nakamura-ku, Nagoya, 453-8511, Japan
| | - Daiki Yamashita
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, 3-35 Michishita-cho, Nakamura-ku, Nagoya, 453-8511, Japan
| | - Ryo Maemura
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, 3-35 Michishita-cho, Nakamura-ku, Nagoya, 453-8511, Japan
| | - Hironobu Kitazawa
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, 3-35 Michishita-cho, Nakamura-ku, Nagoya, 453-8511, Japan
| | - Hirotoshi Sakaguchi
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, 3-35 Michishita-cho, Nakamura-ku, Nagoya, 453-8511, Japan
| | - Nao Yoshida
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, 3-35 Michishita-cho, Nakamura-ku, Nagoya, 453-8511, Japan
| | - Asahito Hama
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, 3-35 Michishita-cho, Nakamura-ku, Nagoya, 453-8511, Japan.
| |
Collapse
|
14
|
Sahasrabudhe SA, Kartha RV, Ng M, Basso LM, Mishra U, Cloyd JC, Orchard PJ, Brundage RC, Coles LD. Population Pharmacokinetic Analysis of N-Acetylcysteine in Pediatric Patients With Inherited Metabolic Disorders Undergoing Hematopoietic Stem Cell Transplant. J Clin Pharmacol 2021; 61:1638-1645. [PMID: 34275158 DOI: 10.1002/jcph.1943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/14/2021] [Indexed: 11/06/2022]
Abstract
N-acetylcysteine (NAC) has been used in patients with cerebral adrenoleukodystrophy as an antioxidant agent in association with hematopoietic stem cell transplant (HSCT). However, an understanding of the pharmacokinetic characteristics of intravenous NAC dosing in these patients is limited. If and how NAC pharmacokinetics change following the transplant is unknown. Toward that end, a total of 260 blood samples obtained from 18 pediatric patients with inherited metabolic disorders who underwent HSCT were included in a population pharmacokinetic analysis using nonlinear mixed-effects modeling. NAC clearance (CL) and volume of distribution (V) were explored on 3 occasions: -7, +7, and +21 days relative to transplant. Additionally, the effect of transplant procedure on NAC disposition was explored by accounting for between-occasion variability. The covariate OCC was modeled as a fixed-effect parameter on CL and/or V1. A 2-compartment model adequately described the pharmacokinetics of total NAC. Weight-based allometric scaling on pharmacokinetic parameters was assumed using standard coefficients. Estimates for CL, central (V1), and peripheral volume (V2), and intercompartment clearance were 14.7 L/h, 23.2 L, 17.1 L, 3.99 L/h, respectively, for a 70-kg person. The data only supported between-subject variability in CL (12%) and V1 (41%). Residual variability was estimated to be 16%. HSCT did not change CL and V1 significantly, and analysis across occasions did not reveal any trends. Pharmacokinetic parameter estimates were in general comparable to those reported previously in different populations. These results suggest that dosing of NAC does not need to be altered following HSCT.
Collapse
Affiliation(s)
- Siddhee A Sahasrabudhe
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michelle Ng
- Department of Hematology and Oncology, Perth Children's Hospital, Nedlands, WA, Australia.,Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lisa M Basso
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Usha Mishra
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - James C Cloyd
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Richard C Brundage
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lisa D Coles
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
15
|
Köse S, Aerts-Kaya F, Uçkan Çetinkaya D, Korkusuz P. Stem Cell Applications in Lysosomal Storage Disorders: Progress and Ongoing Challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:135-162. [PMID: 33977438 DOI: 10.1007/5584_2021_639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Lysosomal storage disorders (LSDs) are rare inborn errors of metabolism caused by defects in lysosomal function. These diseases are characterized by accumulation of completely or partially degraded substrates in the lysosomes leading to cellular dysfunction of the affected cells. Currently, enzyme replacement therapies (ERTs), treatments directed at substrate reduction (SRT), and hematopoietic stem cell (HSC) transplantation are the only treatment options for LSDs, and the effects of these treatments depend strongly on the type of LSD and the time of initiation of treatment. However, some of the LSDs still lack a durable and curative treatment. Therefore, a variety of novel treatments for LSD patients has been developed in the past few years. However, despite significant progress, the efficacy of some of these treatments remains limited because these therapies are often initiated after irreversible organ damage has occurred.Here, we provide an overview of the known effects of LSDs on stem cell function, as well as a synopsis of available stem cell-based cell and gene therapies that have been/are being developed for the treatment of LSDs. We discuss the advantages and disadvantages of use of hematopoietic stem cell (HSC), mesenchymal stem cell (MSC), and induced pluripotent stem cell (iPSC)-related (gene) therapies. An overview of current research data indicates that when stem cell and/or gene therapy applications are used in combination with existing therapies such as ERT, SRT, and chaperone therapies, promising results can be achieved, showing that these treatments may result in alleviation of existing symptoms and/or prevention of progression of the disease. All together, these studies offer some insight in LSD stem cell biology and provide a hopeful perspective for the use of stem cells. Further development and improvement of these stem cell (gene) combination therapies may greatly improve the current treatment options and outcomes of patients with a LSD.
Collapse
Affiliation(s)
- Sevil Köse
- Department of Medical Biology, Faculty of Medicine, Atilim University, Ankara, Turkey
| | - Fatima Aerts-Kaya
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey.,Hacettepe University Center for Stem Cell Research and Development (PEDI-STEM), Ankara, Turkey
| | - Duygu Uçkan Çetinkaya
- Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Hematology, Hacettepe University Center for Stem Cell Research and Development (PEDI-STEM), Ankara, Turkey.,Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Hacettepe University Faculty of Medicine, Ankara, Turkey.
| |
Collapse
|
16
|
Sugiman-Marangos SN, Beilhartz GL, Zhao X, Zhou D, Hua R, Kim PK, Rini JM, Minassian BA, Melnyk RA. Exploiting the diphtheria toxin internalization receptor enhances delivery of proteins to lysosomes for enzyme replacement therapy. SCIENCE ADVANCES 2020; 6:6/50/eabb0385. [PMID: 33310843 PMCID: PMC7732195 DOI: 10.1126/sciadv.abb0385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 10/21/2020] [Indexed: 05/17/2023]
Abstract
Enzyme replacement therapy, in which a functional copy of an enzyme is injected either systemically or directly into the brain of affected individuals, has proven to be an effective strategy for treating certain lysosomal storage diseases. The inefficient uptake of recombinant enzymes via the mannose-6-phosphate receptor, however, prohibits the broad utility of replacement therapy. Here, to improve the efficiency and efficacy of lysosomal enzyme uptake, we exploited the strategy used by diphtheria toxin to enter into the endolysosomal network of cells by creating a chimera between the receptor-binding fragment of diphtheria toxin and the lysosomal hydrolase TPP1. We show that chimeric TPP1 binds with high affinity to target cells and is efficiently delivered into lysosomes. Further, we show superior uptake of chimeric TPP1 over TPP1 alone in brain tissue following intracerebroventricular injection in mice lacking TPP1, demonstrating the potential of this strategy for enhancing lysosomal storage disease therapy.
