1
|
Kose S, Cinar E, Akyel H, Cakir-Aktas C, Tel BC, Karatas H, Kelicen-Ugur P. Cerliponase alfa decreases Aβ load and alters autophagy- related pathways in mouse hippocampal neurons exposed to fAβ 1-42. Life Sci 2024; 357:123105. [PMID: 39362589 DOI: 10.1016/j.lfs.2024.123105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/30/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Extracellular aggregation of amyloid-beta (Aβ) in the brain plays a central role in the onset and progression of Alzheimer's disease (AD). Moreover, intraneuronal accumulation of Aβ via oligomer internalization might play an important role in the progression of AD. Deficient autophagy, which is a lysosomal degradation process, occurs during the early stages of AD. Tripeptidyl peptidase-1 (TPP1) functions as a lysosomal enzyme, and TPP1 gene mutations are associated with type 2 late infantile neuronal ceroid lipofuscinosis (LINCL). Nevertheless, there is little information about the role of TPP1 in the pathogenesis of AD; therefore, the present study aimed to measure the decrease in intraneuronal Aβ accumulation by a recombinant analog of the TPP1 enzyme, cerliponase alfa (CER) (Brineura®), and to determine whether autophagy pathways play a role in this decrease. In this study, endogenous Aβ accumulation was induced by fAβ1-42 (a toxic fragment of full-length Aβ) exposure, and mouse hippocampal neuronal cells (HT-22) were treated with CER (human recombinant rhTPP1 1 mg mL-1). Soluble Aβ, TPP1, and the proteins involved in autophagy, including mammalian target of rapamycin (p-mTOR/mTOR), p62/sequestosome-1 (p62/SQSTM1), and microtubule-associated protein 1 A/1B-light chain 3 (LC3), were evaluated using western blotting. The sirtuin-1, beclin-1, and Atg5 genes were also studied using RT-PCR. Aβ and TPP1 localizations were observed via immunocytochemistry. CER reduced the Aβ load in HT-22 cells by inducing TPP1 expression and converting pro-TPP1 into the mature form. Furthermore, exposure to CER and fAβ1-42 induced the autophagy-regulatory/related pathways in HT-22 cells and exposure to CER alone increased sirtuin-1 activity. Based on the present findings, we suggest that augmentation of TPP1 with enzyme replacement therapy may be a potential therapeutic option for the treatment of AD.
Collapse
Affiliation(s)
- Selma Kose
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Sihhiye, Ankara, Turkiye
| | - Elif Cinar
- Istanbul University-Cerrahpasa, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkiye.
| | - Hilal Akyel
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Sihhiye, Ankara, Turkiye; Baskent University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkiye
| | - Canan Cakir-Aktas
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Sihhiye, Ankara, Turkiye.
| | - Banu Cahide Tel
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Sihhiye, Ankara, Turkiye.
| | - Hulya Karatas
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Sihhiye, Ankara, Turkiye.
| | - Pelin Kelicen-Ugur
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Sihhiye, Ankara, Turkiye.
| |
Collapse
|
2
|
Bellamy KKL, Skedsmo FS, Hultman J, Jansen JH, Lingaas F. Neuronal ceroid lipofuscinosis in a Schapendoes dog is caused by a missense variant in CLN6. Anim Genet 2024; 55:612-620. [PMID: 38866396 DOI: 10.1111/age.13457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/03/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
Neuronal ceroid lipofuscinosis (NCL) is a group of neurodegenerative disorders that occur in humans, dogs, and several other species. NCL is characterised clinically by progressive deterioration of cognitive and motor function, epileptic seizures, and visual impairment. Most forms present early in life and eventually lead to premature death. Typical pathological changes include neuronal accumulation of autofluorescent, periodic acid-Schiff- and Sudan black B-positive lipopigments, as well as marked loss of neurons in the central nervous system. Here, we describe a 19-month-old Schapendoes dog, where clinical signs were indicative of lysosomal storage disease, which was corroborated by pathological findings consistent with NCL. Whole genome sequencing of the affected dog and both parents, followed by variant calling and visual inspection of known NCL genes, identified a missense variant in CLN6 (c.386T>C). The variant is located in a highly conserved region of the gene and predicted to be harmful, which supports a causal relationship. The identification of this novel CLN6 variant enables pre-breeding DNA-testing to prevent future cases of NCL6 in the Schapendoes breed, and presents a potential natural model for NCL6 in humans.
