146 results on '"Lund TC"'
Search Results
2. Phototherapy Alters the Plasma Metabolite Profile in Infants Born Preterm with Hyperbilirubinemia.
- Author
-
Satrom KM, Wang J, Lock EF, Snook K, Lund TC, and Rao RB
- Subjects
- Humans, Infant, Newborn, Male, Female, Prospective Studies, Bilirubin blood, Metabolome, Phototherapy methods, Hyperbilirubinemia, Neonatal therapy, Hyperbilirubinemia, Neonatal blood, Infant, Premature blood, Gestational Age
- Abstract
Objective: To investigate the effects of gestational age (GA) and phototherapy on the plasma metabolite profile of preterm infants with neonatal hyperbilirubinemia (NHB)., Study Design: From a cohort of prospectively enrolled infants born preterm (n = 92), plasma samples of very preterm (VPT; GA, 28 + 0 to 31 + 6 weeks, n = 27) and moderate/late preterm (M/LPT; GA, 32 + 0 to 35 + 6 weeks, n = 33) infants requiring phototherapy for NHB were collected prior to the initiation of phototherapy and 24 hours after starting phototherapy. An additional sample was collected 48 hours after starting phototherapy in a randomly selected subset (n = 30; VPT n = 15; M/LPT n = 15). Metabolite profiles were determined using ultraperformance liquid chromatography tandem mass spectroscopy. Two-way ANCOVA was used to identify metabolites that differed between GA groups and timepoints after adjusting for total serum bilirubin levels (false discovery rate q-value < 0.05). Top impacted pathways were identified using pathway over-representation analysis., Results: Phototherapy was initiated at lower total serum bilirubin (mean ± SD mg/dL) levels in VPT compared with M/LPT infants (7.3 ± 1.4 vs 9.9 ± 1.9, P < .01). We identified 664 metabolites that were significant for a phototherapy effect, 191 metabolites significant for GA, and 46 metabolites significant for GA × phototherapy interaction (false discovery rate q-value < 0.05). Longer duration phototherapy had a larger mean effect size (24 hours postphototherapy: d = 0.36; 48 hours postphototherapy: d = 0.43). Top pathways affected by phototherapy included membrane lipid metabolism, one-carbon metabolism, creatine biosynthesis, and oligodendrocyte differentiation., Conclusion: Phototherapy alters the plasma metabolite profile more than GA in preterm infants with NHB, affecting pathways related to lipid and one-carbon metabolism, energy biosynthesis, and oligodendrocyte differentiation., Competing Interests: Declaration of Competing Interest T.L. and R.R. made substantial contributions to the concept and design of the study and critically reviewed the manuscript for important intellectual content. K.S. and T.L. report financial support was provided by University of Minnesota Twin Cities Department of Pediatrics. The other authors declare no conflicts of interest. Funded by the Department of Pediatrics, University of Minnesota for this work through the Cross-Divisional Grant Mechanism., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Lentiviral Gene Therapy for Cerebral Adrenoleukodystrophy.
- Author
-
Eichler F, Duncan CN, Musolino PL, Lund TC, Gupta AO, De Oliveira S, Thrasher AJ, Aubourg P, Kühl JS, Loes DJ, Amartino H, Smith N, Folloni Fernandes J, Sevin C, Sankar R, Hussain SA, Gissen P, Dalle JH, Platzbecker U, Downey GF, McNeil E, Demopoulos L, Dietz AC, Thakar HL, Orchard PJ, and Williams DA
- Subjects
- Adolescent, Child, Child, Preschool, Humans, Male, Brain diagnostic imaging, Brain pathology, Hematopoietic Stem Cell Transplantation, Magnetic Resonance Imaging, Follow-Up Studies, Treatment Outcome, Myelodysplastic Syndromes epidemiology, Myelodysplastic Syndromes genetics, Adrenoleukodystrophy diagnosis, Adrenoleukodystrophy genetics, Adrenoleukodystrophy mortality, Adrenoleukodystrophy therapy, ATP Binding Cassette Transporter, Subfamily D, Member 1 genetics, Genetic Therapy adverse effects, Genetic Therapy methods, Genetic Vectors administration & dosage, Genetic Vectors adverse effects, Lentivirus genetics
- Abstract
Background: Cerebral adrenoleukodystrophy is a severe form of X-linked adrenoleukodystrophy characterized by white-matter disease, loss of neurologic function, and early death. Elivaldogene autotemcel (eli-cel) gene therapy, which consists of autologous CD34+ cells transduced with Lenti-D lentiviral vector containing ABCD1 complementary DNA, is being tested in persons with cerebral adrenoleukodystrophy., Methods: In a phase 2-3 study, we evaluated the efficacy and safety of eli-cel therapy in boys with early-stage cerebral adrenoleukodystrophy and evidence of active inflammation on magnetic resonance imaging (MRI). The primary efficacy end point was survival without any of six major functional disabilities at month 24. The secondary end points included overall survival at month 24 and the change from baseline to month 24 in the total neurologic function score., Results: A total of 32 patients received eli-cel; 29 patients (91%) completed the 24-month study and are being monitored in the long-term follow-up study. At month 24, none of these 29 patients had major functional disabilities; overall survival was 94%. At the most recent assessment (median follow-up, 6 years), the neurologic function score was stable as compared with the baseline score in 30 of 32 patients (94%); 26 patients (81%) had no major functional disabilities. Four patients had adverse events that were directly related to eli-cel. Myelodysplastic syndrome (MDS) with excess blasts developed in 1 patient at month 92; the patient underwent allogeneic hematopoietic stem-cell transplantation and did not have MDS at the most recent follow-up., Conclusions: At a median follow-up of 6 years after lentiviral gene therapy, most patients with early cerebral adrenoleukodystrophy and MRI abnormalities had no major functional disabilities. However, insertional oncogenesis is an ongoing risk associated with the integration of viral vectors. (Funded by Bluebird Bio; ALD-102 and LTF-304 ClinicalTrials.gov numbers NCT01896102 and NCT02698579, respectively.)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
4. Secondary failure of lentiviral vector gene therapy in a cerebral adrenoleukodystrophy patient with an ABCD1 whole-gene deletion.
- Author
-
Lund TC, Orchard PJ, Nascene DR, King CJ, Braun J, Thakkar S, Durose W, Shestopalov I, Thakar H, and Gupta AO
- Subjects
- Humans, Male, Child, Magnetic Resonance Imaging, ATP-Binding Cassette Transporters genetics, Adrenoleukodystrophy therapy, Adrenoleukodystrophy genetics, ATP Binding Cassette Transporter, Subfamily D, Member 1 genetics, Genetic Therapy methods, Genetic Vectors genetics, Genetic Vectors administration & dosage, Lentivirus genetics, Hematopoietic Stem Cell Transplantation methods, Gene Deletion
- Abstract
A 9-year-old boy with adrenoleukodystrophy due to ABCD1 whole-gene deletion was diagnosed with active cerebral adrenoleukodystrophy characterized by demyelination and gadolinium enhancement on brain MRI. He underwent hematopoietic cell transplant (HCT) with autologous CD34
+ cells transduced with an ABCD1-expressing lentiviral vector (eli-cel [elivaldogene autotemcel]) as part of the ALD-104 clinical trial. Fifty days after HCT, the patient's MRI showed gadolinium resolution; the whole-blood vector copy number (VCN) was 0.666 copies/mL. Six months following HCT, an MRI showed re-emergence of gadolinium enhancement; the VCN had decreased to 0.029 copies/mL. Polyclonal antibodies to the ABCD1 gene product were detectable 9 months after transplant, showing reactivity to peroxisomes, suggesting an immune response; however, no antibody binding to human CD34+ cells could be shown. The patient underwent a successful allogeneic HCT 12 months after gene therapy with resultant gadolinium resolution, cerebral disease stabilization, and the disappearance of antibodies. The coincident VCN loss and appearance of antibody to the ABCD1 gene product is of interest, and we postulate that it is related to the patient's whole ABCD1 gene deletion. We suggest close monitoring of loss of gene therapy efficacy due to immune response in patients with full deletions who are considering gene therapy., Competing Interests: Declaration of interests H.T. and I.S. are employees of bluebird bio and own bluebird bio stock/stock options. bluebird bio provided funding for clinical trial ALD-104 (ClinicalTrials.gov: NCT03852498). T.C.L., P.J.O., D.R.N., and A.O.G. were investigators in the ALD-104 clinical trial., (Copyright © 2024 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
5. Meconium Ileal Obstruction and Functional Immaturity: Review.
- Author
-
Jesus LE, Lund TC, Regadas CT, Oliveira APP, Bruno RR, Moraes ACG, and Dekermacher S
- Abstract
Introduction: Ileal obstruction caused by thick meconium associated with functional immaturity (IOMFI) is an uncommon disease associated with prematurity. IOMFI is not well known, and late or wrong diagnosis is a problem. In this research, we review the clinical characteristics and therapeutic methods of IOMFI., Methods: Critical descriptive literature review., Results: Most patients eliminate meconium previously to IOMFI. More premature babies tend to become symptomatic in their second week of life, with progressive abdominal distension. The most frequent complication is perforation. Radiologically there is diffuse intestinal distention without air-fluid levels. In contrast enema a caliber transition zone is observed in the distal ileum with multiple filling defects in the ileum and colon. Neural ganglia are present in biopsies from the rectum, colon, and stoma, mostly with characteristics of immature ganglia. Most patients respond to treatment with water soluble contrast enemas. Surgery is needed for patients who do not respond to enemas and those presenting perforations., Discussion: IOMFI literature is limited to retrospective mainly small and heterogeneous cohorts. Patients usually respond promptly to water soluble enemas, but a favorable response is highly dependent in contrast reflux through the ileocecal valve and success is related to early treatment. Contrast inflow may be controlled by intermittent radiographies or real time ultrasound., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Lipidomic biomarkers in plasma correlate with disease severity in adrenoleukodystrophy.
- Author
-
Jaspers YRJ, Yska HAF, Bergner CG, Dijkstra IME, Huffnagel IC, Voermans MMC, Wever E, Salomons GS, Vaz FM, Jongejan A, Hermans J, Tryon RK, Lund TC, Köhler W, Engelen M, and Kemp S
- Abstract
Background: X-linked adrenoleukodystrophy (ALD) is a neurometabolic disorder caused by pathogenic variants in ABCD1 resulting very long-chain fatty acids (VLCFA) accumulation in plasma and tissues. Males can present with various clinical manifestations, including adrenal insufficiency, spinal cord disease, and leukodystrophy. Female patients typically develop spinal cord disease and peripheral neuropathy. Predicting the clinical outcome of an individual patient remains impossible due to the lack of genotype-phenotype correlation and predictive biomarkers., Methods: The availability of a large prospective cohort of well-characterized patients and associated biobank samples allowed us to investigate the relationship between lipidome and disease severity in ALD. We performed a lipidomic analysis of plasma samples from 24 healthy controls, 92 male and 65 female ALD patients., Results: Here we show that VLCFA are incorporated into different lipid classes, including lysophosphatidylcholines, phosphatidylcholines, triglycerides, and sphingomyelins. Our results show a strong association between higher levels of VLCFA-containing lipids and the presence of leukodystrophy, adrenal insufficiency, and severe spinal cord disease in male ALD patients. In female ALD patients, VLCFA-lipid levels correlate with X-inactivation patterns in blood mononuclear cells, and higher levels are associated with more severe disease manifestations. Finally, hematopoietic stem cell transplantation significantly reduces, but does not normalize, plasma C26:0-lysophosphatidylcholine levels in male ALD patients. Our findings are supported by the concordance of C26:0-lysophosphatidylcholine and total VLCFA analysis with the lipidomics results., Conclusions: This study reveals the profound impact of ALD on the lipidome and provides potential biomarkers for predicting clinical outcomes in ALD patients., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Targeting VEGF-mediated blood-brain barrier disruption in advanced cerebral leukodystrophy.