Collapse
Affiliation(s)
| | - Greg L Beilhartz
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Xiaochu Zhao
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Dongxia Zhou
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Rong Hua
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Cell Biology Program, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, Canada
| | - Peter K Kim
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Cell Biology Program, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, Canada
| | - James M Rini
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, ON M5S1A8, Canada
| | - Berge A Minassian
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Pediatrics and Dallas Children's Medical Center, University of Texas Southwestern, Dallas, TX 75390-9063, USA
| | - Roman A Melnyk
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Julien DC, Woolgar K, Pollard L, Miller H, Desai A, Lindstrom K, Kishnani PS. Immune Modulation for Enzyme Replacement Therapy in A Female Patient With Hunter Syndrome. Front Immunol 2020; 11:1000. [PMID: 32508845 PMCID: PMC7253587 DOI: 10.3389/fimmu.2020.01000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/27/2020] [Indexed: 12/21/2022] Open
Abstract
A 3.5 year old Hispanic female presented with signs and symptoms concerning for MPS II (Hunter Syndrome). The diagnosis of MPS II was confirmed by enzyme and molecular testing. Genetic evaluation revealed undetectable plasma iduronate-2-sulfatase enzyme activity and an inversion between intron 7 of the IDS gene and a region near exon 3 of IDS-2. This inversion is the molecular cause for ~8% of cases of MPS II and often results in a severe phenotype. X-inactivation studies revealed an inactivation ratio of 100:0. Given the patient's undetectable enzyme level, in combination with a severe IDS gene mutation, classic features at time of presentation, and the significantly skewed X inactivation, there was concern that she was at high risk of developing high and sustained antibody titers to idursulfase which would limit her benefit from enzyme replacement therapy (ERT). Anti-drug neutralizing antibodies to idursulfase have been associated with reduced systemic exposure to idursulfase and poorer clinical outcomes. Therefore, the decision was made to concurrently treat the patient with immune tolerance induction therapy during the first month of treatment with idursulfase in order to decrease the risk of developing high sustained antibody titers. The immune tolerance induction protocol consisted of rituximab weekly for 4 weeks, methotrexate three times a week for 3 weeks and monthly IVIG through B-cell and immunoglobulin recovery. Immune tolerance induction was initiated concurrently with the start of ERT. The patient had no significant adverse effects related to undergoing immune tolerance induction therapy and two and half years later is doing well with significantly reduced urine glycosaminoglycans and very low anti-drug antibody titers. This immune tolerance induction protocol could be considered for other patients with MPS II as well as patients with other lysosomal storage disorders who are starting on enzyme replacement therapy and are at high risk of developing neutralizing anti-drug antibodies.
Collapse
Affiliation(s)
- Daniel C Julien
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Kara Woolgar
- Division of Genetics and Metabolism, Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Laura Pollard
- Division of Medical Genetics, Greenwood, SC, United States
| | - Holly Miller
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Ankit Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Kristin Lindstrom
- Division of Genetics and Metabolism, Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
18
|
Lund TC, Miller WP, Liao AY, Tolar J, Shanley R, Pasquali M, Sando N, Bigger BW, Polgreen LE, Orchard PJ. Post-transplant laronidase augmentation for children with Hurler syndrome: biochemical outcomes. Sci Rep 2019; 9:14105. [PMID: 31575939 PMCID: PMC6773848 DOI: 10.1038/s41598-019-50595-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 09/11/2019] [Indexed: 01/15/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) benefits children with Hurler syndrome (MPS-IH). However, survivors remain burdened by substantial MPS-IH related residual disease. We studied the feasibility, safety and biochemical impact of augmentative recombinant intravenous enzyme replacement therapy (IV-ERT) post transplantation. Ten children with MPS-IH and ≥2 years from successful HCT underwent IV-ERT for 2 years’ duration. Patients were monitored for anti-drug antibody (ADA) development, including inhibitory capacity and changes in urinary excretion of glycosaminoglycans (uGAG). Three patients demonstrated low-level ADA at baseline, though all children tolerated IV-ERT well. Eight patients developed ADA over the 2-year study, with 3 (38%) meeting criteria for an inhibitory ADA response. The aggregate cohort experienced a reduction in uGAG from baseline to study end, which was enhanced in children with low or no ADA response. Conversely, children with inhibitory ADA showed increase in uGAG over time. IV-ERT in previously transplanted children with MPS-IH appears safe and can reduce uGAG, although this is reversed by the presence of inhibitory ADA. These data show a biochemical change after initiation of post-HCT IV-ERT, but the occurrence of ADA and inhibitory antibodies are a concern and should be monitored in future efficacy trials. This trial was registered at www.clinicaltrials.gov, NCT01173016, 07/30/2010.
Collapse
Affiliation(s)
- Troy C Lund
- University of Minnesota, Division of Pediatric Blood and Marrow Transplant, Minneapolis, 55455, USA.
| | - Weston P Miller
- University of Minnesota, Division of Pediatric Blood and Marrow Transplant, Minneapolis, 55455, USA.,Sangamo Therapeutics, Richmond, 94804, USA
| | - Ai Yin Liao
- University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Jakub Tolar
- University of Minnesota, Division of Pediatric Blood and Marrow Transplant, Minneapolis, 55455, USA
| | - Ryan Shanley
- University of Minnesota, Division of Biostatistics, Minneapolis, 55455, USA
| | | | - Nicole Sando
- University of Minnesota, Division of Pediatric Blood and Marrow Transplant, Minneapolis, 55455, USA
| | - Brian W Bigger
- University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Lynda E Polgreen
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, 90502, USA
| | - Paul J Orchard
- University of Minnesota, Division of Pediatric Blood and Marrow Transplant, Minneapolis, 55455, USA
| |
Collapse
|
19
|
Taylor M, Khan S, Stapleton M, Wang J, Chen J, Wynn R, Yabe H, Chinen Y, Boelens JJ, Mason RW, Kubaski F, Horovitz DDG, Barth AL, Serafini M, Bernardo ME, Kobayashi H, Orii KE, Suzuki Y, Orii T, Tomatsu S. Hematopoietic Stem Cell Transplantation for Mucopolysaccharidoses: Past, Present, and Future. Biol Blood Marrow Transplant 2019; 25:e226-e246. [PMID: 30772512 DOI: 10.1016/j.bbmt.2019.02.012] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/11/2019] [Indexed: 12/16/2022]
Abstract
Allogenic hematopoietic stem cell transplantation (HSCT) has proven to be a viable treatment option for a selected group of patients with mucopolysaccharidoses (MPS), including those with MPS types I, II, IVA, VI, and VII. Early diagnosis and timely referral to an expert in MPS are critical, followed by a complete examination and evaluation by a multidisciplinary team, including a transplantation physician. Treatment recommendations for MPS are based on multiple biological, sociological, and financial factors, including type of MPS, clinical severity, prognosis, present clinical signs and symptoms (disease stage), age at onset, rate of progression, family factors and expectations, financial burden, feasibility, availability, risks and benefits of available therapies such as HSCT, enzyme replacement therapy (ERT), surgical interventions, and other supportive care. International collaboration and data review are critical to evaluating the therapeutic efficacy and adverse effects of HSCT for MPS. Collaborative efforts to assess HSCT for MPS have been ongoing since the first attempt at HSCT in a patient with MPS reported in 1981. The accumulation of data since then has made it possible to identify early outcomes (ie, transplantation outcomes) and long-term disease-specific outcomes resulting from HSCT. The recent identification of predictive factors and the development of innovative regimens have significantly improved the outcomes of both engraftment failure and transplantation-related mortality. Assessment of long-term outcomes has considered a variety of factors, including type of MPS, type of graft, age at transplantation, and stage of disease progression, among others. Studies on long-term outcomes are considered a key factor in the use of HSCT in patients with MPS. These studies have shown the effects and limitations of HSCT on improving disease manifestations and quality of life. In this review, we summarize the efficacy, side effects, risks, and cost of HSCT for each type of MPS.