Collapse
Affiliation(s)
| | - Fredrik S Skedsmo
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Josefin Hultman
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Johan Høgset Jansen
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Frode Lingaas
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
3
|
Critchley BJ, Gaspar HB, Benedetti S. Targeting the central nervous system in lysosomal storage diseases: Strategies to deliver therapeutics across the blood-brain barrier. Mol Ther 2023; 31:657-675. [PMID: 36457248 PMCID: PMC10014236 DOI: 10.1016/j.ymthe.2022.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/18/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are multisystem inherited metabolic disorders caused by dysfunctional lysosomal activity, resulting in the accumulation of undegraded macromolecules in a variety of organs/tissues, including the central nervous system (CNS). Treatments include enzyme replacement therapy, stem/progenitor cell transplantation, and in vivo gene therapy. However, these treatments are not fully effective in treating the CNS as neither enzymes, stem cells, nor viral vectors efficiently cross the blood-brain barrier. Here, we review the latest advancements in improving delivery of different therapeutic agents to the CNS and comment upon outstanding questions in the field of neurological LSDs.
Collapse
Affiliation(s)
- Bethan J Critchley
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK
| | - H Bobby Gaspar
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; Orchard Therapeutics Ltd., London EC4N 6EU, UK
| | - Sara Benedetti
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
4
|
Nittari G, Tomassoni D, Roy P, Martinelli I, Tayebati SK, Amenta F. Batten disease through different in vivo and in vitro models: A review. J Neurosci Res 2023; 101:298-315. [PMID: 36434776 DOI: 10.1002/jnr.25147] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022]
Abstract
Batten disease consists of a family of primarily autosomal recessive, progressive neuropediatric disorders, also known as neuronal ceroid lipofuscinoses (NCLs). These pathologies are characterized by seizures and visual, cognitive and motor decline, and premature death. The pathophysiology of this rare disease is still unclear despite the years of trials and financial aids. This paper has reviewed advantages and limits of in vivo and in vitro models of Batten disease from murine and larger animal models to primitive unicellular models, until the most recently developed patient-derived induced pluripotent stem cells. For each model advantages, limits and applications were analyzed. The first prototypes investigated were murine models that due to their limits were replaced by larger animals. In vitro models gradually replaced animal models for practical, cost, and ethical reasons. Using induced pluripotent stem cells to study neurodegeneration is a new way of studying the disease, since they can be distinguished into differentiating elements like neurons, which are susceptible to neurodegeneration. In vivo and in vitro models have contributed to clarifying to some extent the pathophysiology of the disease. The collection and sharing of suitable human bio samples likely through biobanks can contribute to a better understanding, prevention, and to identify possible treatment strategies of Batten disease.
Collapse
Affiliation(s)
- Giulio Nittari
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Ilenia Martinelli
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Seyed Khosrow Tayebati
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Francesco Amenta
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| |
Collapse
|
5
|
Sampaio LPDB, Manreza MLGD, Pessoa A, Gurgel-Giannetti J, Coan AC, Júnior HVDL, Embiruçu EK, Henriques-Souza AMDM, Kok F. Clinical management and diagnosis of CLN2 disease: consensus of the Brazilian experts group. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:284-295. [PMID: 37059438 PMCID: PMC10104757 DOI: 10.1055/s-0043-1761434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
Neuronal ceroid lipofuscinosis type 2 (CLN2) is a rare neurodegenerative genetic disease that affects children in early life. Its classic form is rapidly progressive, leading to death within the first 10 years. The urge for earlier diagnosis increases with the availability of enzyme replacement therapy. A panel of nine Brazilian child neurologists combined their expertise in CLN2 with evidence from the medical literature to establish a consensus to manage this disease in Brazil. They voted 92 questions including diagnosis, clinical manifestations, and treatment of the disease, considering the access to healthcare in this country. Clinicians should suspect CLN2 disease in any child, from 2 to 4 years old, with language delay and epilepsy. Even though the classic form is the most prevalent, atypical cases with different phenotypes can be found. Electroencephalogram, magnetic resonance imaging, molecular and biochemical testing are the main tools to investigate and confirm the diagnosis. However, we have limited access to molecular testing in Brazil, and rely on the support from the pharmaceutical industry. The management of CLN2 should involve a multidisciplinary team and focus on the quality of life of patients and on family support. Enzyme replacement therapy with Cerliponase α is an innovative treatment approved in Brazil since 2018; it delays functional decline and provides quality of life. Given the difficulties for the diagnosis and treatment of rare diseases in our public health system, the early diagnosis of CLN2 needs improvement as enzyme replacement therapy is available and modifies the prognosis of patients.