- Author
-
Gupta AO, Furcich JW, Nascene DR, Kemp S, King CJ, Nolan EE, Durose W, Miller BS, Orchard PJ, and Lund TC
- Subjects
- Humans, Male, Child, Adrenoleukodystrophy drug therapy, Bevacizumab therapeutic use, Bevacizumab pharmacology, Child, Preschool, Angiogenesis Inhibitors therapeutic use, Angiogenesis Inhibitors pharmacology, Blood-Brain Barrier drug effects, Blood-Brain Barrier metabolism, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
The earliest clinical manifestation of cerebral adrenoleukodystrophy (CALD) is adrenal insufficiency (AI) characterized by elevations in ACTH and loss of cortisol. We showed high (though physiologically achievable) levels of ACTH increases endothelial permeability, increases anisotropy, and increases VEGF secretion. An ACBD1 knockout endothelial cell line had increased sensitivity to ACTH and VEGF. Inhibition of VEGF via application of anti-VEGF (bevacizumab) improved permeability. Six boys with advanced CALD were treated with bevacizumab combined with dexamethasone and ruxolitinib as immune suppressants. Most boys had decreases in gadolinium enhancement on MRI indicating improvement in endothelial function, though all boys continued to progress symptomatically., Competing Interests: Declaration of competing interest AOG, JWF, DRN, SK, CJK, EEN, and WD have no conflicts. BSM is a consultant for Ascendis Pharma, BioMarin, Bristol Myers Squibb, EMD Serono, Endo Pharmaceuticals, Novo Nordisk, Pfizer, proventionbio and Tolmar and has received research support from Alexion, Abbvie, Aeterna Zentaris, Amicus, Lumos Pharma, Lysogene, Novo Nordisk, OPKO Health, Pfizer, Prevail Therapeutics and Sangamo Therapeutics. PJO and TCL have been principle investigators with bluebird bio., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
8. Hemangiosarcoma Cells Promote Conserved Host-derived Hematopoietic Expansion.
- Author
-
Kim JH, Schulte AJ, Sarver AL, Lee D, Angelos MG, Frantz AM, Forster CL, O'Brien TD, Cornax I, O'Sullivan MG, Cheng N, Lewellen M, Oseth L, Kumar S, Bullman S, Pedamallu CS, Goyal SM, Meyerson M, Lund TC, Breen M, Lindblad-Toh K, Dickerson EB, Kaufman DS, and Modiano JF
- Subjects
- Dogs, Animals, Humans, Mice, Tumor Microenvironment, Hematopoietic Stem Cells pathology, Hematopoiesis, Cell Differentiation, Hemangiosarcoma pathology, Hemangiosarcoma veterinary, Hemangiosarcoma genetics
- Abstract
Hemangiosarcoma and angiosarcoma are soft-tissue sarcomas of blood vessel-forming cells in dogs and humans, respectively. These vasoformative sarcomas are aggressive and highly metastatic, with disorganized, irregular blood-filled vascular spaces. Our objective was to define molecular programs which support the niche that enables progression of canine hemangiosarcoma and human angiosarcoma. Dog-in-mouse hemangiosarcoma xenografts recapitulated the vasoformative and highly angiogenic morphology and molecular characteristics of primary tumors. Blood vessels in the tumors were complex and disorganized, and they were lined by both donor and host cells. In a series of xenografts, we observed that the transplanted hemangiosarcoma cells created exuberant myeloid hyperplasia and gave rise to lymphoproliferative tumors of mouse origin. Our functional analyses indicate that hemangiosarcoma cells generate a microenvironment that supports expansion and differentiation of hematopoietic progenitor populations. Furthermore, gene expression profiling data revealed hemangiosarcoma cells expressed a repertoire of hematopoietic cytokines capable of regulating the surrounding stromal cells. We conclude that canine hemangiosarcomas, and possibly human angiosarcomas, maintain molecular properties that provide hematopoietic support and facilitate stromal reactions, suggesting their potential involvement in promoting the growth of hematopoietic tumors., Significance: We demonstrate that hemangiosarcomas regulate molecular programs supporting hematopoietic expansion and differentiation, providing insights into their potential roles in creating a permissive stromal-immune environment for tumor progression., (© 2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
9. CSF1R-related disorder: State of the art, challenges, and proposition of a new terminology.
- Author
-
Dulski J, Muthusamy K, Lund TC, and Wszolek ZK
- Subjects
- Adolescent, Adult, Humans, Mutation, Phenotype, Leukoencephalopathies genetics, Neuroglia
- Abstract
Recent developments in adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) and other disorders due to CSF1R variants led to the emergence of symptomatic and prophylactic treatment options. The growing body of knowledge on genetics, pathomechanisms, clinical, and radiological features in patients harboring CSF1R variants challenges the current concepts and terminology to define the disorders, in addition to bringing up new questions on genotype-phenotype relationships. Therefore, this paper discusses the present complexities and challenges in the research on ALSP due to CSF1R variants. We illustrate our new concepts with two cases that are compound heterozygotes for CSF1R variants. Although their clinical phenotype resembles ALSP, the diagnosis of brain abnormalities, neurodegeneration, and dysosteosclerosis (BANDDOS) seems more appropriate based on their genotype. As the diagnostic classification dilemma cannot be resolved with currently used concepts and terminology on these disorders, we propose a new nomenclature of "CSF1R-related disorder" with subcategories of "early-onset (<18 years old) and late-onset (≥18 years old) forms". We highlight the heterogeneity of CSF1R variant carriers in age at onset, spectrum and severity of clinical presentation, and progression rate, even within the same family. We argue that multiple factors, including genetic architecture and environment, converge to result in an individual's disease phenotype., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. IDH-mutant astrocytoma arising from a demyelinating plaque in a child with X-linked adrenoleukodystrophy.
- Author
-
Kalter JA, Yang RA, Toland A, Milla S, Lund TC, Hankinson T, and Dahl NA
- Subjects
- Child, Humans, Isocitrate Dehydrogenase genetics, Mutation genetics, Adrenoleukodystrophy genetics, Astrocytoma genetics, Brain Neoplasms diagnostic imaging, Brain Neoplasms genetics
- Published
- 2024
- Full Text
- View/download PDF
11. Natural history of craniovertebral abnormalities in a single-center study in 54 patients with Hurler syndrome.
- Author
-
Huang S, Nascene DR, Shanley R, Pena-Pino I, Lund TC, Gupta AO, Orchard PJ, and Sandoval-Garcia C
- Subjects
- Humans, Male, Female, Child, Preschool, Child, Retrospective Studies, Adolescent, Infant, Hematopoietic Stem Cell Transplantation, Decompression, Surgical methods, Disease Progression, Cervical Vertebrae surgery, Cervical Vertebrae diagnostic imaging, Young Adult, Mucopolysaccharidosis I complications, Mucopolysaccharidosis I surgery, Mucopolysaccharidosis I diagnostic imaging, Mucopolysaccharidosis I pathology
- Abstract
Objective: Craniovertebral junction (CVJ) abnormalities are common and well documented in mucopolysaccharidosis type I-Hurler syndrome (MPS IH), often causing severe spinal canal narrowing. However, the requirement for surgical decompression and/or fusion is uncommon. Although hematopoietic cell transplant (HCT) has been shown to prolong the lives of patients with MPS IH, its effect in halting or reversing musculoskeletal abnormalities is less clear. Unfortunately, there are currently no universal guidelines for imaging or indication for surgical interventions in these patients. The goal of this study was to track the progression of the CVJ anatomy in patients with MPS IH following HCT, and to examine radiographic features in patients who needed surgical intervention., Methods: Patients with MPS IH treated at the University of Minnesota with allogeneic HCT between 2008 and 2020 were retrospectively reviewed. Patients who underwent CVJ surgery were identified with chart review. All MPS IH cervical scans were examined, and the odontoid retroflexion angle, clivoaxial angle (CXA), canal width, and Grabb-Oakes distance (pB-C2) were measured yearly for up to 7 years after HCT. Longitudinal models based on the measurements were made. An intraclass correlation coefficient was used to measure interrater reliability. Nine children without MPS IH were examined for control CVJ measurements., Results: A total of 253 cervical spine MRI scans were reviewed in 54 patients with MPS IH. Only 4 (7.4%) patients in the study cohort required surgery. Three of them had posterior fossa and C1 decompression, and 1 had a C1-2 fusion. There was no statistically significant difference in the spinal parameters that were examined between surgery and nonsurgery groups. Among the measurements, canal width and CXA varied drastically in patients with different neck positions. Odontoid retroflexion angle and CXA tended to decrease with age. Canal width and pB-C2 tended to increase with age., Conclusions: Based on the data, the authors observed an increase in canal width and pB-C2, whereas the CXA and odontoid retroflexion angle became more acute as the patients aged after HCT. The longitudinal models derived from these data mirrored the development in children without MPS IH. Spinal measurements obtained on MR images alone are not sufficient in identifying patients who require surgical intervention. Symptom monitoring and clinical examination, as well as pathological spinal cord changes on MRI, are more crucial in assessing the need for surgery than is obtaining serial imaging.
- Published
- 2024
- Full Text
- View/download PDF
12. Comparative dose effectiveness of intravenous and intrathecal AAV9.CB7.hIDS, RGX-121, in mucopolysaccharidosis type II mice.
- Author
-
Smith MC, Belur LR, Karlen AD, Erlanson O, Furcich J, Lund TC, Seelig D, Kitto KF, Fairbanks CA, Kim KH, Buss N, and McIvor RS
- Abstract
Mucopolysaccharidosis type II (MPS II) is an X-linked recessive lysosomal disease caused by iduronate-2-sulfatase (IDS) deficiency, leading to accumulation of glycosaminoglycans (GAGs) and the emergence of progressive disease. Enzyme replacement therapy is the only currently approved treatment, but it leaves neurological disease unaddressed. Cerebrospinal fluid (CSF)-directed administration of AAV9.CB7.hIDS (RGX-121) is an alternative treatment strategy, but it is unknown if this approach will affect both neurologic and systemic manifestations. We compared the effectiveness of intrathecal (i.t.) and intravenous (i.v.) routes of administration (ROAs) at a range of vector doses in a mouse model of MPS II. While lower doses were completely ineffective, a total dose of 1 × 10
9 gc resulted in appreciable IDS activity levels in plasma but not tissues. Total doses of 1 × 1010 and 1 × 1011 gc by either ROA resulted in supraphysiological plasma IDS activity, substantial IDS activity levels and GAG reduction in nearly all tissues, and normalized zygomatic arch diameter. In the brain, a dose of 1 × 1011 gc i.t. achieved the highest IDS activity levels and the greatest reduction in GAG content, and it prevented neurocognitive deficiency. We conclude that a dose of 1 × 1010 gc normalized metabolic and skeletal outcomes, while neurologic improvement required a dose of 1 × 1011 gc, thereby suggesting the prospect of a similar direct benefit in humans., Competing Interests: The work detailed in this article was sponsored by REGENXBIO. K.H.K. and N.B. are current or former employees of Regenxbio. L.R.B., T.C.L., C.A.F., N.B., and R.S.M. are coinventors on patents related to the contents of this manuscript. R.S.M. is also an employee of Immusoft., (© 2024 The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
13. Loss of early myeloid donor cell engraftment into the central nervous system with nonmyeloablative conditioning.
- Author
-
Nolan EE, Durose W, Taghizadeh LA, King CJ, Gupta AO, Orchard PJ, Lorentson M, Braaten K, Furcich JW, and Lund TC
- Subjects
- Transplantation, Homologous, Bone Marrow Transplantation, Central Nervous System
- Published
- 2023
- Full Text
- View/download PDF
14. Hurler Syndrome Glycosaminoglycans Decrease in Cerebrospinal Fluid without Brain-Targeted Therapy.
- Author
-
Lund TC, Braunlin E, Polgreen LE, Gupta AO, Orchard PJ, and Eisengart JB
- Subjects
- Humans, Glycosaminoglycans therapeutic use, Brain, Blood-Brain Barrier, Enzyme Replacement Therapy, Mucopolysaccharidosis I drug therapy
- Abstract
Novel therapies for Hurler syndrome aim to cross the blood-brain barrier (BBB) to target neurodegeneration by degrading glycosaminoglycans (GAG). BBB penetration has been assumed with decreased cerebrospinal fluid (CSF) GAG, yet little is known about CSF GAG without brain-targeting therapies. We compared pre-transplant CSF GAG in patients who were treatment naïve (n = 19) versus receiving standard non-BBB penetrating enzyme replacement therapy (ERT, n = 12). In the ERT versus treatment naïve groups, CSF GAG was significantly lower across all content assayed, raising questions about using CSF GAG decrements to show BBB penetration. Future studies should compare GAG reduction in standard versus novel therapies. ANN NEUROL 2023;94:1182-1186., (© 2023 The Authors. Annals of Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.)
