Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Patricia Dickson is active.

Publication


Featured researches published by Patricia Dickson.


American Journal of Medical Genetics Part A | 2008

Intrathecal enzyme replacement therapy in a patient with mucopolysaccharidosis type I and symptomatic spinal cord compression

Maria-Veronica Muñoz-Rojas; Taiane Alves Vieira; R.G. Costa; Simone Fagondes; Angela Beatriz John; Laura Bannach Jardim; Leonardo Modesti Vedolin; Marcia Mocellin Raymundo; Patricia Dickson; Emil D. Kakkis; Roberto Giugliani

In mucopolysaccharidosis I, deficiency of α‐L‐iduronidase can cause spinal cord compression (SCC) due to storage of glycosaminoglycans (GAGs) within the cervical meninges. As intravenous enzyme replacement therapy (ERT) is not likely to provide enzyme across the blood–brain barrier, standard treatment for this complication is usually surgical, which has a high morbidity and mortality risk. We report on the use of intrathecal (IT) laronidase in a MPS I patient with SCC who refused the surgical treatment. Assessments were performed at baseline, with clinical and biochemical evaluations, 4‐extremity somatosensory evoked potentials, 12 min walk test and MRI studies of the CNS. Changes on these parameters were evaluated after 4 IT infusions of laronidase administered monthly via lumbar puncture. To our knowledge, this was the first MPS patient who received IT ERT. No major adverse events were observed. There were no clinically significant changes in serum chemistries. CSF GAG results revealed pretreatment values slightly above normal standards: 13.3 mg/L (NV < 12 mg/L) which after IT laronidase infusions were within normal levels (10.3 mg/L). 12MWT presented a 14% improvement, with better performance on stability and gait control. Maximum voluntary ventilation showed 55.6% improvement considering the percentage of predicted (26.7% at baseline compared to 41.9%); Maximum Inspiration Pressure improved 36.6% of predicted (26.8% at baseline to 36.7%); Pulmonary diffusion improved 17.6% of predicted %. In conclusion, although the improvement observed in this case with IT laronidase should be confirmed in further patients, this procedure seems to be a safe treatment for SCC in MPS I.


Journal of Clinical Investigation | 2008

Immune tolerance improves the efficacy of enzyme replacement therapy in canine mucopolysaccharidosis I

Patricia Dickson; Maryn Peinovich; Michael F. McEntee; Thomas Lester; Steven Q. Le; Aimee Krieger; Hayden Manuel; Catherine Jabagat; Merry Passage; Emil D. Kakkis

Mucopolysaccharidoses (MPSs) are lysosomal storage diseases caused by a deficit in the enzymes needed for glycosaminoglycan (GAG) degradation. Enzyme replacement therapy with recombinant human alpha-L-iduronidase successfully reduces lysosomal storage in canines and humans with iduronidase-deficient MPS I, but therapy usually also induces antibodies specific for the recombinant enzyme that could reduce its efficacy. To understand the potential impact of alpha-L-iduronidase-specific antibodies, we studied whether inducing antigen-specific immune tolerance to iduronidase could improve the effectiveness of recombinant iduronidase treatment in canines. A total of 24 canines with MPS I were either tolerized to iduronidase or left nontolerant. All canines received i.v. recombinant iduronidase at the FDA-approved human dose or a higher dose for 9-44 weeks. Nontolerized canines developed iduronidase-specific antibodies that proportionally reduced in vitro iduronidase uptake. Immune-tolerized canines achieved increased tissue enzyme levels at either dose in most nonreticular tissues and a greater reduction in tissue GAG levels, lysosomal pathology, and urinary GAG excretion. Tolerized MPS I dogs treated with the higher dose received some further benefit in the reduction of GAGs in tissues, urine, and the heart valve. Therefore, immune tolerance to iduronidase improved the efficacy of enzyme replacement therapy with recombinant iduronidase in canine MPS I and could potentially improve outcomes in patients with MPS I and other lysosomal storage diseases.