Collapse
Affiliation(s)
| | | | - André Pessoa
- Universidade Estadual do Ceará, Hospital Infantil Albert Sabin, Fortaleza CE, Brazil
| | - Juliana Gurgel-Giannetti
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Hospital das Clínicas, Belo Horizonte MG, Brazil
| | - Ana Carolina Coan
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Campinas SP, Brazil
| | | | - Emília Katiane Embiruçu
- Universidade do Estado da Bahia, Hospital Universitário Professor Edgard Santos, Salvador BA, Brazil
| | | | - Fernando Kok
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo SP, Brazil
| |
Collapse
|
6
|
Kick GR, Whiting REH, Ota-Kuroki J, Castaner LJ, Morgan-Jack B, Sabol JC, Meiman EJ, Ortiz F, Katz ML. Intravitreal gene therapy preserves retinal function in a canine model of CLN2 neuronal ceroid lipofuscinosis. Exp Eye Res 2023; 226:109344. [PMID: 36509165 PMCID: PMC9839638 DOI: 10.1016/j.exer.2022.109344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/02/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
CLN2 neuronal ceroid lipofuscinosis is a rare hereditary neurodegenerative disorder characterized by deleterious sequence variants in TPP1 that result in reduced or abolished function of the lysosomal enzyme tripeptidyl peptidase 1 (TPP1). Children with this disorder experience progressive neurological decline and vision loss starting around 2-4 years of age. Ocular disease is characterized by progressive retinal degeneration and impaired retinal function culminating in total loss of vision. Similar retinal pathology occurs in a canine model of CLN2 disease with a null variant in TPP1. A study using the dog model was performed to evaluate the efficacy of ocular gene therapy to provide a continuous, long-term source of human TPP1 (hTPP1) to the retina, inhibit retinal degeneration and preserve retinal function. TPP1-/- dogs received an intravitreal injection of 1 x 1012 viral genomes of AAV2.CAG.hTPP1 in one eye and AAV2.CAG.GFP in the contralateral eye at 4 months of age. Ophthalmic exams, in vivo ocular imaging and electroretinography were repeated monthly to assess retinal structure and function. Retinal morphology, hTPP1 and GFP expression in the retina, optic nerve and lateral geniculate nucleus, and hTPP1 concentrations in the vitreous were evaluated after the dogs were euthanized at end stage neurological disease at approximately 10 months of age. Intravitreal administration of AAV2.CAG.hTPP1 resulted in stable, widespread expression of hTPP1 throughout the inner retina, prevented disease-related declines in retinal function and inhibited disease-related cell loss and storage body accumulation in the retina for at least 6 months. Uveitis occurred in eyes treated with the hTPP1 vector, but this did not prevent therapeutic efficacy. The severity of the uveitis was ameliorated with anti-inflammatory treatments. These results indicate that a single intravitreal injection of AAV2.CAG.hTPP1 is an effective treatment to inhibit ocular disease progression in canine CLN2 disease.
Collapse
Affiliation(s)
- Grace Robinson Kick
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Rebecca E H Whiting
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Juri Ota-Kuroki
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Leilani J Castaner
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Brandie Morgan-Jack
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Julianna C Sabol
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Elizabeth J Meiman
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Francheska Ortiz
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA
| | - Martin L Katz
- Neurodegenerative Diseases Research Laboratory, University of Missouri School of Medicine, Columbia, MO, USA.