- Published
- 2023
- Full Text
- View/download PDF
15. No impact of CD34 + cell dose on outcome among children undergoing autologous hematopoietic stem cell transplant for high-risk neuroblastoma.
- Author
-
Knight TE, Ahn KW, Hebert KM, Atshan R, Wall DA, Chiengthong K, Lund TC, Prestidge T, Rangarajan HG, Dvorak CC, Auletta JJ, Kent M, Hashem H, Talano JA, Rotz SJ, Fraint E, Myers KC, Leung W, Sharma A, Bhatt NS, Driscoll TA, Yu LC, Schultz KR, Qayed M, Broglie L, Eapen M, and Yanik GA
- Subjects
- Humans, Child, Hematopoietic Stem Cells, Transplantation, Autologous, Antigens, CD34, Peripheral Blood Stem Cell Transplantation, Neuroblastoma therapy, Hematopoietic Stem Cell Transplantation
- Published
- 2023
- Full Text
- View/download PDF
16. Hematopoietic cell transplantation for Mucopolysaccharidosis I in the presence of decreased cardiac function.
- Author
-
Pillai NR, Elsbecker SA, Gupta AO, Lund TC, Orchard PJ, and Braunlin E
- Subjects
- Infant, Humans, Infant, Newborn, Retrospective Studies, Milrinone therapeutic use, Heart, Enzyme Replacement Therapy methods, Mucopolysaccharidosis I diagnosis, Hematopoietic Stem Cell Transplantation methods
- Abstract
Background: Severe mucopolysaccharidosis type I, (MPS IH) is a rare inherited lysosomal disorder resulting in progressive storage of proteoglycans (GAGs) in central nervous system and somatic tissues and, if left untreated, causing death within the first decade of life. Hematopoietic cell transplantation (HCT) arrests many of the features of MPS IH but carries a 10-15% risk of mortality. Decreased cardiac function can occur in MPS IH and increase the risk of HCT., Methods: Retrospective chart review was performed to determine the long-term outcome of individuals evaluated for HCT with MPS IH who had decreased cardiac function as measured by cardiac echocardiogram (echo) and ejection fraction (EF) of <50% at the time of initial evaluation., Results: Six patients ranging in age from 1 week to 21 months (median: 4 months) had EFs ranging from 25 to 47% (median: 32%) at diagnosis and were initiated on enzyme replacement therapy (ERT) with improvement in EF in three patients by 5 months. The remaining three patients continued to have EFs <50% and continuous milrinone infusion was added in the pre-HCT period. On average, milrinone infusion was able to be discontinued post-HCT, prior to hospital discharge, within a mean of 37 days. Five patients survived HCT and are alive today with normal EFs. One patient receiving milrinone died of sepsis during HCT with a normal EF., Conclusion: Decreased cardiac systolic function in infants with MPS IH that fails to normalize with ERT alone may benefit from the addition of continuous milrinone infusion during HCT., Competing Interests: Declaration of Competing Interest The authors have no relevant conflicts of interest to disclose., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Biomarker-based risk prediction for the onset of neuroinflammation in X-linked adrenoleukodystrophy.
- Author
-
Weinhofer I, Rommer P, Gleiss A, Ponleitner M, Zierfuss B, Waidhofer-Söllner P, Fourcade S, Grabmeier-Pfistershammer K, Reinert MC, Göpfert J, Heine A, Yska HAF, Casasnovas C, Cantarín V, Bergner CG, Mallack E, Forss-Petter S, Aubourg P, Bley A, Engelen M, Eichler F, Lund TC, Pujol A, Köhler W, Kühl JS, and Berger J
- Subjects
- Humans, Male, Child, Child, Preschool, Adolescent, Prognosis, Neuroinflammatory Diseases etiology, Neuroinflammatory Diseases blood, Neuroinflammatory Diseases diagnosis, Neuroinflammatory Diseases pathology, Cytokines blood, Retrospective Studies, Neurofilament Proteins blood, Risk Assessment, Adrenoleukodystrophy diagnosis, Adrenoleukodystrophy blood, Biomarkers blood
- Abstract
Background: X-linked adrenoleukodystrophy (X-ALD) is highly variable, ranging from slowly progressive adrenomyeloneuropathy to severe brain demyelination and inflammation (cerebral ALD, CALD) affecting males with childhood peak onset. Risk models integrating blood-based biomarkers to indicate CALD onset, enabling timely interventions, are lacking. Therefore, we evaluated the prognostic value of blood biomarkers in addition to current neuroimaging predictors for early detection of CALD., Methods: We measured blood biomarkers in a retrospective, male CALD risk-assessment cohort consisting of 134 X-ALD patients and 66 controls and in a phenotype-blinded validation set (25 X-ALD boys, 4-13 years) using Simoa®and Luminex® technologies., Findings: Among 25 biomarkers indicating axonal damage, astrocye/microglia activation, or immune-cell recruitment, neurofilament light chain (NfL) had the highest prognostic value for early indication of childhood/adolescent CALD. A plasma NfL cut-off level of 8.33 pg/mL, determined in the assessment cohort, correctly discriminated CALD with an accuracy of 96% [95% CI: 80-100] in the validation group. Multivariable logistic regression models revealed that combining NfL with GFAP or cytokines/chemokines (IL-15, IL-12p40, CXCL8, CCL11, CCL22, and IL-4) that were significantly elevated in CALD vs healthy controls had no additional benefit for detecting neuroinflammation. Some cytokines/chemokines were elevated only in childhood/adolescent CALD and already upregulated in asymptomatic X-ALD children (IL-15, IL-12p40, and CCL7). In adults, NfL levels distinguished CALD but were lower than in childhood/adolescent CALD patients with similar (MRI) lesion severity. Blood GFAP did not differentiate CALD from non-inflammatory X-ALD., Interpretation: Biomarker-based risk prediction with a plasma NfL cut-off value of 8.33 pg/mL, determined by ROC analysis, indicates CALD onset with high sensitivity and specificity in childhood X-ALD patients. A specific pro-inflammatory cytokine/chemokine profile in asymptomatic X-ALD boys may indicate a primed, immanent inflammatory state aligning with peak onset of CALD. Age-related differences in biomarker levels in adult vs childhood CALD patients warrants caution in predicting onset and progression of CALD in adults. Further evaluations are needed to assess clinical utility of the NfL cut-off for risk prognosis of CALD onset., Funding: Austrian Science Fund, European Leukodystrophy Association., Competing Interests: Declaration of interests MP received support from Amicus, Merck, Novartis and Sanofi-Genzyme; BZ received support from ACTRIMS 2022 and 2023 endMS SPRINT; JG received support from Quanterix; HAY was supported by an emerging investigator grant from ALD connect; CGB received grants from the German Research Foundation and the Ministry for Science and Culture of Lower Saxony; ME received support from Minoryx and is member of the advisory board of Minoryx, Poxel and SwanBio Therapeutics; FE is holding a license for “Intrathecal delivery of nucleic acid sequences encoding ABCD1 for treatment of Adrenomyeloneuropathy” (NO. 29539-021PCT), received consulting fees from SwanBio Therapeutics and UpToDate, is founder of SwanBio Therapeutics, ALD Connect and organizer of trial sites for ASPA, Bluebird Bio Therapeutics, Ionis Pharmaceuticals and Sanofi; AP received consulting fees from Swanbio Therapeutics and Sanofi and is member of the Advisory Board of Bluebird Bio Therapeutics and MedDay Therapeutics. JSK is member of the advisory board for Krabbe Disease of PassageBio. MCR received a grant from Novartis. EM has received funding from the National Institutes of Health (K23NS118044). All remaining authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Blood Levels of Environmental Heavy Metals are Associated with Poorer Iron Status in Ugandan Children: A Cross-Sectional Study.
- Author
-
Park S, Mupere E, Lund TC, Hodges JS, Moody EC, Colicino E, Georgieff MK, and Cusick SE
- Subjects
- Humans, Child, Child, Preschool, Iron metabolism, Cross-Sectional Studies, Uganda, Prospective Studies, Ferritins, Manganese, Biomarkers, Receptors, Transferrin, Metals, Heavy, Iron Deficiencies, Anemia, Iron-Deficiency
- Abstract
Background: Iron deficiency (ID) and environmental exposure to metals frequently co-occur among Ugandan children, but little is known about their associations, although iron and other divalent metals share the same intestinal absorption transporter, divalent metal transporter 1 (DMT1)., Objectives: We examined associations between iron status and blood concentrations of lead, manganese (Mn), cobalt (Co), and cadmium, both singly and as a mixture., Methods: We used data on sociodemographic status, iron biomarkers, and blood concentrations of heavy metals collected from a cross-sectional survey of 100 children aged 6-59 mo in Kampala, Uganda. We compared blood concentrations of metals in ID with iron-sufficient children. We examined associations between a metal mixture and iron biomarkers using multiple linear regression and weighted quintile sum regression., Results: The median (interquartile range) blood Mn (μg/L) was higher in ID children defined by soluble transferrin receptor (sTfR) and ferritin (ID compared with iron-sufficient children): (sTfR [21.3 {15.1, 28.8}, 11.2 {8.6, 18.5}], ferritin [19.5 {15.0, 27.2}, 11.2 {8.8, 19.6}]; P < 0.001 for both). Similarly, the median (interquartile range) blood Co (μg/L) was higher in ID children by ferritin ([0.5 {0.4, 0.9}, 0.4 {0.3, 0.5}], P = 0.05). Based on the multiple linear regression results, higher blood Co and Mn were associated with poorer iron status (defined by all 4 iron indicators for Co and by sTfR for Mn). The weighted quintile sum regression result showed that higher blood concentrations of a metal mixture were associated with poorer iron status represented by sTfR, ferritin, and hepcidin, mainly driven by Co and Mn., Conclusions: Our study findings suggest that poorer iron status is associated with overall heavy metal burden, predominantly Co and Mn, among Ugandan children. Further prospective studies should confirm our primary findings and investigate the combined effects of coexposures to neurotoxicants on the neurodevelopment of young children., (Copyright © 2023 American Society for Nutrition. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. VUS: Variant of uncertain significance or very unclear situation?
- Author
-
Kemp S, Orsini JJ, Ebberink MS, Engelen M, and Lund TC
- Subjects
- Male, Infant, Newborn, Humans, Neonatal Screening methods, Mutation, Missense, Adrenoleukodystrophy diagnosis
- Abstract
The advancements in population screening, including newborn screening, enables the identification of disease-causing variants and timely initiation of treatment. However, screening may also identify mild variants, non-disease variants, and variants of uncertain significance (VUS). The identification of a VUS poses a challenge in terms of diagnostic uncertainty and confusion. X-linked adrenoleukodystrophy (ALD) serves as an illustrative example of this complex issue. ALD is a monogenic neurometabolic disease with a complex clinical presentation and a lack of predictive tests for clinical severity. Despite the success of ALD newborn screening, a significant proportion (62%) of missense variants identified through newborn screening exhibit uncertainty regarding their pathogenicity. Resolving this issue requires ongoing efforts to accurately classify variants and refine screening protocols. While it is undisputable that ALD newborn screening greatly benefits boys with the disease, the identification of VUS underscores the need for continuous research and collaboration in improving screening practices., Competing Interests: Declaration of Competing Interest None., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
20. Persistent bone and joint disease despite current treatments for mucopolysaccharidosis types I, II, and VI: Data from a 10-year prospective study.