Journal of Medical Genetics | 2005

ETHE1 mutations are specific to ethylmalonic encephalopathy

Valeria Tiranti; Egill Briem; Eleonora Lamantea; Rossana Mineri; Elena Papaleo; L De Gioia; F Forlani; Piero Rinaldo; Patricia Dickson; B Abu-Libdeh; L Cindro-Heberle; M Owaidha; R M Jack; E Christensen; Alberto Burlina; Massimo Zeviani

Mutations in ETHE1, a gene located at chromosome 19q13, have recently been identified in patients affected by ethylmalonic encephalopathy (EE). EE is a devastating infantile metabolic disorder, characterised by widespread lesions in the brain, hyperlactic acidaemia, petechiae, orthostatic acrocyanosis, and high levels of ethylmalonic acid in body fluids. To investigate to what extent ETHE1 is responsible for EE, we analysed this gene in 29 patients with typical EE and in 11 patients presenting with early onset progressive encephalopathy with ethylmalonic aciduria (non-EE EMA). Frameshift, stop, splice site, and missense mutations of ETHE1 were detected in all the typical EE patients analysed. Western blot analysis of the ETHE1 protein indicated that some of the missense mutations are associated with the presence of the protein, suggesting that the corresponding wild type amino acid residues have a catalytic function. No ETHE1 mutations were identified in non-EE EMA patients. Experiments based on two dimensional blue native electrophoresis indicated that ETHE1 protein works as a supramolecular, presumably homodimeric, complex, and a three dimensional model of the protein suggests that it is likely to be a mitochondrial matrix thioesterase acting on a still unknown substrate. Finally, the 625G→A single nucleotide polymorphism in the gene encoding the short chain acyl-coenzyme A dehydrogenase (SCAD) was previously proposed as a co-factor in the aetiology of EE and other EMA syndromes. SNP analysis in our patients ruled out a pathogenic role of SCAD variants in EE, but did show a highly significant prevalence of the 625A alleles in non-EE EMA patients.


Science Translational Medicine | 2010

Replacing the Enzyme α-l-Iduronidase at Birth Ameliorates Symptoms in the Brain and Periphery of Dogs with Mucopolysaccharidosis Type I

Ashley Dierenfeld; Michael F. McEntee; Carole Vogler; Charles H. Vite; Agnes H. Chen; Merry Passage; Steven Q. Le; S. Shah; Jacqueline K. Jens; Elizabeth M. Snella; K.L. Kline; J.D. Parkes; Wendy A. Ware; L.E. Moran; A. J. Fales-Williams; J.A. Wengert; R.D. Whitley; D.M. Betts; A.M. Boal; E.A. Riedesel; William Gross; N.M. Ellinwood; Patricia Dickson

Replacing the enzyme α-l-iduronidase at birth ameliorates symptoms in the brain and periphery of dogs with mucopolysaccharidosis type I. When Dogs Really Are Man’s Best Friend For certain diseases, dogs provide an excellent large-animal model and the lysosomal storage disorder mucopolysaccharidosis type I is no exception. In this disease, a defect in the enzyme α-l-iduronidase prevents breakdown of glycosaminoglycans, resulting in their accumulation in the lysosomes of cells, leading to engorged and dysfunctional cells. A variety of serious complications ensue such as enlarged organs, skeletal defects, corneal clouding, abnormal heart valves, and cognitive deficits. Although a human recombinant form of the enzyme has proved successful in treating patients with less severe forms of the disease, only some of the symptoms are ameliorated and patients often develop antibodies to the enzyme. Dierenfeld and colleagues reasoned that starting enzyme replacement therapy at birth could halt or even reverse the more serious disease symptoms such as heart valve defects, skeletal deformities, and cognitive impairment and might prevent antibodies from forming against human recombinant α-l-iduronidase. Working in a naturally occurring dog model of mucopolysaccharidosis type I in which the animals have a defective α-l-iduronidase enzyme and show most of the symptoms of the human disease, Dierenfeld et al. administered two different doses of α-l-iduronidase (0.58 and 1.57 mg/kg) intravenously every week starting a few days after birth. The authors show that starting enzyme replacement therapy at such a young age prevented the formation of antibodies against the enzyme due to neonatal immune tolerance. Treated dogs showed marked reductions in glycosaminoglycans in a variety of different tissues including the mitral heart valve, which has been refractory to traditional enzyme treatment administered at later ages. Furthermore, treated dogs showed relatively normal skeletons and lacked the stiff gait, lax joints, upturned nose, and poor neck flexibility exhibited by untreated animals with the disease. Compellingly, glycosaminoglycan levels and cortical atrophy in the brain decreased and corneal clouding improved slightly (at the higher dose) in the treated dogs. These results show that starting intravenous administration of α-l-iduronidase at higher doses and as soon as possible after birth increases the efficacy of enzyme replacement therapy and ensures immune tolerance to the enzyme. These promising results in man’s best friend suggest that children with severe mucopolysaccharidosis type I or other early-onset lysosomal storage diseases should start enzyme replacement therapy as soon as possible after birth. Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disease caused by loss of activity of α-l-iduronidase and attendant accumulation of the glycosaminoglycans dermatan sulfate and heparan sulfate. Current treatments are suboptimal and do not address residual disease including corneal clouding, skeletal deformities, valvular heart disease, and cognitive impairment. We treated neonatal dogs with MPS I with intravenous recombinant α-l-iduronidase replacement therapy at the conventional 0.58 mg/kg or a higher 1.57 mg/kg weekly dose for 56 to 81 weeks. In contrast to previous results in animals and patients treated at a later age, the dogs failed to mount an antibody response to enzyme therapy, consistent with the induction of immune tolerance in neonates. The higher dose of enzyme led to complete normalization of lysosomal storage in the liver, spleen, lung, kidney, synovium, and myocardium, as well as in the hard-to-treat mitral valve. Cardiac biochemistry and function were restored, and there were improvements in skeletal disease as shown by clinical and radiographic assessments. Glycosaminoglycan levels in the brain were normalized after intravenous enzyme therapy, in the presence or absence of intrathecal administration of recombinant α-l-iduronidase. Histopathological evidence of glycosaminoglycan storage in the brain was ameliorated with the higher-dose intravenous therapy and was further improved by combining intravenous and intrathecal therapy. These findings argue that neonatal testing and early treatment of patients with MPS I may more effectively treat this disease.