Collapse
Affiliation(s)
- Madeleine Taylor
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Deparment of Biological Science, University of Delaware, Newark, Delaware
| | - Shaukat Khan
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Molly Stapleton
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Deparment of Biological Science, University of Delaware, Newark, Delaware
| | - Jianmin Wang
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Chen
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Robert Wynn
- Department of Paediatric Haematology and Cell Therapy, University of Manchester, Manchester, United Kingdom
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Yasutsugu Chinen
- Department of Pediatrics, Faculty of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert W Mason
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Deparment of Biological Science, University of Delaware, Newark, Delaware
| | - Francyne Kubaski
- Medical Genetics Service, Hospital de ClÃnicas de Porto Alegre (HCPA), Department of Genetics and Molecular Biology- Program Partnership Graduate in Genetics and Molecular Biology (PPGBM), Federal University of Rio Grande do Sul (UFRGS), and National Institute of Populational Medical Genetics (INAGEMP), Porto Alegre, Brazil
| | - Dafne D G Horovitz
- Medical Genetics Department, National Institute of Women, Children, and Adolescent Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Anneliese L Barth
- Medical Genetics Department, National Institute of Women, Children, and Adolescent Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Marta Serafini
- Department of Pediatrics, Dulbecco Telethon Institute, University of Milano-Bicocca, Monza, Italy
| | - Maria Ester Bernardo
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Hironori Kobayashi
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane, Japan
| | - Kenji E Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Gifu, Japan
| | - Tadao Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Department of Pediatrics, Shimane University Faculty of Medicine, Shimane, Japan; Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
20
|
Kobayashi H. Recent trends in mucopolysaccharidosis research. J Hum Genet 2018; 64:127-137. [PMID: 30451936 DOI: 10.1038/s10038-018-0534-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023]
Abstract
Mucopolysaccharidosis (MPS) is a group of inherited conditions involving metabolic dysfunction. Lysosomal enzyme deficiency leads to the accumulation of glycosaminoglycan (GAG) resulting in systemic symptoms, and is categorized into seven types caused by deficiency in one of eleven different enzymes. The pathophysiological mechanism of these diseases has been investigated, indicating impaired autophagy in neuronal damage initiation, association of activated microglia and astrocytes with the neuroinflammatory processes, and involvement of tauopathy. A new inherited error of metabolism resulting in a multisystem disorder with features of the MPS was also identified. Additionally, new therapeutic methods are being developed that could improve conventional therapies, such as new recombinant enzymes that can penetrate the blood brain barrier, hematopoietic stem cell transplantation with reduced intensity conditioning, gene therapy using a viral vector system or gene editing, and substrate reduction therapy. In this review, we discuss the recent developments in MPS research and provide a framework for developing strategies.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Division of Gene Therapy, Research Center for Medical Sciences, Department of Pediatrics, The Jikei University School of Medicine, Tokyo, 105-8461, Japan.
| |
Collapse
|
21
|
Abstract
Leukodystrophies are a heterogeneous class of genetic diseases affecting the white matter in the central nervous system with a broad range of clinical manifestations and a frequently progressive course. An interest in precision medicine has emerged over the last several decades, and biomedical research in leukodystrophies has made exciting advances along this front through therapeutic target discovery and novel disease model systems. In this review, we discuss current and emerging therapeutic approaches in leukodystrophies, including gene therapy, antisense oligonucleotide therapy, CRISPR/CAS-based gene editing, and cell and stem cell based therapies.
Collapse
Affiliation(s)
- Eliza Gordon-Lipkin
- 1 Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA .,2 Departments of Neurology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ali Fatemi
- 1 Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA .,2 Departments of Neurology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,3 Moser Center for Leukodystrophies and Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA
| |
Collapse
|
22
|
Selvanathan A, Ellaway C, Wilson C, Owens P, Shaw PJ, Bhattacharya K. Effectiveness of Early Hematopoietic Stem Cell Transplantation in Preventing Neurocognitive Decline in Mucopolysaccharidosis Type II: A Case Series. JIMD Rep 2018; 41:81-89. [PMID: 29671225 DOI: 10.1007/8904_2018_104] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 03/18/2018] [Accepted: 03/19/2018] [Indexed: 12/02/2022] Open
Abstract
The early progressive form of the X-linked disorder, Hunter syndrome or mucopolysaccharidosis type II (MPS II) (OMIM #309900), is characterized by cognitive decline, and pulmonary and cardiac complications that often cause death before 20 years of age. Deficiency of the lysosomal enzyme, iduronate-2-sulfatase (EC 3.1.6.13) results in deposition of the glycosaminoglycans, dermatan, and heparan sulfate in various tissues. In recent years, enzyme replacement therapy (ERT) has become the mainstay of treatment, but is expensive and ineffective in arresting cognitive decline. Hematopoietic stem cell transplantation (HSCT) also provides enzyme replacement, and may be effective in stabilizing neurocognitive function if initiated early, though data are limited. We present a case series of four patients who demonstrated neurocognitive stabilization with early HSCT. HSCT is a potentially underutilized treatment strategy for select groups of MPS II patients.
Collapse
Affiliation(s)
- A Selvanathan
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Westmead, NSW, Australia.
- Discipline of Child and Adolescent Health, The University of Sydney, Camperdown, NSW, Australia.
| | - C Ellaway
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Camperdown, NSW, Australia
| | - C Wilson
- Starship Paediatric Metabolic Service, Starship Children's Hospital, Auckland, New Zealand
| | - P Owens
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - P J Shaw
- Discipline of Child and Adolescent Health, The University of Sydney, Camperdown, NSW, Australia
- Blood and Marrow Transplant Service, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - K Bhattacharya
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
23
|
Guo N, DeAngelis V, Zhu C, Schuchman EH, Simonaro CM. Pentosan Polysulfate Treatment of Mucopolysaccharidosis Type IIIA Mice. JIMD Rep 2018; 43:37-52. [PMID: 29654542 PMCID: PMC6323024 DOI: 10.1007/8904_2018_96] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/05/2018] [Accepted: 02/08/2018] [Indexed: 02/06/2023] Open
Abstract
Overall Goal: This study was designed to evaluate the impact of pentosan polysulfate (PPS) treatment on mice with mucopolysaccharidosis (MPS) type IIIA (Sanfilippo A syndrome; OMIM 252900). Protocol: Three groups of MPS IIIA mice were evaluated: 1-week-old mice treated with subcutaneous (subQ) PPS at 25 mg/kg once weekly for 31 weeks (group 1); 5-month-old mice treated with subQ PPS once weekly at 50 mg/kg for 12 weeks (group 2); and 5-week-old mice treated by continual intracerebroventricular (ICV) PPS infusion for 11 weeks (60 μg/kg/day). Treated MPS IIIA mice and controls were assessed by measuring plasma cytokine levels, histologic analyses of systemic organs, and analyses of various neuroinflammatory, neurodegenerative, and lysosomal disease markers in their brains. Neurobehavioral testing also was carried out. Results: As seen in other MPS animal models, subQ PPS treatment reduced plasma cytokine levels and macrophage infiltration in systemic tissues. ICV administration did not elicit these systemic effects. SubQ PPS administration also significantly impacted brain neuropathology, inflammation, and behavior. The effect of early subQ treatment was more significant than dose. Surprisingly, ICV PPS treatment had intermediate effects on most of these brain markers, perhaps due to the limited dose and/or duration of treatment. Consistent with these neuropathological findings, we also observed significant improvements in the hyperactivity/anxiety and learning behaviors of the MPS IIIA mice treated with early subQ PPS.