- Author
-
Miller BS, Fung EB, White KK, Lund TC, Harmatz P, Orchard PJ, Whitley CB, and Polgreen LE
- Subjects
- Child, Adult, Humans, Prospective Studies, Mucopolysaccharidosis I drug therapy, Mucopolysaccharidoses therapy, Mucopolysaccharidosis VI drug therapy, Mucopolysaccharidosis II drug therapy, Joint Diseases, Contracture
- Abstract
The mucopolysaccharidosis (MPS) disorders have many potential new therapies on the horizon. Thus, historic control data on disease progression and variability are urgently needed. We conducted a 10-year prospective observational study of 55 children with MPS IH (N = 23), MPS IA (N = 10), non-neuronopathic MPS II (N = 13), and MPS VI (N = 9) to systematically evaluate bone and joint disease. Annual measurements included height, weight, and goniometry. Mixed effects modeling was used to evaluate changes over time. All participants had been treated with hematopoietic cell transplantation and/or enzyme replacement therapy. Height z-score decreased over time in MPS IH, MPS II, and MPS VI, but not MPS IA. Adult heights were 136 ± 10 cm in MPS IH, 161 ± 11 cm in MPS IA, 161 ± 14 cm in MPS II, and 128 ± 15 cm in MPS VI. Adult average BMI percentiles were high: 75 ± 30%ile in MPS IH, 71 ± 37%ile in MPS IA, 71 ± 25%ile in MPS II, and 60 ± 42%ile in MPS VI. Every participant had joint contractures of the shoulders, elbows, hips, and/or knees. Joint contractures remained stable over time. In conclusion, despite current treatments for MPS I, II, and VI, short stature and joint contractures persist. The elevation in average BMI may be related, in part, to physical inactivity due to the ongoing bone and joint disease. Data from this longitudinal historical control study may be used to expedite testing of experimental bone and joint directed therapies and to highlight the need for weight management as part of routine clinical care for patients with MPS., (© 2023 The Authors. Journal of Inherited Metabolic Disease published by John Wiley & Sons Ltd on behalf of SSIEM.)
- Published
- 2023
- Full Text
- View/download PDF
21. Generation and characterization of an immunodeficient mouse model of mucopolysaccharidosis type II.
- Author
-
Smith MC, Belur LR, Karlen AD, Podetz-Pedersen K, Erlanson O, Laoharawee K, Furcich J, Lund TC, You Y, Seelig D, Webber BR, and McIvor RS
- Subjects
- Humans, Animals, Mice, Mice, Inbred NOD, Mice, SCID, Glycosaminoglycans, Mucopolysaccharidosis II therapy, Iduronate Sulfatase genetics
- Abstract
Mucopolysaccharidosis type II (Hunter syndrome, MPS II) is an inherited X-linked recessive disease caused by deficiency of iduronate-2-sulfatase (IDS), resulting in the accumulation of the glycosaminoglycans (GAG) heparan and dermatan sulfates. Mouse models of MPS II have been used in several reports to study disease pathology and to conduct preclinical studies for current and next generation therapies. Here, we report the generation and characterization of an immunodeficient mouse model of MPS II, where CRISPR/Cas9 was employed to knock out a portion of the murine IDS gene on the NOD/SCID/Il2rγ (NSG) immunodeficient background. IDS
-/- NSG mice lacked detectable IDS activity in plasma and all analyzed tissues and exhibited elevated levels of GAGs in those same tissues and in the urine. Histopathology revealed vacuolized cells in both the periphery and CNS of NSG-MPS II mice. This model recapitulates skeletal disease manifestations, such as increased zygomatic arch diameter and decreased femur length. Neurocognitive deficits in spatial memory and learning were also observed in the NSG-MPS II model. We anticipate that this new immunodeficient model will be appropriate for preclinical studies involving xenotransplantation of human cell products intended for the treatment of MPS II., (Copyright © 2023. Published by Elsevier Inc.)- Published
- 2023
- Full Text
- View/download PDF
22. Intracerebral lentiviral ABCD1 gene therapy in an early disease onset ALD mouse model.
- Author
-
Gong J, Liu Y, Chung TH, Xu L, Lund TC, and Chang LJ
- Subjects
- Animals, Mice, ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters metabolism, Tissue Distribution, Mice, Knockout, Genetic Therapy, ATP Binding Cassette Transporter, Subfamily D, Member 1 genetics, ATP Binding Cassette Transporter, Subfamily D, Member 1 metabolism, Adrenoleukodystrophy genetics, Adrenoleukodystrophy therapy, Adrenoleukodystrophy metabolism
- Abstract
X-linked adrenoleukodystrophy (ALD) is a genetic disorder of the ABCD1 gene. We aimed to treat ALD via direct intracerebral injection of lentiviral ABCD1 (LV.ABCD1). Lentiviral vectors (LVs) were injected into the brain of wild type mice to access toxicities and biodistribution. Confocal microscopy illustrated supraphysiological ABCD1 expression surrounding the injection sites, and LVs were also detected in the opposite site of the unilaterally injected brain. In multi-site bilateral injections (4, 6, 8, and 9 sites), LV.ABCD1 transduced most brain regions including the cerebellum. Investigation of neuronal loss, astrogliosis and microglia activation did not detect abnormality. For efficacy evaluation, a novel ALD knockout (KO) mouse model was established by deleting exons 3 to 9 of the ABCD1 gene based on CRISPR/Cas9 gene editing. The KO mice showed behavioral deficit in open-field test (OFT) and reduced locomotor activities in rotarod test at 6 and 7 months of age, respectively. We treated 3-month-old KO mice with bilateral LV.ABCD1 injections into the external capsule and thalamus. ABCD1 expression was detected 15 days later, and the impaired motor ability was gradually alleviated. Our studies established an early onset ALD model and illustrated neurological improvement after LV.ABCD1 intracerebral injection without immunopathological toxicity., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
23. A case series of adrenoleukodystrophy in children conceived through in vitro fertilization with an egg donor.
- Author
-
Chang C, Gupta AO, Orchard PJ, Nascene DR, Kierstein J, Tryon RK, and Lund TC
- Abstract
Objective: To report 3 cases of adrenoleukodystrophy (ALD) in children conceived by in vitro fertilization (IVF) and egg donation., Design: A case report., Patients: Patients aged 4-5 years old, evaluated by the University of Minnesota Leukodystrophy Center, who were diagnosed with ALD after being conceived by IVF with oocytes provided by the same donor., Interventions: One patient received a hematopoietic stem cell transplant from a human leukocyte antigen-matched donor, and 1 patient received autologous lentiviral corrected hematopoietic cells. The disease state in 1 patient was unfortunately too advanced for effective treatment to be administered., Main Outcome Measures: Progression of disease after diagnosis or treatment was observed by cerebral magnetic resonance imaging and monitoring the development or advancement of any cognitive, adaptive, and motor deficits., Results: Patients who received a transplant for ALD successfully experienced little to no disease progression at least 6 months to 1 year after treatment., Conclusions: These 3 cases of transmission of ALD through oocyte donation and IVF highlight the potential need to implement more comprehensive genetic screening of gamete donors to prevent the transfer of rare but severe genetic diseases through IVF. Further, these cases highlight limitations in carrier screening guidelines that limit reportable variants to pathogenic and likely pathogenic variants., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
24. Hematopoietic stem cell transplant for Hurler syndrome: does using bone marrow or umbilical cord blood make a difference?
- Author
-
Orchard PJ, Gupta AO, Eisengart JB, Polgreen LE, Pollard LM, Braunlin E, Pasquali M, and Lund TC
- Subjects
- Humans, Fetal Blood, Bone Marrow, Bone Marrow Transplantation, Mucopolysaccharidosis I therapy, Hematopoietic Stem Cell Transplantation
- Published
- 2022
- Full Text
- View/download PDF
25. Hematopoietic Stem Cell Transplantation in CSF1R -Related Leukoencephalopathy: Retrospective Study on Predictors of Outcomes.
- Author
-
Dulski J, Heckman MG, White LJ, Żur-Wyrozumska K, Lund TC, and Wszolek ZK
- Abstract
Mutations in the CSF1R gene are the most common cause of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), a neurodegenerative disease with rapid progression and ominous prognosis. Hematopoietic stem cell transplantation (HSCT) has been increasingly offered to patients with CSF1R -ALSP. However, different therapy results were observed, and it was not elucidated which patient should be referred for HSCT. This study aimed to determine predictors of good and bad HSCT outcomes in CSF1R -ALSP. We retrospectively analyzed 15 patients, 14 symptomatic and 1 asymptomatic, with CSF1R -ALSP that underwent HSCT. Median age of onset was 39 years, and the median age of HSCT was 43 years. Cognitive impairment was the most frequent initial manifestation (43%), followed by gait problems (21%) and neuropsychiatric symptoms (21%). Median post-HSCT follow-up was 26 months. Good outcomes were associated with gait problems as initial ( p = 0.041) and predominant ( p = 0.017) manifestation and younger age at HSCT ( p = 0.044). Cognitive impairment as first manifestation was a predictor of a bad outcome ( p = 0.016) and worsening of cognition post-HSCT ( p = 0.025). In conclusion, gait problems indicated a milder phenotype with better response to HSCT and good therapy outcomes. In contrast, patients with a higher burden of cognitive symptoms were most likely not to benefit from HSCT.
- Published
- 2022
- Full Text
- View/download PDF
26. Phenotypic Correction of Murine Mucopolysaccharidosis Type II by Engraftment of Ex Vivo Lentiviral Vector-Transduced Hematopoietic Stem and Progenitor Cells.
- Author
-
Smith MC, Belur LR, Karlen AD, Erlanson O, Podetz-Pedersen KM, McKenzie J, Detellis J, Gagnidze K, Parsons G, Robinson N, Labarre S, Shah S, Furcich J, Lund TC, Tsai HC, McIvor RS, and Bonner M
- Subjects
- Animals, Mice, Humans, Leukocytes, Mononuclear, Enzyme Replacement Therapy, Disease Models, Animal, Hematopoietic Stem Cells, Mucopolysaccharidosis II genetics, Mucopolysaccharidosis II therapy, Iduronate Sulfatase genetics
- Abstract
Mucopolysaccharidosis type II (MPS II, Hunter syndrome) is an X-linked recessive lysosomal disease caused by deficiency of iduronate-2-sulfatase (IDS). The absence of IDS results in the accumulation of the glycosaminoglycans (GAGs) heparan sulfate and dermatan sulfate. Currently, the only approved treatment option for MPS II is enzyme replacement therapy (ERT), Elaprase. However, ERT is demanding for the patient and does not ameliorate neurological manifestations of the disease. Using an IDS-deficient mouse model that phenocopies the human disease, we evaluated hematopoietic stem and progenitor cells (HSPCs) transduced with a lentiviral vector (LVV) carrying a codon-optimized human IDS coding sequence regulated by a ubiquitous MNDU3 promoter (MNDU3-IDS). Mice treated with MNDU3-IDS LVV-transduced cells showed supraphysiological levels of IDS enzyme activity in plasma, peripheral blood mononuclear cells, and in most analyzed tissues. These enzyme levels were sufficient to normalize GAG storage in analyzed tissues. Importantly, IDS levels in the brains of MNDU3-IDS-engrafted animals were restored to 10-20% than that of wild-type mice, sufficient to normalize GAG content and prevent emergence of cognitive deficit as evaluated by neurobehavioral testing. These results demonstrate the potential effectiveness of ex vivo MNDU3-IDS LVV-transduced HSPCs for treatment of MPS II.
- Published
- 2022
- Full Text
- View/download PDF
27. International Recommendations for the Diagnosis and Management of Patients With Adrenoleukodystrophy: A Consensus-Based Approach.