Pharmacology & Therapeutics | 2014

Intrathecal delivery of protein therapeutics to the brain: a critical reassessment.

Pericles Calias; William A. Banks; David J. Begley; Maurizio Scarpa; Patricia Dickson

Disorders of the central nervous system (CNS), including stroke, neurodegenerative diseases, and brain tumors, are the worlds leading causes of disability. Delivery of drugs to the CNS is complicated by the blood-brain barriers that protect the brain from the unregulated leakage and entry of substances, including proteins, from the blood. Yet proteins represent one of the most promising classes of therapeutics for the treatment of CNS diseases. Many strategies for overcoming these obstacles are in development, but the relatively straightforward approach of bypassing these barriers through direct intrathecal administration has been largely overlooked. Originally discounted because of its lack of usefulness for delivering small, lipid-soluble drugs to the brain, the intrathecal route has emerged as a useful, in some cases perhaps the ideal, route of administration for certain therapeutic protein and targeted disease combinations. Here, we review blood-brain barrier functions and cerebrospinal fluid dynamics and their relevance to drug delivery via the intrathecal route, discuss animal and human studies that have investigated intrathecal delivery of protein therapeutics, and outline several characteristics of protein therapeutics that can allow them to be successfully delivered intrathecally.


Molecular Genetics and Metabolism | 2010

Early versus late treatment of spinal cord compression with long-term intrathecal enzyme replacement therapy in canine mucopolysaccharidosis type I

Patricia Dickson; Stephen Hanson; Michael F. McEntee; Charles H. Vite; Carole Vogler; Anton Mlikotic; Agnes H. Chen; Katherine P. Ponder; Mark E. Haskins; Brigette L. Tippin; Steven Q. Le; Merry Passage; Catalina Guerra; Ashley Dierenfeld; Jackie K. Jens; Elizabeth M. Snella; Shih-hsin Kan; N. Matthew Ellinwood