Collapse
Affiliation(s)
- Ningning Guo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Victor DeAngelis
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Changzhi Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edward H Schuchman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Calogera M Simonaro
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
24
|
Beckmann NB, Miller WP, Dietrich MS, Orchard PJ. Quality of life among boys with adrenoleukodystrophy following hematopoietic stem cell transplant. Child Neuropsychol 2017; 24:986-998. [DOI: 10.1080/09297049.2017.1380176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Nicole B. Beckmann
- University of Minnesota Masonic Children’s Hospital, Pediatric Blood & Marrow Transplantation, Minneapolis, MN, USA
- Vanderbilt University Schools of Medicine, Nashville, TN, USA
| | - Weston P. Miller
- University of Minnesota Medical School, Department of Pediatrics, Division of Blood & Marrow Transplantation, Minneapolis, MN, USA
| | - Mary S. Dietrich
- Vanderbilt University Schools of Medicine and Nursing, Nashville, TN, USA
| | - Paul J. Orchard
- University of Minnesota Medical School, Department of Pediatrics, Division of Blood & Marrow Transplantation, Minneapolis, MN, USA
| |
Collapse
|
25
|
Saute JAM, Souza CFMD, Poswar FDO, Donis KC, Campos LG, Deyl AVS, Burin MG, Vargas CR, Matte UDS, Giugliani R, Saraiva-Pereira ML, Vedolin LM, Gregianin LJ, Jardim LB. Neurological outcomes after hematopoietic stem cell transplantation for cerebral X-linked adrenoleukodystrophy, late onset metachromatic leukodystrophy and Hurler syndrome. ARQUIVOS DE NEURO-PSIQUIATRIA 2017; 74:953-966. [PMID: 27991992 DOI: 10.1590/0004-282x20160155] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/24/2016] [Indexed: 01/12/2023]
Abstract
Objective To describe survival and neurological outcomes after HSCT for these disorders. Methods Seven CALD, 2 MLD and 2 MPS-IH patients underwent HSCT between 2007 and 2014. Neurological examinations, magnetic resonance imaging, molecular and biochemical studies were obtained at baseline and repeated when appropriated. Results Favorable outcomes were obtained with 4/5 related and 3/6 unrelated donors. Two patients died from procedure-related complications. Nine transplanted patients were alive after a median of 3.7 years: neurological stabilization was obtained in 5/6 CALD, 1/2 MLD, and one MPS-IH patient. Brain lesions of the MPS-IH patient were reduced four years after HSCT. Conclusion Good outcomes were obtained when HSCT was performed before adulthood, early in the clinical course, and/or from a related donor.
Collapse
Affiliation(s)
- Jonas Alex Morales Saute
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre RS, Brasil.,Hospital de Clínicas de Porto Alegre, Laboratório de Identificação Genética, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre RS, Brasil
| | | | - Fabiano de Oliveira Poswar
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Genética e Biologia Molecular; Porto Alegre RS, Brasil
| | - Karina Carvalho Donis
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Porto Alegre RS, Brasil
| | - Lillian Gonçalves Campos
- Hospital de Clínicas de Porto Alegre, Serviço de Radiologia, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre RS, Brasil
| | | | - Maira Graeff Burin
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre RS, Brasil
| | - Carmen Regla Vargas
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Faculdade de Farmacia, Porto Alegre, Brasil
| | - Ursula da Silveira Matte
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Genética e Biologia Molecular; Porto Alegre RS, Brasil.,Hospital de Clínicas de Porto Alegre, Laboratório de Terapia Gênica, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Departamento de Genética e Biologia Molecular, Porto Alegre RS, Brasil
| | - Roberto Giugliani
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Genética e Biologia Molecular; Porto Alegre RS, Brasil.,Hospital de Clínicas de Porto Alegre, Laboratório de Terapia Gênica, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Departamento de Genética e Biologia Molecular, Porto Alegre RS, Brasil
| | - Maria Luiza Saraiva-Pereira
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre RS, Brasil.,Hospital de Clínicas de Porto Alegre, Laboratório de Identificação Genética, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Genética e Biologia Molecular; Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Departamento de Bioquímica, Porto Alegre RS, Brasil
| | - Leonardo Modesti Vedolin
- Hospital de Clínicas de Porto Alegre, Serviço de Radiologia, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre RS, Brasil
| | - Lauro José Gregianin
- Hospital de Clínicas de Porto Alegre, Serviço de Oncologia Pediátrica, Porto Alegre, Brasil.,Universidade Federal do Rio Grande do Sul, Departamento de Pediatria, Porto Alegre RS, Brasil
| | - Laura Bannach Jardim
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre RS, Brasil.,Hospital de Clínicas de Porto Alegre, Laboratório de Identificação Genética, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Genética e Biologia Molecular; Porto Alegre RS, Brasil.,Universidade Federal do Rio Grande do Sul, Departamento de Medicina Interna, Porto Alegre RS, Brasil
| |
Collapse
|
26
|
Bradley LA, Haddow HRM, Palomaki GE. Treatment of mucopolysaccharidosis type II (Hunter syndrome): results from a systematic evidence review. Genet Med 2017. [PMID: 28640238 DOI: 10.1038/gim.2017.30] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PurposeA pilot systematic evidence review to establish methodology utility in rare genetic diseases, support clinical recommendations, and identify important knowledge gaps.MethodsBroad-based published/gray-literature searches through December 2015 for studies of males with confirmed mucopolysaccharidosis type II (any age, phenotype, genotype, family history) treated with enzyme replacement therapy or hematopoietic stem cell transplantation. Preset inclusion criteria employed for abstract and full document selection, and standardized methods for data extraction and assessment of quality and strength of evidence.ResultsTwelve outcomes reported included benefits of urinary glycosaminoglycan and liver/spleen volume reductions and harms of immunoglobulin G/neutralizing antibody development (moderate strength of evidence). Less clear were benefits of improved 6-minute walk tests, height, early treatment, and harms of other adverse reactions (low strength of evidence). Benefits and harms of other outcomes were unclear (insufficient strength of evidence). Current benefits and harms of hematopoietic stem cell transplantation are unclear, based on dated, low-quality studies. A critical knowledge gap is long-term outcomes. Consensus on selection of critical outcomes and measures is needed to definitively evaluate treatment safety and effectiveness.ConclusionMinor methodology modifications and a focus on critical evidence can reduce review time and resources. Summarized evidence was sufficient to support guidance development and highlight important knowledge gaps.
Collapse
Affiliation(s)
- Linda A Bradley
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital/Warren Alpert, Medical School at Brown University, Providence, Rhode Island, USA
| | | | - Glenn E Palomaki
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital/Warren Alpert, Medical School at Brown University, Providence, Rhode Island, USA
| |
Collapse
|
27
|
Lizzi Lagranha V, Zambiasi Martinelli B, Baldo G, Ávila Testa G, Giacomet de Carvalho T, Giugliani R, Matte U. Subcutaneous implantation of microencapsulated cells overexpressing α-L-iduronidase for mucopolysaccharidosis type I treatment. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:43. [PMID: 28150116 DOI: 10.1007/s10856-017-5844-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/03/2017] [Indexed: 06/06/2023]
Abstract
Mucopolysaccharidosis type I (MPS I) is caused by a deficiency of α-L-iduronidase (IDUA), resulting in accumulation of glycosaminoglycans (GAG) in lysosomes. Microencapsulation of recombinant cells is a promising gene/cell therapy approach that could overcome the limitations of the current available treatments. In the present study we produced alginate-poly-L-lysine-alginate (APA) microcapsules containing recombinant cells overexpressing IDUA, which were implanted in the subcutaneous space of MPS I mice in order to evaluate their potential effect as a treatment for this disease. APA microcapsules enclosing genetically modified Baby Hamster Kidney cells overexpressing IDUA were produced and implanted in the subcutaneous space of 4-month-old MPS I mice (Idua -/-). Treatment was performed using two cell concentrations: 8.3 × 107 and 8.3 × 106 cells/mL. Untreated MPS I and normal mice were used as controls. Microcapsules were retrieved and analyzed after treatment. Increased IDUA in the liver, kidney and heart was detected 24 h postimplantation. After 120 days, higher IDUA activity was detected in the liver, kidney and heart, in both groups, whereas GAG accumulation was reduced only in the high cell concentration group. Microcapsules analysis showed blood vessels around them, as well as inflammatory cells and a fibrotic layer. Microencapsulated cells were able to ameliorate some aspects of the disease, indicating their potential as a treatment. To achieve better performance of the microcapsules, improvements such as the modulation of inflammatory response are suggested.