| |
Collapse
|
7
|
Pinkstaff J, McCullagh E, Grover A, Melton AC, Cherukuri A, Wait JC, Nguyen A, Butt MT, Trombley JL, Reed RP, Adams EL, Boyd RB, Chandra S, Henshaw J, O'Neill CA, Zanelli E, Kovalchin J. Safety, pharmacokinetics and CNS distribution of tralesinidase alfa administered via intracerebroventricular infusion to juvenile cynomolgus monkeys. Toxicol Rep 2023; 10:357-366. [PMID: 36923444 PMCID: PMC10009680 DOI: 10.1016/j.toxrep.2023.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Mucopolysaccharidosis Type IIIB (MPS IIIB) is an ultrarare, fatal pediatric disease with no approved therapy. It is caused by mutations in the gene encoding for lysosomal enzyme alpha-N-acetylglucosaminidase (NAGLU). Tralesinidase alfa (TA) is a fusion protein comprised of recombinant NAGLU and a modified human insulin-like growth factor 2 that is being developed as an enzyme replacement therapy for MPS IIIB. Since MPS IIIB is a pediatric disease the safety/toxicity, pharmacokinetics and biodistribution of TA were evaluated in juvenile non-human primates that were administered up to 5 weekly intracerebroventricular (ICV) or single intravenous (IV) infusions of TA. TA administered by ICV slow-, ICV isovolumetric bolus- or IV-infusion was well-tolerated, and no effects were observed on clinical observations, electrocardiographic or ophthalmologic parameters, or respiratory rates. The drug-related changes observed were limited to increased cell infiltrates in the CSF and along the ICV catheter track after ICV administration. These findings were not associated with functional changes and are associated with the use of ICV catheters. The CSF PK profiles were consistent across all conditions tested and TA distributed widely in the CNS after ICV administration. Anti-drug antibodies were observed but did not appear to significantly affect the exposure to TA. Correlations between TA concentrations in plasma and brain regions in direct contact with the cisterna magna suggest glymphatic drainage may be responsible for clearance of TA from the CNS. The data support the administration of TA by isovolumetric bolus ICV infusion to pediatric patients with MPS IIIB.
Collapse
Key Words
- ADA, anti-drug antibodies
- AUC, area under the curve
- CLN2, Neuronal Ceroid Lipofuscinosis Type 2
- CNS, central nervous system
- CSF, cerebrospinal fluid
- Cmax, maximal concentration
- ERT, enzyme replacement therapy
- Enzyme replacement therapy
- H&E, Hematoxylin and Eosin
- HS, heparan sulfate
- ICV, intracerebroventricular
- IGF2, insulin-like growth factor 2
- IT-L, intrathecal lumbar
- IV, intravenous
- Intracerebroventricular
- LLOQ, lower limit of quantitation
- MPS IIIB
- MPS IIIB, mucopolysaccharidosis type IIIB
- NAGLU
- NAGLU, alpha-N-acetylglucosaminidase
- NBF, neutral buffered formalin
- NHP, non-human primate
- PK, pharmacokinetics
- QW, once weekly
- Sanfilippo syndrome
- T1/2, time required for compound concentration to decrease by 50%
- TA, tralesinidase alfa
- Tmax, time at which maximal concentration is achieved
- WBC, white blood cell count
- aCSF, artificial cerebrospinal fluid
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Randall P Reed
- Northern Biomedical Research Inc., Norton Shores, MI, USA
| | - Eric L Adams
- Northern Biomedical Research Inc., Norton Shores, MI, USA
| | - Robert B Boyd
- Northern Biomedical Research Inc., Norton Shores, MI, USA
| | | | | | | | | | | |
Collapse
|
8
|
Nelvagal HR, Eaton SL, Wang SH, Eultgen EM, Takahashi K, Le SQ, Nesbitt R, Dearborn JT, Siano N, Puhl AC, Dickson PI, Thompson G, Murdoch F, Brennan PM, Gray M, Greenhalgh SN, Tennant P, Gregson R, Clutton E, Nixon J, Proudfoot C, Guido S, Lillico SG, Whitelaw CBA, Lu JY, Hofmann SL, Ekins S, Sands MS, Wishart TM, Cooper JD. Cross-species efficacy of enzyme replacement therapy for CLN1 disease in mice and sheep. J Clin Invest 2022; 132:163107. [PMID: 36040802 PMCID: PMC9566914 DOI: 10.1172/jci163107] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
CLN1 disease, also called infantile neuronal ceroid lipofuscinosis (NCL) or infantile Batten disease, is a fatal neurodegenerative lysosomal storage disorder resulting from mutations in the CLN1 gene encoding the soluble lysosomal enzyme palmitoyl-protein thioesterase 1 (PPT1). Therapies for CLN1 disease have proven challenging because of the aggressive disease course and the need to treat widespread areas of the brain and spinal cord. Indeed, gene therapy has proven less effective for CLN1 disease than for other similar lysosomal enzyme deficiencies. We therefore tested the efficacy of enzyme replacement therapy (ERT) by administering monthly infusions of recombinant human PPT1 (rhPPT1) to PPT1-deficient mice (Cln1-/-) and CLN1R151X sheep to assess how to potentially scale up for translation. In Cln1-/- mice, intracerebrovascular (i.c.v.) rhPPT1 delivery was the most effective route of administration, resulting in therapeutically relevant CNS levels of PPT1 activity. rhPPT1-treated mice had improved motor function, reduced disease-associated pathology, and diminished neuronal loss. In CLN1R151X sheep, i.c.v. infusions resulted in widespread rhPPT1 distribution and positive treatment effects measured by quantitative structural MRI and neuropathology. This study demonstrates the feasibility and therapeutic efficacy of i.c.v. rhPPT1 ERT. These findings represent a key step toward clinical testing of ERT in children with CLN1 disease and highlight the importance of a cross-species approach to developing a successful treatment strategy.