- Author
-
Engelen M, van Ballegoij WJC, Mallack EJ, Van Haren KP, Köhler W, Salsano E, van Trotsenburg ASP, Mochel F, Sevin C, Regelmann MO, Tritos NA, Halper A, Lachmann RH, Davison J, Raymond GV, Lund TC, Orchard PJ, Kuehl JS, Lindemans CA, Caruso P, Turk BR, Moser AB, Vaz FM, Ferdinandusse S, Kemp S, Fatemi A, Eichler FS, and Huffnagel IC
- Subjects
- Infant, Newborn, Humans, Male, Consensus, Neonatal Screening methods, Adrenoleukodystrophy diagnosis, Adrenoleukodystrophy genetics, Adrenoleukodystrophy therapy, Hematopoietic Stem Cell Transplantation adverse effects, Adrenal Insufficiency diagnosis
- Abstract
Pathogenic variants in the ABCD1 gene cause adrenoleukodystrophy (ALD), a progressive metabolic disorder characterized by 3 core clinical syndromes: a slowly progressive myeloneuropathy, a rapidly progressive inflammatory leukodystrophy (cerebral ALD), and primary adrenal insufficiency. These syndromes are not present in all individuals and are not related to genotype. Cerebral ALD and adrenal insufficiency require early detection and intervention and warrant clinical surveillance because of variable penetrance and age at onset. Newborn screening has increased the number of presymptomatic individuals under observation, but clinical surveillance protocols vary. We used a consensus-based modified Delphi approach among 28 international ALD experts to develop best-practice recommendations for diagnosis, clinical surveillance, and treatment of patients with ALD. We identified 39 discrete areas of consensus. Regular monitoring to detect the onset of adrenal failure and conversion to cerebral ALD is recommended in all male patients. Hematopoietic cell transplant (HCT) is the treatment of choice for cerebral ALD. This guideline addresses a clinical need in the ALD community worldwide as the number of overall diagnoses and presymptomatic individuals is increasing because of newborn screening and greater availability of next-generation sequencing. The poor ability to predict the disease course informs current monitoring intervals but remains subject to change as more data emerge. This knowledge gap should direct future research and illustrates once again that international collaboration among physicians, researchers, and patients is essential to improving care., (Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.)
- Published
- 2022
- Full Text
- View/download PDF
28. Treatment of cerebral adrenoleukodystrophy: allogeneic transplantation and lentiviral gene therapy.
- Author
-
Gupta AO, Raymond G, Pierpont EI, Kemp S, McIvor RS, Rayannavar A, Miller B, Lund TC, and Orchard PJ
- Subjects
- Adult, Genetic Therapy, Humans, Male, Transplantation, Homologous, Adrenoleukodystrophy genetics, Adrenoleukodystrophy pathology, Adrenoleukodystrophy therapy, Hematopoietic Stem Cell Transplantation
- Abstract
Introduction: Adrenoleukodystrophy (ALD) is an X-linked peroxisomal disorder with an incidence of 1 in 14-17,000 male births, caused by pathogenic variants within the ABCD1 gene. By adulthood, approximately 40% of the patients develop cerebral ALD, a severe, neuroinflammatory condition that is generally progressive and fatal without intervention., Areas Covered: Historically, only allogeneic hematopoietic stem cell transplantation (HSCT) has been shown to halt progression of cerebral ALD, with superior outcomes obtained when HSCT is performed early in the disease process. More recently, a lentiviral-based gene therapy approach has been investigated as therapy for cerebral ALD as an alternative to allogeneic transplantation. A focused literature review was performed using the terms 'hematopoietic stem cell transplantation,' 'gene therapy' and 'adrenoleukodystrophy' to include relevant literature, especially comparing the experience with gene therapy and HSCT outcomes. We review the history and experience with HSCT in cerebral ALD and its limitations, as well as the information currently available in association with the gene therapy trials for cerebral ALD., Expert Opinion: The data regarding this lentiviral-based gene therapy approach and its relative risks and benefits is still being evaluated. This information is explored in the context of the experience with allogeneic HSCT for cerebral ALD.
- Published
- 2022
- Full Text
- View/download PDF
29. Glycoprotein nonmetastatic melanoma protein B (GNMPB) as a novel biomarker for cerebral adrenoleukodystrophy.
- Author
-
Taghizadeh LA, King CJ, Nascene DR, Gupta AO, Orchard PJ, Higgins L, Markowski TW, Nolan EE, Furcich JW, and Lund TC
- Subjects
- Biomarkers metabolism, Disease Progression, Humans, Receptors, Fc, Adrenoleukodystrophy diagnosis, Adrenoleukodystrophy genetics, Melanoma, Membrane Glycoproteins metabolism
- Abstract
Adrenoleukodystrophy (ALD) is an X-linked peroxisomal disease caused by a mutation in the ABCD1 gene, producing mutations in the very long chain fatty acid transporter, ALD protein. Cerebral ALD (cALD) is a severe phenotype of ALD with neuroinflammation and neurodegeneration. Elevated levels of Glycoprotein Nonmetastatic Melanoma Protein B (GNMPB) have been recently documented in neurodegenerative diseases such as Alzheimer's disease, Multiple Sclerosis and Amyotrophic Lateral Sclerosis. Our objective was to measure the levels cerebral spinal fluid (CSF) GNMPB in cALD patients to determine if GNMPB could be a potential biomarker in tracking cALD disease progression. CSF GNMPB levels were significantly higher in cALD patients versus controls (2407 ± 1672 pg/mL vs. 639.5 ± 404 pg/mL, p = 0.0009). We found a positive correlation between CSF GNMPB and MRI disease severity score levels (R
2 = 0.3225, p < 0.0001) as well as the gadolinium intensity score (p = 0.0204). Boys with more severe neurologic deficits also had higher levels of CSF GNMPB (p < 0.0001). A positive correlation was shown between CSF GNMPB and another biomarker, chitotriosidase (R2 = 0.2512, p = 0.0244). These data show that GNMPB could be a potential biomarker of cALD disease state and further studies should evaluate it as a predictor of the disease progression., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
30. Primary Adrenal Insufficiency in a Boy with Type I Diabetes: The Importance of Considering X-linked Adrenoleukodystrophy.
- Author
-
Wiersma RE, Gupta AO, Lund TC, Sarafoglou K, Pierpont EI, Orchard PJ, and Miller BS
- Abstract
Primary adrenal insufficiency (PAI) is often the first clinical sign of X-linked adrenoleukodystrophy (X-ALD), a rare genetic disorder that can present with various clinical phenotypes. A subset of boys with X-ALD develop cerebral ALD (cALD), characterized by progressive central demyelination, neurocognitive decline, and ultimately death. Timely intervention with hematopoietic cell transplant (HCT) can be a life-saving therapy by stopping progression of cerebral disease. We report the case of an 11-year-old boy with type 1 diabetes mellitus who presented with PAI, growth delay, and symptoms of attention deficit hyperactivity disorder. Given his history of T1DM, his PAI was presumed to be autoimmune and he was started on hydrocortisone and fludrocortisone. Eleven months later brain magnetic resonance imaging revealed white matter hyperintensity consistent with advanced cALD. The degree of disease progression at the time of diagnosis rendered the patient ineligible for transplant and he has continued to experience progressive neurologic decline. Initial symptoms of cALD are often subtle but should not be overlooked, as early identification of X-ALD is critical to allow early intervention with lifesaving HCT. PAI typically presents prior to the onset of neurologic symptoms. All boys who present with PAI should undergo workup for X-ALD with plasma very long chain fatty acid testing, even in the setting of underlying autoimmune disease., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2022
- Full Text
- View/download PDF
31. Evaluation of Neurofilament Light Chain as a Biomarker of Neurodegeneration in X-Linked Childhood Cerebral Adrenoleukodystrophy.
- Author
-
Wang H, Davison MD, Kramer ML, Qiu W, Gladysheva T, Chiang RMS, Kayatekin C, Nascene DR, Taghizadeh LA, King CJ, Nolan EE, Gupta AO, Orchard PJ, and Lund TC
- Subjects
- Biomarkers metabolism, Child, Humans, Intermediate Filaments metabolism, Male, Sweden, Adrenoleukodystrophy diagnosis, Adrenoleukodystrophy genetics, Adrenoleukodystrophy metabolism
- Abstract
Cerebral adrenoleukodystrophy (CALD) is a devastating, demyelinating neuroinflammatory manifestation found in up to 40% of young males with an inherited mutation in ABCD1 , the causative gene in adrenoleukodystrophy. The search for biomarkers which correlate to CALD disease burden and respond to intervention has long been sought after. We used the Olink Proximity Extension Assay (Uppsala, Sweden) to explore the cerebral spinal fluid (CSF) of young males with CALD followed by correlative analysis with plasma. Using the Target 96 Neuro Exploratory panel, we found that, of the five proteins significantly increased in CSF, only neurofilament light chain (NfL) showed a significant correlation between CSF and plasma levels. Young males with CALD had a 11.3-fold increase in plasma NfL compared with controls. Importantly, 9 of 11 young males with CALD who underwent HCT showed a mean decrease in plasma NfL of 50% at 1 year after HCT compared with pre-HCT levels. In conclusion, plasma NfL could be a great value in determining outcomes in CALD and should be scrutinized in future studies in patients prior to CALD development and after therapeutic intervention.
- Published
- 2022
- Full Text
- View/download PDF
32. Busulfan dose Recommendation in Inherited Metabolic Disorders: Population Pharmacokinetic Analysis.
- Author
-
Takahashi T, Illamola SM, Jennissen CA, Long SE, Lund TC, Orchard PJ, Gupta AO, and Long-Boyle JR
- Subjects
- Body Weight, Busulfan therapeutic use, Child, Humans, Transplantation Conditioning, Young Adult, Hematopoietic Stem Cell Transplantation adverse effects, Metabolic Diseases chemically induced
- Abstract
Busulfan is a commonly used alkylating agent in the conditioning regimens of hematopoietic cell transplantation (HCT). Population pharmacokinetic (popPK) models enable description of busulfan PK and optimization of exposure, which leads to improvement of event-free survival after HCT. Prior busulfan popPK analysis has been limited by small numbers in patients with inherited metabolic disorders (IMD). The primary objective was to characterize population PK of busulfan in a large cohort of children and young adults undergoing HCT for IMD. PopPK analysis of busulfan drug concentrations was performed using data from 78 patients with IMD who received intravenous busulfan (every 24 hours, 4 doses) as part of pretransplantation combination chemotherapy. The final model for busulfan drug clearance was used to estimate individual doses aimed to achieve a target cumulative area under the curve (cAUC) of 80 to 100 mg · h/L. We then compared the probability of cAUC within the range of 80 to 100 mg · h/L by the developed dosing regimen versus conventional regimen. A 1-compartment, linear elimination model best described the PK of busulfan. Significant covariates demonstrated to affect busulfan clearance included total body weight and the time (in days) from busulfan infusion start. The probability of target cAUC attainment by the developed dosing versus the conventional dosing were 47% versus 43% for body weight <12 kg, and 48% versus 36% for body weight ≥12 kg. We described population PK of intravenous busulfan in a large IMD cohort. The proposed dosing regimen based on the developed model can improve the target cAUC attainment of busulfan for IMD., (Copyright © 2021 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