Collapse
Affiliation(s)
- Valeska Lizzi Lagranha
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Barbara Zambiasi Martinelli
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Baldo
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program in Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Talita Giacomet de Carvalho
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Roberto Giugliani
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ursula Matte
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
28
|
Grosse SD, Lam WKK, Wiggins LD, Kemper AR. Cognitive outcomes and age of detection of severe mucopolysaccharidosis type 1. Genet Med 2017; 19:975-982. [PMID: 28125077 DOI: 10.1038/gim.2016.223] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/12/2016] [Indexed: 11/09/2022] Open
Abstract
The US Secretary of Health and Human Services recommended in February 2016 that mucopolysaccharidosis type 1 (MPS I) be added to the recommended uniform screening panel for state newborn screening programs. One of the key factors in this decision was the evidence suggesting that earlier treatment with hematopoietic cell transplantation (HCT) for the most severe form, Hurler syndrome (MPS IH), would lead to improved cognitive outcomes. Consistent evidence from peer-reviewed studies suggests that transplantation in the first year of life is associated with improved developmental quotient or intelligence quotient and continued cognitive growth, with earlier age of treatment associated with improved outcomes. However, available evidence suggests that cognitive functioning and attention can still lag behind unaffected age-matched children, leading to the need for special education services. Verbal and nonverbal cognitive abilities outcomes may be affected differently by HCT. With the recent addition of MPS I to the recommended uniform screening panel, future work is needed to evaluate the impact of earlier, presymptomatic detection and treatment initiation and other supportive therapies on cognitive outcomes.Genet Med advance online publication 26 January 2017.
Collapse
Affiliation(s)
- Scott D Grosse
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Wendy K K Lam
- Duke Clinical Translational Science Institute, Duke University, Durham, North Carolina, USA
| | - Lisa D Wiggins
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Alex R Kemper
- Duke Clinical Translational Science Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
29
|
Winner LK, Beard H, Hassiotis S, Lau AA, Luck AJ, Hopwood JJ, Hemsley KM. A Preclinical Study Evaluating AAVrh10-Based Gene Therapy for Sanfilippo Syndrome. Hum Gene Ther 2016; 27:363-75. [PMID: 26975339 DOI: 10.1089/hum.2015.170] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA) is predominantly a disorder of the central nervous system, caused by a deficiency of sulfamidase (SGSH) with subsequent storage of heparan sulfate-derived oligosaccharides. No widely available therapy exists, and for this reason, a mouse model has been utilized to carry out a preclinical assessment of the benefit of intraparenchymal administration of a gene vector (AAVrh10-SGSH-IRES-SUMF1) into presymptomatic MPS IIIA mice. The outcome has been assessed with time, measuring primary and secondary storage material, neuroinflammation, and intracellular inclusions, all of which appear as the disease progresses. The vector resulted in predominantly ipsilateral distribution of SGSH, with substantially less detected in the contralateral hemisphere. Vector-derived SGSH enzyme improved heparan sulfate catabolism, reduced microglial activation, and, after a time delay, ameliorated GM3 ganglioside accumulation and halted ubiquitin-positive lesion formation in regions local to, or connected by projections to, the injection site. Improvements were not observed in regions of the brain distant from, or lacking connections with, the injection site. Intraparenchymal gene vector administration therefore has therapeutic potential provided that multiple brain regions are targeted with vector, in order to achieve widespread enzyme distribution and correction of disease pathology.
Collapse
Affiliation(s)
- Leanne K Winner
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI) , Adelaide, Australia
| | - Helen Beard
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI) , Adelaide, Australia
| | - Sofia Hassiotis
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI) , Adelaide, Australia
| | - Adeline A Lau
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI) , Adelaide, Australia
| | - Amanda J Luck
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI) , Adelaide, Australia
| | - John J Hopwood
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI) , Adelaide, Australia
| | - Kim M Hemsley
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI) , Adelaide, Australia
| |
Collapse
|
30
|
Mallhi KK, Smith AR, DeFor TE, Lund TC, Orchard PJ, Miller WP. Allele-Level HLA Matching Impacts Key Outcomes Following Umbilical Cord Blood Transplantation for Inherited Metabolic Disorders. Biol Blood Marrow Transplant 2016; 23:119-125. [PMID: 27989932 DOI: 10.1016/j.bbmt.2016.10.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/25/2016] [Indexed: 01/06/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation has demonstrated efficacy for numerous inherited metabolic disorders (IMDs). Umbilical cord blood transplant (UCBT) is increasingly used as a graft source in IMDs, but little is known of the impact of cord blood unit (CBU)/recipient HLA allelic disparity on key outcomes following UCBT for IMD. We reviewed outcomes of 106 consecutive first, single UCBTs for IMD at the University of Minnesota with regard to CBU/recipient HLA allelic matching (HLA-A, -B, -C, and -DRB1). The median age at UCBT was 1 year, and 87 patients (82%) received myeloablative conditioning. Primary diagnoses were Hurler syndrome (41%), cerebral adrenoleukodystrophy (35%), metachromatic leukodystrophy/globoid cell leukodystrophy (9%), and other (16%). The 5-year overall survival (OS) for the entire cohort was 70% (95% confidence interval, 59% to 79%). Rates of severe acute and chronic graft-versus-host disease were low (6% for each). CBU/recipient HLA conventional matching was based on antigen-level matching at HLA-A and -B, and on allele-level matching at HLA-DRB1. Of 46 conventional matched UCBTs, 20 (43%) were mismatched at 1 or more alleles. Of 49 conventional 5/6 UCBTs, 30 (61%) were mismatched at ≥2 alleles and 19 (39%) were mismatched at ≥3 alleles. Within the 6/6 conventional match stratum, comparisons of key outcomes between allele-matched and allele-mismatched UCBT were as follows: 5-year OS, 88% versus 42% (P < .01); 1-year engrafted survival (ES) with ≥90% donor chimerism, 73% versus 60% (P = .33); graft failure, 8% versus 30% (P = .05); and transplantation-related mortality (TRM), 8% versus 30% (P = .04). For patients undergoing conventional 5/6 HLA-matched UCBT, better allelic matching was associated with similar outcomes: 5-year OS, 77% versus 74% (P = .72); 1-year ES, 73% versus 47% (P = .06); graft failure, 17% versus 42% (P = .05); and TRM, 10% versus 16% (P = .54). On multivariable analyses, fewer allele-level mismatches within each conventional match stratum continued to predict more favorable outcomes following UCBT. These data provide evidence that allele-level HLA matching considerations within a conventional HLA match stratum may better predict outcomes of interest after UCBT for IMD. Larger studies are warranted to confirm these findings and explore other allele-level HLA match dynamics.
Collapse
Affiliation(s)
- Kanwaldeep K Mallhi
- Division of Pediatric Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Angela R Smith
- Division of Pediatric Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Todd E DeFor
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Troy C Lund
- 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
| | - Weston P Miller
- Division of Pediatric Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
31
|
Svenberg P, Remberger M, Uzunel M, Mattsson J, Gustafsson B, Fjaertoft G, Sundin M, Winiarski J, Ringdén O. Improved overall survival for pediatric patients undergoing allogeneic hematopoietic stem cell transplantation - A comparison of the last two decades. Pediatr Transplant 2016; 20:667-74. [PMID: 27251184 DOI: 10.1111/petr.12723] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/26/2016] [Indexed: 01/06/2023]
Abstract
Pediatric protocols for allogeneic hematopoietic SCT have been altered during the last two decades. To compare the outcomes in children (<18 yr old), who underwent SCT at our center during 1992-2002 (P1) and 2003-2013 (P2). We retrospectively analyzed 188 patients in P1 and 201 patients in P2. The most significant protocol changes during P2 compared with P1 were a decrease in MAC protocols, particularly those containing TBI, an increase in RIC protocols, and altered GvHD prophylaxis. In addition, P2 had more patients with nonmalignant diagnoses (p = 0.002), more mismatched (MM) donors (p = 0.01), and more umbilical CB grafts (p = 0.03). Mesenchymal or DSCs were used for severe acute GvHD during P2. Three-yr OS in P1 was 58%, and in P2, it was 78% (p < 0.001). Improved OS was seen in both malignant disorders (51% vs. 68%; p = 0.05) and nonmalignant disorders (77% vs. 87%; p = 0.04). Multivariate analysis showed that SCT during P2 was associated with reduced mortality (HR = 0.57; p = 0.005), reduced TRM (HR = 0.57; p = 0.03), unchanged relapse rate, similar rate of GF, less chronic GvHD (HR = 0.49; p = 0.01), and more acute GvHD (HR = 1.77, p = 0.007). During recent years, OS has improved at our center, possibly reflecting the introduction of less toxic conditioning regimens and a number of other methodological developments in SCT.