Collapse
Affiliation(s)
- Hemanth R. Nelvagal
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Samantha L. Eaton
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Sophie H. Wang
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth M. Eultgen
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Keigo Takahashi
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Steven Q. Le
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Rachel Nesbitt
- Department of Medicine, Washington University in St. Louis, School of Medicine, St .Louis, Missouri, USA
| | - Joshua T. Dearborn
- Department of Medicine, Washington University in St. Louis, School of Medicine, St .Louis, Missouri, USA
| | - Nicholas Siano
- Discovery Science Division, Amicus Therapeutics Inc., Philadelphia, Pennsylvania, USA
| | - Ana C. Puhl
- Collaborations Pharmaceuticals Inc., Lab 3510, Raleigh, North Carolina, USA
| | - Patricia I. Dickson
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Gerard Thompson
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, Edinburgh, Scotland, United Kingdom
- Department of Clinical Neurosciences, NHS Lothian, Edinburgh, Scotland, United Kingdom
| | - Fraser Murdoch
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Paul M. Brennan
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, Edinburgh, Scotland, United Kingdom
- Department of Clinical Neurosciences, NHS Lothian, Edinburgh, Scotland, United Kingdom
| | - Mark Gray
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Stephen N. Greenhalgh
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Peter Tennant
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Rachael Gregson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Eddie Clutton
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - James Nixon
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Chris Proudfoot
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Stefano Guido
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Simon G. Lillico
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - C. Bruce A. Whitelaw
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Jui-Yun Lu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sandra L. Hofmann
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., Lab 3510, Raleigh, North Carolina, USA
| | - Mark S. Sands
- Department of Medicine, Washington University in St. Louis, School of Medicine, St .Louis, Missouri, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Thomas M. Wishart
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Jonathan D. Cooper
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
9
|
Swier VJ, White KA, Johnson TB, Sieren JC, Johnson HJ, Knoernschild K, Wang X, Rohret FA, Rogers CS, Pearce DA, Brudvig JJ, Weimer JM. A Novel Porcine Model of CLN2 Batten Disease that Recapitulates Patient Phenotypes. Neurotherapeutics 2022; 19:1905-1919. [PMID: 36100791 PMCID: PMC9723024 DOI: 10.1007/s13311-022-01296-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2022] [Indexed: 12/13/2022] Open
Abstract
CLN2 Batten disease is a lysosomal disorder in which pathogenic variants in CLN2 lead to reduced activity in the enzyme tripeptidyl peptidase 1. The disease typically manifests around 2 to 4 years of age with developmental delay, ataxia, seizures, inability to speak and walk, and fatality between 6 and 12 years of age. Multiple Cln2 mouse models exist to better understand the etiology of the disease; however, these models are unable to adequately recapitulate the disease due to differences in anatomy and physiology, limiting their utility for therapeutic testing. Here, we describe a new CLN2R208X/R208X porcine model of CLN2 disease. We present comprehensive characterization showing behavioral, pathological, and visual phenotypes that recapitulate those seen in CLN2 patients. CLN2R208X/R208X miniswine present with gait abnormalities at 6 months of age, ERG waveform declines at 6-9 months, vision loss at 11 months, cognitive declines at 12 months, seizures by 15 months, and early death at 18 months due to failure to thrive. CLN2R208X/R208X miniswine also showed classic storage material accumulation and glial activation in the brain at 6 months, and cortical atrophy at 12 months. Thus, the CLN2R208X/R208X miniswine model is a valuable resource for biomarker discovery and therapeutic development in CLN2 disease.