33. Open-Label Pilot Study of Interferon Gamma-1b in Patients With Non-Infantile Osteopetrosis.
- Author
-
Nguyen A, Miller WP, Gupta A, Lund TC, Schiferl D, Lam LSK, Arzumanyan Z, Orchard PJ, and Polgreen LE
- Abstract
The only treatment currently available for patients with severe infantile osteopetrosis is hematopoietic cell transplantation (HCT). HCT-related toxicity and mortality risks typically preclude its use in non-infantile patients, and other therapies are needed for these patients who have significant disease-related morbidity. Interferon gamma-1b is currently approved by the U.S. Food and Drug Administration (FDA) for treatment of severe infantile osteopetrosis (autosomal recessive osteopetrosis [ARO]). However, little is known about the effects of interferon gamma-1b in non-infantile osteopetrosis. Thus, this pilot study aimed at testing the safety and tolerability of interferon gamma-1b in patients with non-infantile osteopetrosis and assessing the clinical effects. We performed a 12-month, open-label, multi-center pilot study involving patients >1 year-old diagnosed radiographically with osteopetrosis. Patients were initiated on interferon gamma-1b subcutaneously 15 μg/m
2 three times weekly, to be titrated over 3 weeks to a goal of 100 μg/m2 three times weekly. The primary aim was safety and tolerability. The secondary aims were to assess changes in peripheral quantitative computed tomography (pQCT), dual-energy x-ray absorptiometry (DXA) bone mineral density (BMD) Z-scores, bone biomarkers, and quality-of-life (QOL) measures. Four of the five participants enrolled withdrew from the study between 3 and 9 months due to intolerability of interferon gamma-1b-related flu-like symptoms. The last participant completed the study with the addition of prednisone on days of interferon gamma-1b administration. DXA and pQCT outcomes were stable over 6-12 months, and there were no clear trends in bone biomarkers or QOL measures. No serious drug-related adverse events were reported during this study. Interferon gamma-1b was only tolerable in one of five participants with the addition of prednisone. The stabilization of BMD and other measures of bone health during this study suggest possible positive effects of interferon gamma-1b on osteopetrosis; however, additional data are needed before conclusions on treatment efficacy can be made. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research., Competing Interests: The content of this manuscript is solely the responsibility of the authors. WPM is a full‐time employee of Audentes Therapeutics, and Astellas Company; he was employed by the University of Minnesota at the time of data collection. There are no other relevant conflicts of interest to report., (© 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.)- Published
- 2022
- Full Text
- View/download PDF
34. Isoprostanoid Plasma Levels Are Relevant to Cerebral Adrenoleukodystrophy Disease.
- Author
-
Signorini C, De Felice C, Durand T, Galano JM, Oger C, Leoncini S, Hayek J, Lee JC, Lund TC, and Orchard PJ
- Abstract
Cerebral adrenoleukodystrophy (ALD) is a rare neuroinflammatory disorder characterized by progressive demyelination. Mutations within the ABCD1 gene result in very long-chain fatty acid (VLCFA) accumulation within the peroxisome, particularly in the brain. While this VLCFA accumulation is known to be the driving cause of the disease, oxidative stress can be a contributing factor. For patients with early cerebral disease, allogeneic hematopoietic stem cell transplantation (HSCT) is the standard of care, and this can be supported by antioxidants. To evaluate the involvement of fatty acid oxidation in the disease, F
2 -isoprostanes (F2 -IsoPs), F2- dihomo-isoprostanes (F2 -dihomo-IsoPs) and F4 -neuroprostanes (F4 -NeuroPs)-which are oxygenated metabolites of arachidonic (ARA), adrenic (AdA) and docosahexaenoic (DHA) acids, respectively-in plasma samples from ALD subjects ( n = 20)-with various phenotypes of the disease-were measured. Three ALD groups were classified according to patients with: (1) confirmed diagnosis of ALD but without cerebral disease; (2) cerebral disease in early period post-HSCT (<100 days post-HSCT) and on intravenous N-acetyl-L-cysteine (NAC) treatment; (3) cerebral disease in late period post-HSCT (beyond 100 days post-HSCT) and off NAC therapy. In our observation, when compared to healthy subjects ( n = 29), in ALD (i), F2 -IsoPs levels were significantly ( p < 0.01) increased in all patients, with the single exception of the early ALD and on NAC subjects; (ii) significant elevated ( p < 0.0001) amounts of F2 -dihomo-IsoPs were detected, with the exception of patients with a lack of cerebral disease; (iii), a significant increase ( p < 0.003) in F4 -NeuroP plasma levels was detected in all ALD patients. Moreover, F2 -IsoPs plasma levels were significantly higher ( p = 0.038) in early ALD in comparison to late ALD stage, and F4 -NeuroPs were significantly lower ( p = 0.012) in ALD subjects with a lack of cerebral disease in comparison to the late disease stage. Remarkably, plasma amounts of all investigated isoprostanoids were shown to discriminate ALD patients vs. healthy subjects. Altogether, isoprostanoids are relevant to the phenotype of X-ALD and may be helpful in predicting the presence of cerebral disease and establishing the risk of progression.- Published
- 2022
- Full Text
- View/download PDF
35. Hematopoietic cell transplantation for sialidosis type I.
- Author
-
Gupta AO, Patterson MC, Wood T, Eisengart JB, Orchard PJ, and Lund TC
- Abstract
We report the clinical and laboratory follow-up data of an adolescent female with Type I Sialidosis who underwent bone marrow transplant (BMT). After BMT, plasma and urine biomarkers responded concurrently with engraftment. Neuropsychiatry data showed preservation in some domains, but she did have overall decline in motor performance. Sialidosis is a very rare lysosomal condition, and we believe this to be the first report of a case of Type I Sialidosis undergoing BMT., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
36. Differential outcomes for frontal versus posterior demyelination in childhood cerebral adrenoleukodystrophy.
- Author
-
Gupta AO, Nascene DR, Shanley R, Kenney-Jung DL, Eisengart JB, Lund TC, Orchard PJ, and Pierpont EI
- Subjects
- Adolescent, Adrenoleukodystrophy complications, Adrenoleukodystrophy diagnostic imaging, Child, Child, Preschool, Demyelinating Diseases diagnostic imaging, Emotions, Frontal Lobe diagnostic imaging, Humans, Magnetic Resonance Imaging, Male, Mental Disorders diagnostic imaging, Neuropsychological Tests, Retrospective Studies, Severity of Illness Index, Treatment Outcome, White Matter diagnostic imaging, Adrenoleukodystrophy surgery, Demyelinating Diseases etiology, Frontal Lobe pathology, Hematopoietic Stem Cell Transplantation, Mental Disorders etiology, White Matter pathology
- Abstract
In the most common variant of childhood cerebral adrenoleukodystrophy (cALD), demyelinating brain lesions are distributed predominately in parieto-occipital white matter. Less frequently, lesions first develop in frontal white matter. This matched cohort study examined whether outcomes after standard treatment with hematopoietic cell transplantation (HCT) differ in patients with early stage frontal lesions as compared to parieto-occipital lesions. Retrospective chart review identified seven pediatric patients with frontal cALD lesions and MRI severity score < 10 who underwent a single HCT at our center between 1990 and 2019. Concurrent MRI, neurocognitive and psychiatric outcomes at last comprehensive follow-up (mean 1.2 years; range 0.5-2.1 years) were compared with a group of seven boys with the parieto-occipital variant matched on pre-HCT MRI severity score. Both groups showed similar rates of transplant complications and radiographic disease advancement. Neurocognitive outcomes were broadly similar, with more frequent working memory deficits among individuals with frontal lesions. Psychiatric problems (hyperactivity, aggression, and atypical behavior) were considerably more common and severe among patients with frontal lesions. Aligned with the critical role of the frontal lobes in emotional and behavioral regulation, functional disruption of self-regulation skills is widely observed among patients with frontal lesions. Comprehensive care for cALD should address needs for psychiatric care and management., (© 2021 The Authors. Journal of Inherited Metabolic Disease published by John Wiley & Sons Ltd on behalf of SSIEM.)
- Published
- 2021
- Full Text
- View/download PDF
37. An irradiated marrow niche reveals a small noncollagenous protein mediator of homing, dermatopontin.
- Author
-
Kramer AC, Astuti Y, Elfstrum A, Lehrke MJ, Tolar J, Blazar BR, Blake AL, Taisto ME, Furcich JW, Nolan EE, Durose WW, Webber BR, Geisness A, Wood DK, and Lund TC
- Subjects
- Animals, Cell Adhesion, Hematopoietic Stem Cells, Mice, Zebrafish, Bone Marrow, Hematopoietic Stem Cell Transplantation
- Abstract
Hematopoietic cell homing after hematopoietic cell transplant (HCT) is governed by several pathways involving marrow niche cells that are evoked after pre-HCT conditioning. To understand the factors that play a role in homing, we performed expression analysis on zebrafish marrow niche cells following conditioning. We determined that the noncollagenous protein extracellular matrix related protein dermatopontin (Dpt) was upregulated sevenfold in response to irradiation. Studies in mice revealed DPT induction with radiation and lipopolysaccharide exposure. Interestingly, we found that coincubation of zebrafish or murine hematopoietic cells with recombinant DPT impedes hematopoietic stem and progenitor cell homing by 50% and 86%, respectively. Similarly, this translated into a 24% reduction in long-term engraftment (vs control; P = .01). We found DPT to interact with VLA-4 and block hematopoietic cell-endothelial cell adhesion and transendothelial migration. Finally, a DPT-knockout mouse displayed a 60% increase in the homing of hematopoietic cells vs wild-type mice (P = .03) with a slight improvement in long-term lin-SCA1+cKIT+-SLAM cell engraftment (twofold; P = .04). These data show that the extracellular matrix-related protein DPT increases with radiation and transiently impedes the transendothelial migration of hematopoietic cells to the marrow., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
38. Differences in MPS I and MPS II Disease Manifestations.
- Author
-
Hampe CS, Yund BD, Orchard PJ, Lund TC, Wesley J, and McIvor RS
- Subjects
- Animals, Humans, Mucopolysaccharidosis I etiology, Mucopolysaccharidosis II etiology, Corneal Diseases complications, Epidermal Cells pathology, Mucopolysaccharidosis I pathology, Mucopolysaccharidosis II pathology, Nervous System Diseases complications
- Abstract
Mucopolysaccharidosis (MPS) type I and II are two closely related lysosomal storage diseases associated with disrupted glycosaminoglycan catabolism. In MPS II, the first step of degradation of heparan sulfate (HS) and dermatan sulfate (DS) is blocked by a deficiency in the lysosomal enzyme iduronate 2-sulfatase (IDS), while, in MPS I, blockage of the second step is caused by a deficiency in iduronidase (IDUA). The subsequent accumulation of HS and DS causes lysosomal hypertrophy and an increase in the number of lysosomes in cells, and impacts cellular functions, like cell adhesion, endocytosis, intracellular trafficking of different molecules, intracellular ionic balance, and inflammation. Characteristic phenotypical manifestations of both MPS I and II include skeletal disease, reflected in short stature, inguinal and umbilical hernias, hydrocephalus, hearing loss, coarse facial features, protruded abdomen with hepatosplenomegaly, and neurological involvement with varying functional concerns. However, a few manifestations are disease-specific, including corneal clouding in MPS I, epidermal manifestations in MPS II, and differences in the severity and nature of behavioral concerns. These phenotypic differences appear to be related to different ratios between DS and HS, and their sulfation levels. MPS I is characterized by higher DS/HS levels and lower sulfation levels, while HS levels dominate over DS levels in MPS II and sulfation levels are higher. The high presence of DS in the cornea and its involvement in the arrangement of collagen fibrils potentially causes corneal clouding to be prevalent in MPS I, but not in MPS II. The differences in neurological involvement may be due to the increased HS levels in MPS II, because of the involvement of HS in neuronal development. Current treatment options for patients with MPS II are often restricted to enzyme replacement therapy (ERT). While ERT has beneficial effects on respiratory and cardiopulmonary function and extends the lifespan of the patients, it does not significantly affect CNS manifestations, probably because the enzyme cannot pass the blood-brain barrier at sufficient levels. Many experimental therapies, therefore, aim at delivery of IDS to the CNS in an attempt to prevent neurocognitive decline in the patients.
- Published
- 2021
- Full Text
- View/download PDF
39. Consensus opinion on immune-mediated cytopenias after hematopoietic cell transplant for inherited metabolic disorders.
- Author
-
Gupta AO, Jan Boelens J, Ebens CL, Kurtzberg J, Lund TC, Smith AR, Wagner JE, Wynn R, Blazar BR, and Orchard PJ
- Subjects
- Consensus, Humans, Transplantation Conditioning, Hematopoietic Stem Cell Transplantation adverse effects, Metabolic Diseases etiology, Thrombocytopenia etiology
- Abstract
Hematopoietic stem cell transplantation (HCT) has been increasingly used for patients with inherited metabolic disorders (IMD). Immune mediated cytopenias (IMCs) after HCT, manifesting as hemolytic anemia, thrombocytopenia, and/or neutropenia, are recognized as a significant complication in this patient population, yet our understanding of the incidence, risk factors, and pathophysiology is currently limited. Review of the published literature demonstrates a higher incidence in younger patients who undergo HCT for a nonmalignant disease indication. However, a few reports suggest that the incidence is even higher among those with IMD (incidence ranging from 10 to 56%). This review summarizes the literature, provides an approach to better understanding of the possible etiology of IMCs, and proposes a diagnostic and management plan for patients with IMD who develop single or multi-lineage cytopenias after HCT.