Collapse
Affiliation(s)
- Petter Svenberg
- Oncology/Coagulation Section, Karolinska University Hospital, Stockholm, Sweden
| | - Mats Remberger
- Department of Oncology/Pathology, Karolinska Institute, Stockholm, Sweden.,Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Mehmet Uzunel
- Division of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Jonas Mattsson
- Department of Oncology/Pathology, Karolinska Institute, Stockholm, Sweden.,Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Britt Gustafsson
- Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden.,Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Gustav Fjaertoft
- Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Sundin
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden.,Hematology/Immunology Section, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Jacek Winiarski
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden.,Hematology/Immunology Section, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Olle Ringdén
- Division of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
32
|
Long-Term Follow-Up after Reduced-Intensity Conditioning and Stem Cell Transplantation for Childhood Nonmalignant Disorders. Biol Blood Marrow Transplant 2016; 22:1467-1472. [DOI: 10.1016/j.bbmt.2016.04.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 04/30/2016] [Indexed: 12/21/2022]
|
33
|
Lum S, Jones S, Ghosh A, Bigger B, Wynn R. Hematopoietic stem cell transplant for the mucopolysaccharidoses. Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1147948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
34
|
Affiliation(s)
- Gregory M T Guilcher
- Section of Pediatric Oncology/BMT, Alberta Children's Hospital; Departments of Oncology and Pediatrics, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
35
|
Kato S, Yabe H, Takakura H, Mugishima H, Ishige M, Tanaka A, Kato K, Yoshida N, Adachi S, Sakai N, Hashii Y, Ohashi T, Sasahara Y, Suzuki Y, Tabuchi K. Hematopoietic stem cell transplantation for inborn errors of metabolism: A report from the Research Committee on Transplantation for Inborn Errors of Metabolism of the Japanese Ministry of Health, Labour and Welfare and the Working Group of the Japan Society for Hematopoietic Cell Transplantation. Pediatr Transplant 2016; 20:203-14. [PMID: 26806759 DOI: 10.1111/petr.12672] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2015] [Indexed: 11/29/2022]
Abstract
A total of 216 patients with IEM were treated by allogeneic HSCT in Japan from 1985 until 2010. The results of UCBT have improved, and the OS rate of UCBT (81.9%) was not different from those of RBMT (87.2%) or UBMT (73.9%) in 2000-2010. However, EFS rates in RBMT (73.2%) and UBMT (62.2%) were better than that in UCBT (49.5%), and the difference between RBMT and UCBT was significant (p = 0.01). The EFS rate of patients conditioned by RIC (74.6%) was comparable or slightly better than in those who underwent MAC with irradiation (57.9%) or without irradiation (54.2%) in 2000-2010. A more pronounced trend was observed toward differential EFS for UCBT in 2000-2010: RIC (62.9%), MAC with irradiation (20.0%), and MAC without irradiation (42.1%). The difference between RIC and MAC with irradiation was significant (p < 0.03). In summary, we report a Japanese registry analysis of HSCT for IEM with improving survival in UCBT. The introduction of RIC after 2000 was considered to contribute to this improvement. UCBT could be recommended for those who lack an HLA-identical sibling donor.
Collapse
Affiliation(s)
- Shunichi Kato
- Departments of Cell Transplantation and Pediatrics, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiromasa Yabe
- Departments of Cell Transplantation and Pediatrics, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiromitsu Takakura
- Departments of Cell Transplantation and Pediatrics, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hideo Mugishima
- Department of Pediatrics, Nihon University School of Medicine, Tokyo, Japan
| | - Mika Ishige
- Department of Pediatrics, Nihon University School of Medicine, Tokyo, Japan
| | - Akemi Tanaka
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koji Kato
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Aichi, Japan
| | - Nao Yoshida
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Aichi, Japan
| | - Souichi Adachi
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norio Sakai
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Suita, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Suita, Japan
| | - Toya Ohashi
- Department of Gene Therapy, Institute of DNA Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoji Sasahara
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University School of Medicine, Gifu, Japan
| | - Ken Tabuchi
- Department of Pediatrics and Data Center, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| |
Collapse
|
36
|
Biffi A. Gene therapy for lysosomal storage disorders: a good start. Hum Mol Genet 2015; 25:R65-75. [PMID: 26604151 DOI: 10.1093/hmg/ddv457] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022] Open
Abstract
Lysosomal storage disorders (LSDs) are a heterogeneous group of inherited diseases with a collective frequency of ∼1 in 7000 births, resulting from the deficiency in one or more enzymes or transporters that normally reside within the lysosomes. Pathology results from the progressive accumulation of uncleaved lipids, glycoproteins and/or glycosaminoglycans in the lysosomes and secondary damages that affect the brain, viscera, bones and connective tissues. Most treatment modalities developed for LSD, including gene therapy (GT), are based on the lysosome-specific cross-correction mechanism, by which close proximity of normal cells leads to the correction of the biochemical consequences of enzymatic deficiency within the neighboring cells. Here, GT efforts addressing these disorders are reviewed with an up-to-date discussion of their impact on the LSD disease phenotype in animal models and patients.
Collapse
Affiliation(s)
- Alessandra Biffi
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
37
|
Tylki-Szymańska A, Jurecka A. Prospective therapies for mucopolysaccharidoses. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1089167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
38
|
Abstract
Lysosomal storage diseases are a group of rare, inborn, metabolic errors characterized by deficiencies in normal lysosomal function and by intralysosomal accumulation of undegraded substrates. The past 25 years have been characterized by remarkable progress in the treatment of these diseases and by the development of multiple therapeutic approaches. These approaches include strategies aimed at increasing the residual activity of a missing enzyme (enzyme replacement therapy, hematopoietic stem cell transplantation, pharmacological chaperone therapy and gene therapy) and approaches based on reducing the flux of substrates to lysosomes. As knowledge has improved about the pathophysiology of lysosomal storage diseases, novel targets for therapy have been identified, and innovative treatment approaches are being developed.