Collapse
Affiliation(s)
- Vicki J Swier
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Tyler B Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Jessica C Sieren
- Department of Radiology, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Hans J Johnson
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Kevin Knoernschild
- Department of Radiology, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | | | | | | | - David A Pearce
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Jon J Brudvig
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA.
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA.
| |
Collapse
|
10
|
Takahashi K, Nelvagal HR, Lange J, Cooper JD. Glial Dysfunction and Its Contribution to the Pathogenesis of the Neuronal Ceroid Lipofuscinoses. Front Neurol 2022; 13:886567. [PMID: 35444603 PMCID: PMC9013902 DOI: 10.3389/fneur.2022.886567] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/16/2022] [Indexed: 01/05/2023] Open
Abstract
While significant efforts have been made in developing pre-clinical treatments for the neuronal ceroid lipofuscinoses (NCLs), many challenges still remain to bring children with NCLs a cure. Devising effective therapeutic strategies for the NCLs will require a better understanding of pathophysiology, but little is known about the mechanisms by which loss of lysosomal proteins causes such devastating neurodegeneration. Research into glial cells including astrocytes, microglia, and oligodendrocytes have revealed many of their critical functions in brain homeostasis and potential contributions to neurodegenerative diseases. Genetically modified mouse models have served as a useful platform to define the disease progression in the central nervous system across NCL subtypes, revealing a wide range of glial responses to disease. The emerging evidence of glial dysfunction questions the traditional “neuron-centric” view of NCLs, and would suggest that directly targeting glia in addition to neurons could lead to better therapeutic outcomes. This review summarizes the most up-to-date understanding of glial pathologies and their contribution to the pathogenesis of NCLs, and highlights some of the associated challenges that require further research.
Collapse
Affiliation(s)
- Keigo Takahashi
- Pediatric Storage Disorders Laboratory, Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Hemanth R. Nelvagal
- Department of Pharmacology, School of Pharmacy, University College London, London, United Kingdom
| | - Jenny Lange
- Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Jonathan D. Cooper
- Pediatric Storage Disorders Laboratory, Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Genetics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- *Correspondence: Jonathan D. Cooper
| |
Collapse
|
11
|
Clark GD, Lotze TE. Novel Treatments and Clinical Research in Child Neurology. Neurol Clin 2021; 39:719-722. [PMID: 34215383 DOI: 10.1016/j.ncl.2021.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The molecular understanding of the pathogenic mechanisms responsible for neurologic diseases of children has led to a remarkable period of research that addresses the root causes of diseases. The promise of this research has been realized with cures and treatments that correct underlying deficiencies. The breakneck rate at which new research is being proposed promises to usher in a transformation of child neurology from a diagnostic and supportive field into an interventional one. Training child neurology residents in clinical research and therapeutic intervention is increasingly important to assure the ongoing ability to support research discoveries and treatment.
Collapse
Affiliation(s)
- Gary D Clark
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, 6701 Fannin Street Suite 1250, Houston, TX 77030, USA
| | - Timothy E Lotze
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, 6701 Fannin Street Suite 1250, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Hammon K, de Hart G, Vuillemenot BR, Kennedy D, Musson D, O'Neill CA, Katz ML, Henshaw JW. Dose selection for intracerebroventricular cerliponase alfa in children with CLN2 disease, translation from animal to human in a rare genetic disease. Clin Transl Sci 2021; 14:1810-1821. [PMID: 34076336 PMCID: PMC8504808 DOI: 10.1111/cts.13028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 11/28/2022] Open
Abstract
Neuronal ceroid lipofuscinosis type 2 (CLN2 disease) is an ultra‐rare pediatric neurodegenerative disorder characterized by deficiency of the lysosomal enzyme tripeptidyl peptidase‐1 (TPP1). In the absence of adequate TPP1, lysosomal storage material accumulation occurs in the central nervous system (CNS) accompanied by neurodegeneration and neurological decline that culminates in childhood death. Cerliponase alfa is a recombinant human TPP1 enzyme replacement therapy administered via intracerebroventricular infusion and approved for the treatment of CLN2 disease. Here, we describe two allometric methods, calculated by scaling brain mass across species, that informed the human dose selection and exposure prediction of cerliponase alfa from preclinical studies in monkeys and a dog model of CLN2 disease: (1) scaling of dose using a human‐equivalent dose factor; and (2) scaling of compartmental pharmacokinetic (PK) model parameters. Source PK data were obtained from cerebrospinal fluid (CSF) samples from dogs and monkeys, and the human exposure predictions were confirmed with CSF data from the first‐in‐human clinical study. Nonclinical and clinical data were analyzed using noncompartmental analysis and nonlinear mixed‐effect modeling approaches. Both allometric methods produced CSF exposure predictions within twofold of the observed exposure parameters maximum plasma concentration (Cmax) and area under the curve (AUC). Furthermore, cross‐species qualification produced consistent and reasonable PK profile predictions, which supported the allometric scaling of model parameters. The challenges faced in orphan drug development place an increased importance on, and opportunity for, data translation from research and nonclinical development. Our approach to dose translation and human exposure prediction for cerliponase alfa may be applicable to other CNS administered therapies being developed.