Enzyme replacement therapy (ERT) with intravenous recombinant human alpha-l-iduronidase (IV rhIDU) is a treatment for patients with mucopolysaccharidosis I (MPS I). Spinal cord compression develops in MPS I patients due in part to dural and leptomeningeal thickening from accumulated glycosaminoglycans (GAG). We tested long-term and every 3-month intrathecal (IT) and weekly IV rhIDU in MPS I dogs age 12-15months (Adult) and MPS I pups age 2-23days (Early) to determine whether spinal cord compression could be reversed, stabilized, or prevented. Five treatment groups of MPS I dogs were evaluated (n=4 per group): IT+IV Adult, IV Adult, IT + IV Early, 0.58mg/kg IV Early and 1.57mg/kg IV Early. IT + IV rhIDU (Adult and Early) led to very high iduronidase levels in cervical, thoracic, and lumber spinal meninges (3600-29,000% of normal), while IV rhIDU alone (Adult and Early) led to levels that were 8.2-176% of normal. GAG storage was significantly reduced from untreated levels in spinal meninges of IT + IV Early (p<.001), IT+IV Adult (p=.001), 0.58mg/kg IV Early (p=.002) and 1.57mg/kg IV Early (p<.001) treatment groups. Treatment of dogs shortly after birth with IT+IV rhIDU (IT + IV Early) led to normal to near-normal GAG levels in the meninges and histologic absence of storage vacuoles. Lysosomal storage was reduced in spinal anterior horn cells in 1.57mg/kg IV Early and IT + IV Early animals. All dogs in IT + IV Adult and IV Adult groups had compression of their spinal cord at 12-15months of age determined by magnetic resonance imaging and was due to protrusion of spinal disks into the canal. Cord compression developed in 3 of 4 dogs in the 0.58mg/kg IV Early group; 2 of 3 dogs in the IT + IV Early group; and 0 of 4 dogs in the 1.57mg/kg IV Early group by 12-18months of age. IT + IV rhIDU was more effective than IV rhIDU alone for treatment of meningeal storage, and it prevented meningeal GAG accumulation when begun early. High-dose IV rhIDU from birth (1.57mg/kg weekly) appeared to prevent cord compression due to protrusion of spinal disks.


Molecular Genetics and Metabolism | 2015

Intrathecal 2-hydroxypropyl-beta-cyclodextrin in a single patient with Niemann-Pick C1

Timothy Maarup; Agnes H. Chen; Forbes D. Porter; Nicole Y. Farhat; Daniel S. Ory; Rohini Sidhu; Xuntian Jiang; Patricia Dickson

Niemann-Pick C, type 1 (NPC1) is a progressive autosomal recessive neurologic disease caused by defective intracellular cholesterol and lipid trafficking. There are currently no United States Food and Drug Administration approved treatments for NPC1. We undertook a study evaluating the safety, efficacy, and biomarker response of intrathecal 2-hydroxypropyl-β-cyclodextrin (HP-β-CD) in a 12-year old subject with mildly symptomatic NPC. The subject received 200mg intrathecal HP-β-CD administered biweekly via lumbar puncture. To date the subject has received 27 intrathecal HP-β-CD injections. Intrathecal HP-β-CD has been generally safe and well tolerated in this subject. There has been an improvement in vertical gaze. The subject has developed subclinical hearing loss at high frequency that is likely HP-β-CD related. Plasma 24-(S)-hydroxycholesterol, a pharmacodynamic biomarker for cholesterol redistribution in the central nervous system, was significantly increased in response to each of the first 5 drug administrations. Further dosing as well as dose escalations are needed to more completely ascertain the safety and efficacy of intrathecal HP-β-CD.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Delivery of an enzyme-IGFII fusion protein to the mouse brain is therapeutic for mucopolysaccharidosis type IIIB

Shih-hsin Kan; Mika Aoyagi-Scharber; Steven Q. Le; Jon Vincelette; Kazuhiro Ohmi; Sherry Bullens; Daniel J. Wendt; Terri Christianson; Pascale M.N. Tiger; Jillian R. Brown; Roger Lawrence; Bryan K. Yip; John Holtzinger; Anil Bagri; Danielle Crippen-Harmon; Kristen N. Vondrak; Zhi Chen; Chuck Hague; Josh Woloszynek; Diana S. Cheung; Katherine A. Webster; Evan G. Adintori; Melanie J. Lo; Wesley P. Wong; Paul A. Fitzpatrick; Jonathan H. LeBowitz; Brett E. Crawford; Stuart Bunting; Patricia Dickson; Elizabeth F. Neufeld