Collapse
|
39
|
Shapiro EG, Nestrasil I, Rudser K, Delaney K, Kovac V, Ahmed A, Yund B, Orchard PJ, Eisengart J, Niklason GR, Raiman J, Mamak E, Cowan MJ, Bailey-Olson M, Harmatz P, Shankar SP, Cagle S, Ali N, Steiner RD, Wozniak J, Lim KO, Whitley CB. Neurocognition across the spectrum of mucopolysaccharidosis type I: Age, severity, and treatment. Mol Genet Metab 2015; 116:61-8. [PMID: 26095521 PMCID: PMC4561597 DOI: 10.1016/j.ymgme.2015.06.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/13/2015] [Accepted: 06/13/2015] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Precise characterization of cognitive outcomes and factors that contribute to cognitive variability will enable better understanding of disease progression and treatment effects in mucopolysaccharidosis type I (MPS I). We examined the effects on cognition of phenotype, genotype, age at evaluation and first treatment, and somatic disease burden. METHODS Sixty patients with severe MPS IH (Hurler syndrome treated with hematopoietic cell transplant and 29 with attenuated MPS I treated with enzyme replacement therapy), were studied with IQ measures, medical history, genotypes. Sixty-seven patients had volumetric MRI. Subjects were grouped by age and phenotype and MRI and compared to 96 normal controls. RESULTS Prior to hematopoietic cell transplant, MPS IH patients were all cognitively average, but post-transplant, 59% were below average, but stable. Genotype and age at HCT were associated with cognitive ability. In attenuated MPS I, 40% were below average with genotype and somatic disease burden predicting their cognitive ability. White matter volumes were associated with IQ for controls, but not for MPS I. Gray matter volumes were positively associated with IQ in controls and attenuated MPS I patients, but negatively associated in MPS IH. CONCLUSIONS Cognitive impairment, a major difficulty for many MPS I patients, is associated with genotype, age at treatment and somatic disease burden. IQ association with white matter differed from controls. Many attenuated MPS patients have significant physical and/or cognitive problems and receive insufficient support services. Results provide direction for future clinical trials and better disease management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Julian Raiman
- Hospital for Sick Children, University of Toronto, Toronto, CA, United States
| | - Eva Mamak
- Hospital for Sick Children, Toronto, CA, United States
| | - Morton J Cowan
- UCSF Benioff Children's Hospital, University of California San Francisco, United States
| | - Mara Bailey-Olson
- UCSF Benioff Children's Hospital, University of California San Francisco, United States
| | - Paul Harmatz
- UCSF Benioff Children's Hospital Oakland, United States
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Newborn Screening for Lysosomal Storage Disorders: Views of Genetic Healthcare Providers. J Genet Couns 2015; 25:373-84. [DOI: 10.1007/s10897-015-9879-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 08/12/2015] [Indexed: 01/01/2023]
|
41
|
Du H, Chen J, Qin M, Fang J, Li Z, Zhu Y, Sun X, Huang D, Yu J, Tang Y, Hu S, Li J, Zhang Z, Luan Z. Pediatric hematopoietic stem cell transplantation in China: Data and trends during 1998-2012. Pediatr Transplant 2015; 19:563-70. [PMID: 26058853 DOI: 10.1111/petr.12525] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2015] [Indexed: 11/26/2022]
Abstract
The success of treating a wide variety of pediatric diseases with HSCT, hematologic malignancies in particular, has resulted in an increased number of long-term survivors. This study is the first large-scale, multicentre report that describes the evolution of pediatric HSCTs in China during the period of 1998-2012. Of all 1052 patients, 266 cases were treated with autologous HSCs and 786 used allogeneic HSCs. The disease indications for HSCTs mainly included leukemias, lymphoma, solid tumors, and non-malignant disorders. The total number of HSCTs, especially unrelated donor transplants, appeared to be increasing year by year. For patients with neuroblastoma, the therapeutic efficacy seemed to be poor, with a five-yr OS and DFS rate of 34.5 ± 14.3% and 20.7 ± 9.6%, respectively. In contrast, the survival of patients with SAA was prominently improved, and their five-yr OS and DFS rates were 82.8 ± 4% and 80.7 ± 4.1%, respectively. Patients who received cord blood transplants had a lower incidence of acute GVHD than that of PB and/or BM transplants from unrelated donors. This report offers us a valuable resource for evaluating the changes in HSCTs in China over the past 14 yr.
Collapse
Affiliation(s)
- Hong Du
- Navy General Hospital, Beijing, China
| | - Jing Chen
- Shanghai Children's Medical Center, Shanghai, China
| | | | - Jianpei Fang
- Second Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | | | - Yiping Zhu
- West China Second University Hospital of Sichuan University, Chengdu, China
| | - Xin Sun
- Guangzhou Women and Children Medical Center, Guangzhou, China
| | | | - Jie Yu
- Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yongmin Tang
- Zhe Jiang University School of Medicine Children Hospital, Hangzhou, China
| | - Shaoyan Hu
- Soochow University Affiliated Children's Hospital, Suzhou, China
| | - Junhui Li
- Capital Institute of Pediatics, Beijing, China
| | | | - Zuo Luan
- Navy General Hospital, Beijing, China
| |
Collapse
|
42
|
Kantor B, McCown T, Leone P, Gray SJ. Clinical applications involving CNS gene transfer. ADVANCES IN GENETICS 2015; 87:71-124. [PMID: 25311921 DOI: 10.1016/b978-0-12-800149-3.00002-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diseases of the central nervous system (CNS) have traditionally been the most difficult to treat by traditional pharmacological methods, due mostly to the blood-brain barrier and the difficulties associated with repeated drug administration targeting the CNS. Viral vector gene transfer represents a way to permanently provide a therapeutic protein within the nervous system after a single administration, whether this be a gene replacement strategy for an inherited disorder or a disease-modifying protein for a disease such as Parkinson's. Gene therapy approaches for CNS disorders has evolved considerably over the last two decades. Although a breakthrough treatment has remained elusive, current strategies are now considerably safer and potentially much more effective. This chapter will explore the past, current, and future status of CNS gene therapy, focusing on clinical trials utilizing adeno-associated virus and lentiviral vectors.
Collapse
Affiliation(s)
- Boris Kantor
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina, Columbia, SC, USA
| | - Thomas McCown
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paola Leone
- Department of Cell Biology, Rowan University, Camden, NJ, USA
| | - Steven J Gray
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Ophthalmology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
43
|
Andrade F, Aldámiz-Echevarría L, Llarena M, Couce ML. Sanfilippo syndrome: Overall review. Pediatr Int 2015; 57:331-8. [PMID: 25851924 DOI: 10.1111/ped.12636] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/03/2014] [Accepted: 02/13/2015] [Indexed: 12/17/2022]
Abstract
Mucopolysaccharidosis type III (MPS III, Sanfilippo syndrome) is a lysosomal storage disorder, caused by a deficiency in one of the four enzymes involved in the catabolism of glycosaminoglycan heparan sulfate. It is characterized by progressive cognitive decline and severe hyperactivity, with relatively mild somatic features. This review focuses on clinical features, diagnosis, treatment, and follow-up of MPS III, and provides information about supplementary tests and differential diagnosis. Given that few reviews of MPS III have been published, several studies were compiled to establish diagnostic recommendations. Quantitative urinary glycosaminoglycan analysis is strongly recommended, and measurement of disaccharides, heparin cofactor II-thrombin complex and gangliosides is also used. Enzyme activity of the different enzymes in blood serum, leukocytes or fibroblasts, and mutational analysis for SGSH, NAGLU, HGSNAT or GNS genes are required to confirm diagnosis and differentiate four subtypes of MPS III. Although there is no global consensus for treatment, enzyme replacement therapy and gene therapy can provide appropriate results. In this regard, recent publications on treatment and follow-up are discussed.