- Published
- 2021
- Full Text
- View/download PDF
40. MRI surveillance of boys with X-linked adrenoleukodystrophy identified by newborn screening: Meta-analysis and consensus guidelines.
- Author
-
Mallack EJ, Turk BR, Yan H, Price C, Demetres M, Moser AB, Becker C, Hollandsworth K, Adang L, Vanderver A, Van Haren K, Ruzhnikov M, Kurtzberg J, Maegawa G, Orchard PJ, Lund TC, Raymond GV, Regelmann M, Orsini JJ, Seeger E, Kemp S, Eichler F, and Fatemi A
- Subjects
- Child, Child, Preschool, Consensus Development Conferences as Topic, Humans, Infant, Infant, Newborn, Male, Neonatal Screening methods, Adrenoleukodystrophy diagnosis, Magnetic Resonance Imaging
- Abstract
Background: Among boys with X-Linked adrenoleukodystrophy, a subset will develop childhood cerebral adrenoleukodystrophy (CCALD). CCALD is typically lethal without hematopoietic stem cell transplant before or soon after symptom onset. We sought to establish evidence-based guidelines detailing the neuroimaging surveillance of boys with neurologically asymptomatic adrenoleukodystrophy., Methods: To establish the most frequent age and diagnostic neuroimaging modality for CCALD, we completed a meta-analysis of relevant studies published between January 1, 1970 and September 10, 2019. We used the consensus development conference method to incorporate the resulting data into guidelines to inform the timing and techniques for neuroimaging surveillance. Final guideline agreement was defined as >80% consensus., Results: One hundred twenty-three studies met inclusion criteria yielding 1285 patients. The overall mean age of CCALD diagnosis is 7.91 years old. The median age of CCALD diagnosis calculated from individual patient data is 7.0 years old (IQR: 6.0-9.5, n = 349). Ninety percent of patients were diagnosed between 3 and 12. Conventional MRI was most frequently reported, comprised most often of T2-weighted and contrast-enhanced T1-weighted MRI. The expert panel achieved 95.7% consensus on the following surveillance parameters: (a) Obtain an MRI between 12 and 18 months old. (b) Obtain a second MRI 1 year after baseline. (c) Between 3 and 12 years old, obtain a contrast-enhanced MRI every 6 months. (d) After 12 years, obtain an annual MRI., Conclusion: Boys with adrenoleukodystrophy identified early in life should be monitored with serial brain MRIs during the period of highest risk for conversion to CCALD., (© 2020 SSIEM.)
- Published
- 2021
- Full Text
- View/download PDF
41. Aspartylglucosaminuria: Clinical Presentation and Potential Therapies.
- Author
-
Goodspeed K, Feng C, Laine M, and Lund TC
- Subjects
- Aspartylglucosaminuria therapy, Humans, Aspartylglucosaminuria diagnosis, Aspartylglucosaminuria physiopathology
- Abstract
Aspartylglucosaminuria (AGU) is a recessively inherited neurodegenerative lysosomal storage disease characterized by progressive intellectual disability, skeletal abnormalities, connective tissue overgrowth, gait disturbance, and seizures followed by premature death. AGU is caused by pathogenic variants in the aspartylglucosaminidase ( AGA ) gene, leading to glycoasparagine accumulation and cellular dysfunction. Although more prevalent in the Finnish population, more than 30 AGA variants have been identified worldwide. Owing to its rarity, AGU may be largely underdiagnosed. Recognition of the following early clinical features may aid in AGU diagnosis: developmental delays, hyperactivity, early growth spurt, inguinal and abdominal hernias, clumsiness, characteristic facial features, recurring upper respiratory and ear infections, tonsillectomy, multiple sets of tympanostomy tube placement, and sleep problems. Although no curative therapies currently exist, early diagnosis may provide benefit through the provision of anticipatory guidance, management of expectations, early interventions, and prophylaxis; it will also be crucial for increased clinical benefits of future AGU disease-modifying therapies.
- Published
- 2021
- Full Text
- View/download PDF
42. Mucopolysaccharidosis Type I: Current Treatments, Limitations, and Prospects for Improvement.
- Author
-
Hampe CS, Wesley J, Lund TC, Orchard PJ, Polgreen LE, Eisengart JB, McLoon LK, Cureoglu S, Schachern P, and McIvor RS
- Subjects
- Animals, Bone Diseases complications, Bone Diseases therapy, Cognition Disorders complications, Cognition Disorders therapy, Female, Glycosaminoglycans metabolism, Hearing Loss complications, Hearing Loss therapy, Heart Diseases complications, Heart Diseases therapy, Humans, Male, Range of Motion, Articular, Stem Cell Transplantation methods, Transplantation, Homologous, Enzyme Replacement Therapy methods, Hematopoietic Stem Cell Transplantation methods, Iduronidase biosynthesis, Mucopolysaccharidosis I physiopathology, Mucopolysaccharidosis I therapy
- Abstract
Mucopolysaccharidosis type I (MPS I) is a lysosomal disease, caused by a deficiency of the enzyme alpha-L-iduronidase (IDUA). IDUA catalyzes the degradation of the glycosaminoglycans dermatan and heparan sulfate (DS and HS, respectively). Lack of the enzyme leads to pathologic accumulation of undegraded HS and DS with subsequent disease manifestations in multiple organs. The disease can be divided into severe (Hurler syndrome) and attenuated (Hurler-Scheie, Scheie) forms. Currently approved treatments consist of enzyme replacement therapy (ERT) and/or hematopoietic stem cell transplantation (HSCT). Patients with attenuated disease are often treated with ERT alone, while the recommended therapy for patients with Hurler syndrome consists of HSCT. While these treatments significantly improve disease manifestations and prolong life, a considerable burden of disease remains. Notably, treatment can partially prevent, but not significantly improve, clinical manifestations, necessitating early diagnosis of disease and commencement of treatment. This review discusses these standard therapies and their impact on common disease manifestations in patients with MPS I. Where relevant, results of animal models of MPS I will be included. Finally, we highlight alternative and emerging treatments for the most common disease manifestations.
- Published
- 2021
- Full Text
- View/download PDF
43. Outcome After Cord Blood Transplantation Using Busulfan Pharmacokinetics-Targeted Myeloablative Conditioning for Hurler Syndrome.
- Author
-
Lum SH, Orchard PJ, Lund TC, Miller WP, Boelens JJ, and Wynn R
- Subjects
- Busulfan, Child, Humans, Retrospective Studies, Transplantation Conditioning, Cord Blood Stem Cell Transplantation, Mucopolysaccharidosis I therapy
- Abstract
We report the outcomes of cord blood transplantation (CBT) with a busulfan (Bu) pharmacokinetics-targeted myeloablative conditioning regimen in 97 children with Hurler syndrome (HS) performed between 2004 and 2016. The median age at CBT was 10.8 months (range, 0.23 to 63.2 months). The median duration of follow-up for surviving patients was 4.2 years (range, 1.0 to 12.8 years). Five-year overall survival (OS) and engrafted survival (ES) were 88% and 79%, respectively. OS was 95% in patients who received Bu/fludarabine (Flu)/antithymocyte globulin (ATG) conditioning, 90% in those who received Bu/cyclophosphamide (Cy)/ATG, and 74% in those who received Bu/Cy/alemtuzumab (P = .02). ES was 84% for recipients of Bu/Flu/ATG conditioning, 83% for recipients of Bu/Cy/ATG conditioning, and 65% for recipients of Bu/Cy/alemtuzumab conditioning (P = .34). Receipt of washed CB units (P = .03) and HLA matching ≤6/10 (P = .02) were associated with significantly lower ES. The 1-year cumulative incidence of graft failure was 11% (95% confidence interval, 6% to 21%). Five patients (5%) had grade III-IV acute GVHD, 5 patients had limited chronic GVHD, and 1 patient had extensive GVHD. The incidence of veno-occlusive disease was higher in patients conditioned with Bu/Cy compared with those conditioned with Bu/Flu (19% [n = 10] versus 5% [n = 2]: P = .03). Of the 11 patients with graft failure, 8 (73%) were aplastic, and 3 (27%) had autologous reconstitution. Of 11 patients with graft failure, 9 underwent a second CBT, and 8 (89%) survived. Full donor chimerism was observed in 89% patients after first CBT and in all patients after second CBT. Survival after CBT for HS has improved, but better strategies are still needed to improve graft outcomes., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
44. Biomarkers for prediction of skeletal disease progression in mucopolysaccharidosis type I.
- Author
-
Lund TC, Doherty TM, Eisengart JB, Freese RL, Rudser KD, Fung EB, Miller BS, White KK, Orchard PJ, Whitley CB, and Polgreen LE
- Abstract
Background: Orthopedic disease progresses in mucopolysaccharidosis type I (MPS I), even with approved therapies and remains a major factor in persistent suffering and disability. Novel therapies and accurate predictors of response are needed. The primary objective of this study was to identify surrogate biomarkers of future change in orthopedic disease., Methods: As part of a 9-year observational study of MPS I, range-of-motion (ROM), height, pelvic radiographs were measured annually. Biomarkers in year 1 were compared to healthy controls. Linear regression tested for associations of change in biomarkers over the first year with change in long-term outcomes., Results: MPS I participants (N = 19) were age 5 to 16 years and on average 6.9 ± 2.9 years post treatment initiation. Healthy controls (N = 51) were age 9 to 17 years. Plasma IL-1β, TNF-α, osteocalcin, pyridinolines, and deoxypyridinolines were higher in MPS than controls. Within MPS, progression of hip dysplasia was present in 46% to 77%. A 1 pg/mL increase in IL-6 was associated with -22°/year change in ROM (-28 to -15; P < .001), a 20 nmol/mmol creatinine/year increase in urine PYD was associated with a -0.024 Z-score/year change in height Z-score (-0.043 to -0.005; P = .016), and a 20 nmol/mmol creatinine/year increase in urine PYD was associated with a -2.0%/year change in hip dysplasia measured by Reimers migration index (-3.8 to -0.1; P = .037)., Conclusions: Inflammatory cytokines are high in MPS I. IL-6 and PYD were associated with progression in joint contracture, short stature, and hip dysplasia over time. Once validated, these biomarkers may prove useful for predicting response to treatment of skeletal disease in MPS I., Competing Interests: Dr T. C. L. receives research support from Sanofi‐Genzyme. Dr J. B. E. has received honoraria, consulting fees, and/or research support from ArmaGen, JCR pharmaceuticals, Orchard Therapeutics, Bluebird Bio, Shire/Takeda, and Sanofi‐Genzyme. Ms R. L. F. declares that she has no conflict of interest. Dr K. D. R. declares that he has no conflict of interest. Dr E. B. F. is a consultant for BioMarin Pharmaceuticals and Ascendis. Dr B. S. M. is a consultant for AbbVie, Ascendis, Ferring, Novo Nordisk, Pfizer, Sandoz, and Versartis and has received research support from Alexion, Ascendis, Endo Pharmaceuticals, Genentech, Genzyme, Novo Nordisk, Opko, Sandoz, Sangamo, Shire, Tolmar, and Versartis. Dr K. K. W. is a consultant for BioMarin Pharmaceuticals, has received honoraria from BioMarin Pharmaceuticals, Sanofi‐Genzyme, has grant funding from BioMarin Pharmaceuticals and Ultragenyx, and receives royalties from UpToDate.com. Dr C. B. W. is a consultant for Sanofi‐Genzyme. Dr P. J. O. receives research and clinical trial support from Sanofi‐Genzyme, Horizon, Magenta, and Bluebird Bio. Dr L. E. P. is a speaker and consultant for Sanofi‐Genzyme, has received research support from Sanofi‐Genzyme, Shire, BioMarin, and Pfizer‐Therachon, and is a consultant for Sangamo, Immusoft, Pfizer‐Therachon, and BioMarin., (© 2020 The Authors. JIMD Reports published by John Wiley & Sons Ltd on behalf of SSIEM.)