Significance Mucopolysaccharidosis type IIIB (MPS IIIB) is a devastating and currently untreatable disease affecting mainly the brain. The cause is lack of the lysosomal enzyme, α–N-acetylglucosaminidase (NAGLU), and storage of heparan sulfate. Using a mouse model of MPS IIIB, we administered a modified NAGLU by injection into the left ventricle of the brain, bypassing the blood–brain barrier. The modification consisted of a fragment of IGFII, which allows receptor-mediated uptake and delivery to lysosomes. The modified enzyme was taken up avidly by cells in both brain and liver, where it reduced pathological accumulation of heparan sulfate and other metabolites to normal or near-normal levels. The results suggest the possibility of treatment for MPS IIIB. Mucopolysaccharidosis type IIIB (MPS IIIB, Sanfilippo syndrome type B) is a lysosomal storage disease characterized by profound intellectual disability, dementia, and a lifespan of about two decades. The cause is mutation in the gene encoding α–N-acetylglucosaminidase (NAGLU), deficiency of NAGLU, and accumulation of heparan sulfate. Impediments to enzyme replacement therapy are the absence of mannose 6-phosphate on recombinant human NAGLU and the blood–brain barrier. To overcome the first impediment, a fusion protein of recombinant NAGLU and a fragment of insulin-like growth factor II (IGFII) was prepared for endocytosis by the mannose 6-phosphate/IGFII receptor. To bypass the blood–brain barrier, the fusion protein (“enzyme”) in artificial cerebrospinal fluid (“vehicle”) was administered intracerebroventricularly to the brain of adult MPS IIIB mice, four times over 2 wk. The brains were analyzed 1–28 d later and compared with brains of MPS IIIB mice that received vehicle alone or control (heterozygous) mice that received vehicle. There was marked uptake of the administered enzyme in many parts of the brain, where it persisted with a half-life of approximately 10 d. Heparan sulfate, and especially disease-specific heparan sulfate, was reduced to control level. A number of secondary accumulations in neurons [β-hexosaminidase, LAMP1(lysosome-associated membrane protein 1), SCMAS (subunit c of mitochondrial ATP synthase), glypican 5, β-amyloid, P-tau] were reduced almost to control level. CD68, a microglial protein, was reduced halfway. A large amount of enzyme also appeared in liver cells, where it reduced heparan sulfate and β-hexosaminidase accumulation to control levels. These results suggest the feasibility of enzyme replacement therapy for MPS IIIB.


Molecular Genetics and Metabolism | 2014

Glycan-based biomarkers for mucopolysaccharidoses

Roger Lawrence; Jillian R. Brown; Fred Lorey; Patricia Dickson; Brett E. Crawford; Jeffrey D. Esko

The mucopolysaccharidoses (MPS) result from attenuation or loss of enzyme activities required for lysosomal degradation of the glycosaminoglycans, hyaluronan, heparan sulfate, chondroitin/dermatan sulfate, and keratan sulfate. This review provides a summary of glycan biomarkers that have been used to characterize animal models of MPS, for diagnosis of patients, and for monitoring therapy based on hematopoietic stem cell transplantation and enzyme replacement therapy. Recent advances have focused on the non-reducing terminus of the glycosaminoglycans that accumulate as biomarkers, using a combination of enzymatic digestion with bacterial enzymes followed by quantitative liquid chromatography/mass spectrometry. These new methods provide a simple, rapid diagnostic strategy that can be applied to samples of urine, blood, cerebrospinal fluid, cultured cells and dried blood spots from newborn infants. Analysis of the non-reducing end glycans provides a method for monitoring enzyme replacement and substrate reduction therapies and serves as a discovery tool for uncovering novel biomarkers and new forms of mucopolysaccharidoses.


Current Pharmaceutical Biotechnology | 2011

Intrathecal Enzyme Replacement Therapy for Mucopolysaccharidosis I: Translating Success in Animal Models to Patients

Patricia Dickson; Agnes H. Chen

Intrathecal enzyme replacement therapy has been proposed to treat central nervous system (CNS) disease due to mucopolysaccharidosis type I. Our research has shown that repeated injections of recombinant enzyme into the spinal fluid corrects enzyme deficiency and normalizes lysosomal storage in the canine model. The challenge is to translate the success in the animal where there are fewer study limitations to human patients where studies are more restricted. This review will explore what is known about the measurement of clinically-relevant outcomes of intrathecal enzyme replacement therapy for MPS I (including ongoing clinical trials), the challenges in translating therapies for the CNS in rare diseases, and new outcome measures that could aid translation of CNS therapies for MPS disorders.

Collaboration


Dive into the Patricia Dickson's collaboration.

Top Co-Authors

Avatar

Steven Q. Le

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Shih-hsin Kan

University of California

View shared research outputs
Top Co-Authors

Avatar

Agnes H. Chen

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Merry Passage

University of California

View shared research outputs
Top Co-Authors

Avatar

Elsa Shapiro

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Moin U. Vera

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Raymond Y. Wang

Children's Hospital of Orange County

View shared research outputs
Researchain Logo
Decentralizing Knowledge