Collapse
Affiliation(s)
- Fernando Andrade
- Division of Metabolism, BioCruces Health Research Institute, CIBER de Enfermedades Raras (CIBERER), Barakaldo, Spain
| | - Luis Aldámiz-Echevarría
- Division of Metabolism, BioCruces Health Research Institute, CIBER de Enfermedades Raras (CIBERER), Barakaldo, Spain
| | - Marta Llarena
- Division of Metabolism, BioCruces Health Research Institute, CIBER de Enfermedades Raras (CIBERER), Barakaldo, Spain
| | - María Luz Couce
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases, Neonatology Service, Department of Pediatrics, CIBER de Enfermedades Raras (CIBERER), IDIS Clinic University Hospital of Santiago de Compostela, Coruña, Spain
| |
Collapse
|
44
|
Mahmoud HK, Elhaddad AM, Fahmy OA, Samra MA, Abdelfattah RM, El-Nahass YH, Fathy GM, Abdelhady MS. Allogeneic hematopoietic stem cell transplantation for non-malignant hematological disorders. J Adv Res 2015; 6:449-58. [PMID: 26257943 PMCID: PMC4522586 DOI: 10.1016/j.jare.2014.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/29/2014] [Accepted: 11/01/2014] [Indexed: 11/17/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) from a geno-identical matched sibling (MSD) is one of the most successful therapies in patients with non-malignant hematological disorders. This study included 273 patients with severe aplastic anemia (SAA), 152 patients with B-Thalassemia major (BTM), 31 patients with Fanconi's anemia (FA), 20 patients with congenital immunodeficiency diseases (ID), and 13 patients with inherited metabolic disorders (IMD) allografted from a MSD. In SAA, the 8-year overall survival (OS) of the whole group patients was 74%. OS was significantly better in patients conditioned with fludarabine and cyclophosphamide (Flu/Cy) than in those who received cyclophosphamide and antithymocyte globulin (Cy/ATG) (p = 0.021). Acute graft-versus-host disease (aGVHD) grade II-IV occurred in 15% while chronic GVHD (cGVHD) occurred in 28%. In BTM, the 12-year disease-free survival (DFS) of the whole group of BTM patients was 72.4%. DFS was 74% for peripheral blood stem cell (PBSC) group compared to 64% in the BM stem cell group. The incidence of graft rejection was significantly lower in patients who received PBSC than in those who received BM (9% vs 25%) (p = 0.036). AGVHD grade II-IV and cGVHD occurred in 15% and 12% of the whole group of BTM patients respectively. In FA, the 5-year OS was 64.5%. Graft rejection occurred in 10% of patients. Grade II-IV aGVHD occurred in 16% while cGVHD occurred in 4%. In ID, the 5-year OS was 62%. Graft rejection occurred in two (10%) patients. Three patients (15%) developed grade II-IV aGVHD, 2 of them progressed to secondary cGVHD. In IMD, OS was 46% at 5 years. Graft rejection occurred in 8% of patients. AGVHD grade II-IV occurred in 15% while cGVHD occurred in 14%. In conclusion, Allo-HSCT provides a higher DFS rate over conventional therapies for patients with non-malignant hematological disorders with prolonged survival.
Collapse
Affiliation(s)
- Hossam K. Mahmoud
- Department of Medical Oncology, National Cancer Institute, Cairo University, Egypt
| | - Alaa M. Elhaddad
- Department of Pediatric Oncology, National Cancer Institute, Cairo University, Egypt
| | - Omar A. Fahmy
- Department of Internal Medicine, Faculty of Medicine, Cairo University, Egypt
| | - Mohamed A. Samra
- Department of Medical Oncology, National Cancer Institute, Cairo University, Egypt
| | | | - Yasser H. El-Nahass
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Egypt
| | - Gamal M. Fathy
- Department of Hematology and BMT, Nasser Institute for Research and Treatment, Ministry of Health, Egypt
| | - Mohamed S. Abdelhady
- Department of Hematology and BMT, Nasser Institute for Research and Treatment, Ministry of Health, Egypt
| |
Collapse
|
45
|
|
46
|
Parenti G, Andria G, Valenzano KJ. Pharmacological Chaperone Therapy: Preclinical Development, Clinical Translation, and Prospects for the Treatment of Lysosomal Storage Disorders. Mol Ther 2015; 23:1138-1148. [PMID: 25881001 DOI: 10.1038/mt.2015.62] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 04/01/2015] [Indexed: 02/06/2023] Open
Abstract
Lysosomal storage disorders (LSDs) are a group of inborn metabolic diseases caused by mutations in genes that encode proteins involved in different lysosomal functions, in most instances acidic hydrolases. Different therapeutic approaches have been developed to treat these disorders. Pharmacological chaperone therapy (PCT) is an emerging approach based on small-molecule ligands that selectively bind and stabilize mutant enzymes, increase their cellular levels, and improve lysosomal trafficking and activity. Compared to other approaches, PCT shows advantages, particularly in terms of oral administration, broad biodistribution, and positive impact on patients' quality of life. After preclinical in vitro and in vivo studies, PCT is now being translated in the first clinical trials, either as monotherapy or in combination with enzyme replacement therapy, for some of the most prevalent LSDs. For some LSDs, the results of the first clinical trials are encouraging and warrant further development. Future research in the field of PCT will be directed toward the identification of novel chaperones, including new allosteric drugs, and the exploitation of synergies between chaperone treatment and other therapeutic approaches.
Collapse
Affiliation(s)
- Giancarlo Parenti
- Department of Translational Medical Sciences, Section of Pediatrics, Federico II University, Naples, Italy; Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.
| | - Generoso Andria
- Department of Translational Medical Sciences, Section of Pediatrics, Federico II University, Naples, Italy
| | | |
Collapse
|
47
|
Beck M. Enzyme replacement and gene therapy for mucopolysaccharidoses: current progress and future directions. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1021777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
48
|
Abstract
Pharmacological chaperone therapy is an emerging approach to treat lysosomal storage diseases. Small-molecule chaperones interact with mutant enzymes, favor their correct conformation and enhance their stability. This approach shows significant advantages when compared with existing therapies, particularly in terms of the bioavailability of drugs, oral administration and positive impact on the quality of patients' lives. On the other hand, future research in this field must confront important challenges. The identification of novel chaperones is indispensable to expanding the number of patients amenable to this treatment and to optimize therapeutic efficacy. It is important to develop new allosteric drugs, to address the risk of inhibiting target enzymes. Future research must also be directed towards the exploitation of synergies between chaperone treatment and other therapeutic approaches.
Collapse
|
49
|
Milano F, Boelens JJ. Stem cell comparison: what can we learn clinically from unrelated cord blood transplantation as an alternative stem cell source? Cytotherapy 2015; 17:695-701. [PMID: 25795270 DOI: 10.1016/j.jcyt.2015.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 02/24/2015] [Indexed: 02/01/2023]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is an important therapeutic option for a variety of malignant and non-malignant disorders (NMD). The use of umbilical cord blood transplantation (UCBT) has made HCT available to many more patients. The increased level of human leukocyte antigen disparity that can be tolerated makes UCBT a very attractive alternative source of hematopoietic stem cells; however, the increased risk of early death observed after UCBT remains an obstacle. Novel strategies such as ex vivo stem cell expansion are now becoming part of the standard clinical approach, and preliminary results are extremely encouraging with suggestion of reduction of early transplant-related mortality. Although there are no randomized studies that compare the risks and benefits of UCBT relative to those observed with related and unrelated donors both for malignant and NMD, several retrospective studies have compared outcomes between UCBT and other stem cell sources. In this review, we aim to describe and summarize the findings of the principal studies in this field. We hope that what we can learn from these studies and how we can use this information will improve the outcomes of HCT for patients with malignant and NMD.
Collapse
Affiliation(s)
- Filippo Milano
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA.
| | - Jaap Jan Boelens
- University Medical Center Utrecht, Pediatric Blood and Marrow Transplantation Program, Utrecht, The Netherlands; Laboratory Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
50
|
Abstract
Striking therapeutic advances for lysosomal diseases have harnessed the biology of this organelle and illustrate its central rôle in the dynamic economy of the cell. Further Innovation will require improved protein-targetting or realization of therapeutic gene- and cell transfer stratagems. Rescuing function before irreversible injury, mandates a deep knowledge of clinical behaviour as well as molecular pathology – and frequently requires an understanding of neuropathology. Whether addressing primary causes, or rebalancing the effects of disordered cell function, true therapeutic innovation depends on continuing scientific exploration of the lysosome. Genuine partnerships between biotech and the patients affected by this extraordinary family of disorders continue to drive productive pharmaceutical discovery.
Collapse
Affiliation(s)
- Timothy M Cox
- Department of Medicine, University of Cambridge, UK.
| |
Collapse
|