- Published
- 2020
- Full Text
- View/download PDF
45. Inappropriate cathepsin K secretion promotes its enzymatic activation driving heart and valve malformation.
- Author
-
Lu PN, Moreland T, Christian CJ, Lund TC, Steet RA, and Flanagan-Steet H
- Subjects
- Animals, Disease Models, Animal, Enzyme Activation genetics, Genetic Predisposition to Disease, Heart physiopathology, Heart Defects, Congenital physiopathology, Heart Valves growth & development, Humans, Lysosomal Storage Diseases genetics, Lysosomal Storage Diseases physiopathology, Mucolipidoses physiopathology, Mutation, Transforming Growth Factor beta genetics, Zebrafish genetics, Cathepsin K genetics, Heart growth & development, Heart Defects, Congenital genetics, Mucolipidoses genetics, Transferases (Other Substituted Phosphate Groups) genetics
- Abstract
Although congenital heart defects (CHDs) represent the most common birth defect, a comprehensive understanding of disease etiology remains unknown. This is further complicated since CHDs can occur in isolation or as a feature of another disorder. Analyzing disorders with associated CHDs provides a powerful platform to identify primary pathogenic mechanisms driving disease. Aberrant localization and expression of cathepsin proteases can perpetuate later-stage heart diseases, but their contribution toward CHDs is unclear. To investigate the contribution of cathepsins during cardiovascular development and congenital disease, we analyzed the pathogenesis of cardiac defects in zebrafish models of the lysosomal storage disorder mucolipidosis II (MLII). MLII is caused by mutations in the GlcNAc-1-phosphotransferase enzyme (Gnptab) that disrupt carbohydrate-dependent sorting of lysosomal enzymes. Without Gnptab, lysosomal hydrolases, including cathepsin proteases, are inappropriately secreted. Analyses of heart development in gnptab-deficient zebrafish show cathepsin K secretion increases its activity, disrupts TGF-β-related signaling, and alters myocardial and valvular formation. Importantly, cathepsin K inhibition restored normal heart and valve development in MLII embryos. Collectively, these data identify mislocalized cathepsin K as an initiator of cardiac disease in this lysosomal disorder and establish cathepsin inhibition as a viable therapeutic strategy.
- Published
- 2020
- Full Text
- View/download PDF
46. Volume of Gadolinium Enhancement and Successful Repair of the Blood-Brain Barrier in Cerebral Adrenoleukodystrophy.
- Author
-
Lund TC, Ng M, Orchard PJ, Loes DJ, Raymond GV, Gupta A, Kenny-Jung D, and Nascene DR
- Subjects
- Blood-Brain Barrier, Contrast Media, Gadolinium, Humans, Magnetic Resonance Imaging, Male, Adrenoleukodystrophy diagnostic imaging, Adrenoleukodystrophy therapy, Hematopoietic Stem Cell Transplantation
- Abstract
Up to 40% of boys with adrenoleukodystrophy develop a severe central nervous system demyelinating form (cALD) characterized by white matter changes and gadolinium enhancement on magnetic resonance imaging (MRI). Hematopoietic cell transplant (HCT) is the only proven means to attenuate cALD progression. The elimination of active neuroinflammation is indicated radiographically by the resolution of gadolinium (Gd) enhancement and correlates to speed of donor neutrophil recovery. We analyzed 66 boys with cALD undergoing HCT for biomarkers correlating with early (30 days post-HCT) Gd signal resolution. We found that log Gd volume (cm
3 ) on pre-HCT MRI strongly positively correlated to day 30 Gd resolution (P = .0003) with smaller volume correlating to higher proportion resolved, as was the baseline gadolinium intensity score (P = .04), plasma chitotriosidase activity (P = .04), and faster absolute neutrophil count recovery (P = .03). In multivariate analysis, log Gd volume remained superior in determining which patients would have Gd signal resolution by 30 days post-HCT (P = .016). A final analysis indicated that early Gd resolution also correlated with less neurologic progression from baseline to 1 year following HCT (P = .006). MRI Gd volume may serve as a contributing biomarker to better delineate outcomes and an important metric in comparing therapies in the treatment of cALD., (Copyright © 2020 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
47. Mucopolysaccharidosis Type I: A Review of the Natural History and Molecular Pathology.
- Author
-
Hampe CS, Eisengart JB, Lund TC, Orchard PJ, Swietlicka M, Wesley J, and McIvor RS
- Subjects
- Animals, Disease Models, Animal, Early Diagnosis, Humans, Iduronidase deficiency, Iduronidase genetics, Mucopolysaccharidosis I genetics, Mucopolysaccharidosis I physiopathology, Phenotype, Mucopolysaccharidosis I diagnosis, Mucopolysaccharidosis I pathology
- Abstract
Mucopolysaccharidosis type I (MPS I) is a rare autosomal recessive inherited disease, caused by deficiency of the enzyme α-L-iduronidase, resulting in accumulation of the glycosaminoglycans (GAGs) dermatan and heparan sulfate in organs and tissues. If untreated, patients with the severe phenotype die within the first decade of life. Early diagnosis is crucial to prevent the development of fatal disease manifestations, prominently cardiac and respiratory disease, as well as cognitive impairment. However, the initial symptoms are nonspecific and impede early diagnosis. This review discusses common phenotypic manifestations in the order in which they develop. Similarities and differences in the three animal models for MPS I are highlighted. Earliest symptoms, which present during the first 6 months of life, include hernias, coarse facial features, recurrent rhinitis and/or upper airway obstructions in the absence of infection, and thoracolumbar kyphosis. During the next 6 months, loss of hearing, corneal clouding, and further musculoskeletal dysplasias develop. Finally, late manifestations including lower airway obstructions and cognitive decline emerge. Cardiac symptoms are common in MPS I and can develop in infancy. The underlying pathogenesis is in the intra- and extracellular accumulation of partially degraded GAGs and infiltration of cells with enlarged lysosomes causing tissue expansion and bone deformities. These interfere with the proper arrangement of collagen fibrils, disrupt nerve fibers, and cause devastating secondary pathophysiological cascades including inflammation, oxidative stress, and other disruptions to intracellular and extracellular homeostasis. A greater understanding of the natural history of MPS I will allow early diagnosis and timely management of the disease facilitating better treatment outcomes.
- Published
- 2020
- Full Text
- View/download PDF
48. Neurocognitive benchmarks following transplant for emerging cerebral adrenoleukodystrophy.
- Author
-
Pierpont EI, Nascene DR, Shanley R, Kenney-Jung DL, Ziegler RS, Miller WP, Gupta AO, Lund TC, Orchard PJ, and Eisengart JB
- Subjects
- Adolescent, Adrenoleukodystrophy diagnosis, Child, Child, Preschool, Early Diagnosis, Humans, Infant, Newborn, Magnetic Resonance Imaging, Male, Neonatal Screening methods, Treatment Outcome, Adrenoleukodystrophy pathology, Adrenoleukodystrophy therapy, Benchmarking, Hematopoietic Stem Cell Transplantation methods
- Abstract
Objective: To quantify benchmark treatment outcomes that may be enabled by newborn screening surveillance for X-linked adrenoleukodystrophy (ALD), we report neurocognitive, neuropsychiatric, and MRI change for boys who underwent hematopoietic stem cell transplant (HSCT) at initial stages of demyelination, prior to neurocognitive signs of disease., Methods: Retrospective chart review identified 36 patients whose cerebral ALD was detected and treated early, with lesion severity less than 5 on the ALD-specific MRI scoring system. Median age at transplant was 7.3 years (range, 4.0-16.1). Progression of radiologic disease on MRI in the 2 years following HSCT was examined relative to the severity of the initial lesion for 33 patients, and longitudinal neurocognitive and neuropsychiatric outcomes were studied for 30 patients., Results: Patients whose pretransplant lesion extended beyond the splenium of the corpus callosum and adjacent periventricular white matter (MRI severity score >2) demonstrated lower posttransplant neurocognitive scores, more neuropsychiatric symptoms, and more disease progression on MRI than patients with a less severe lesion. Changes from baseline neurocognitive functioning were greater at 2 years posttransplant as compared to 1 year. There was greater variance and risk of lesion progression as pretransplant MRI severity increased., Conclusion: To realize the full benefits of newborn screening, clinicians must detect very small demyelinating lesions during surveillance and intervene quickly. Novel interventions that reduce risks inherent in allogeneic transplantation are needed. Trial endpoints should include direct neurocognitive assessment and extend at least 2 years posttreatment to provide the greatest sensitivity to detect neurocognitive morbidity., (Copyright © 2020 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.)
- Published
- 2020
- Full Text
- View/download PDF
49. Dysostosis Multiplex in Human Mucopolysaccharidosis Type 1 H and in Animal Models of the Disease.
- Author
-
Hampe CS, Polgreen LE, Lund TC, and McIvor RS
- Subjects
- Animals, Disease Models, Animal, Dogs, Humans, Iduronidase, Retrospective Studies, Dysostoses, Mucopolysaccharidosis I
- Abstract
Mucopolysaccharidosis type I (MPS I) is a rare autosomal recessive disorder, caused by deficiency of α-L-iduronidase, and consequent accumulation of dermatan and heparan sulfates. Severity of the disease ranges from mild (Scheie) to moderate (Hurler-Scheie) to severe (Hurler or MPS-IH). A prominent clinical manifestation of MPS-IH is dysostosis multiplex, a constellation of skeletal abnormalities. We performed a retrospective review comparing manifestations of dysostosis multiplex in patients presenting with MPSIH and relevant animal models. Dog, cat and mouse models of MPS-IH are extensively studied to better understand the pathology of the disease. While all animal models display certain characteristics of human MPSIH, species-specific manifestations must be considered when evaluating skeletal abnormalities. Moreover, some skeletal abnormalities emerge at species-specific developmental stages, e.g. thoracolumbar kyphosis is an early manifestation in humans, while it appears late in the mouse model. The choice of the appropriate diagnostic test is of importance to avoid misleading conclusions., (Copyright© of YS Medical Media ltd.)
- Published
- 2020
- Full Text
- View/download PDF
50. The Role of Hematopoietic Cell Transplant in the Glycoprotein Diseases.
- Author
-
Naumchik BM, Gupta A, Flanagan-Steet H, Steet RA, Cathey SS, Orchard PJ, and Lund TC
- Subjects
- Animals, Enzyme Replacement Therapy, Humans, Glycoproteins metabolism, Hematopoietic Stem Cell Transplantation, Lysosomal Storage Diseases therapy
- Abstract
The glycoprotein disorders are a group of lysosomal storage diseases (α-mannosidosis, aspartylglucosaminuria, β-mannosidosis, fucosidosis, galactosialidosis, sialidosis, mucolipidosis II, mucolipidosis III, and Schindler Disease) characterized by specific lysosomal enzyme defects and resultant buildup of undegraded glycoprotein substrates. This buildup causes a multitude of abnormalities in patients including skeletal dysplasia, inflammation, ocular abnormalities, liver and spleen enlargement, myoclonus, ataxia, psychomotor delay, and mild to severe neurodegeneration. Pharmacological treatment options exist through enzyme replacement therapy (ERT) for a few, but therapies for this group of disorders is largely lacking. Hematopoietic cell transplant (HCT) has been explored as a potential therapeutic option for many of these disorders, as HCT introduces functional enzyme-producing cells into the bone marrow and blood along with the engraftment of healthy donor cells in the central nervous system (presumably as brain macrophages or a type of microglial cell). The outcome of HCT varies widely by disease type. We report our institutional experience with HCT as well as a review of the literature to better understand HCT and outcomes for the glycoprotein disorders.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.