Collapse
Affiliation(s)
- Kevin Hammon
- BioMarin Pharmaceutical Inc., Novato, California, USA
| | - Greg de Hart
- BioMarin Pharmaceutical Inc., Novato, California, USA
| | | | - Derek Kennedy
- BioMarin Pharmaceutical Inc., Novato, California, USA
| | - Don Musson
- BioMarin Pharmaceutical Inc., Novato, California, USA
| | | | - Martin L Katz
- Mason Eye Institute, University of Missouri School of Medicine, Columbia, Missouri, USA.,Department of Bioengineering, University of Missouri, Columbia, Missouri, USA
| | | |
Collapse
|
13
|
Cerliponase alfa changes the natural history of children with neuronal ceroid lipofuscinosis type 2: The first French cohort. Eur J Paediatr Neurol 2021; 30:17-21. [PMID: 33348105 DOI: 10.1016/j.ejpn.2020.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 11/30/2020] [Accepted: 12/04/2020] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Neuronal Ceroid Lipofuscinosis type 2 (CLN2) is a neurodegenerative lysosomal disease which leads to early dementia and death without treatment. The recently available therapy consists of intracerebroventricular enzyme substitution: cerliponase alfa. In this report, we describe the evolution of the first French children treated with cerliponase alfa. METHOD CLN2 Clinical Rating Scale Motor-Language (CLN2 ML) assesses the motor and language evolution of CLN2 patients. We retrospectively studied patients' medical records: clinical symptoms, MRI conclusions, gene mutation, side effects of infusions, patient's age and CLN2 ML scores at diagnosis, at the beginning of enzyme replacement therapy (ERT) and at the last evaluation. Seven patients were included. RESULTS Average age at diagnosis was 50 months ( ±10) with CLN2 ML score equal to 3.6 [1.5-5]. Average age at the beginning of ERT was 56 months ( ±13) with CLN2 ML score equal to 3.1 [1-5]. At the last available evaluation, average age was 82 months ( ±20) with CLN2 ML score equal to 2.8 [0-5]. Thus, in 26 months, the mean CLN2 ML score only decreased by 0.3 points. However, patients with a CLN2 ML score greater than three at the onset of ERT experienced a stabilisation or improvement of clinical signs, whereas patients with a CLN2 ML score less than three at baseline continue to deteriorate. CONCLUSION For patients starting ERT at an early stage of the disease, cerliponase alfa changes the natural history of the disease with a halt in disease progression or even a slight improvement in clinical symptoms.
Collapse
|
14
|
Maeser S, Petre BA, Ion L, Rawer S, Kohlschütter A, Santorelli FM, Simonati A, Schulz A, Przybylski M. Enzymatic diagnosis of neuronal lipofuscinoses in dried blood spots using substrates for concomitant tandem mass spectrometry and fluorimetry. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4675. [PMID: 33314470 DOI: 10.1002/jms.4675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/06/2020] [Accepted: 10/20/2020] [Indexed: 06/12/2023]
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are a group of neurodegenerative diseases predominantly in childhood that are characterized by psychomotor deterioration, epilepsy, and early death of patients. The NCLs analyzed in the present study are caused by defects of the specific enzymes, CLN1 (palmitoyl protein thioesterase 1; PPT1), CLN2 (tripeptidyl peptidase 1; TPP1), and CLN10 (cathepsin D). Specific and sensitive diagnostic assays of NCLs were the main goal of this study. They are of increasing importance, particularly since enzyme replacement therapy (ERT) for NCL2 has recently become available for clinical treatment, and ERTs for further NCLs are under development. Here, we report specific and sensitive determinations for CLN1, CLN2, and CLN10 on dried blood spots by tandem mass spectrometry using multiple reaction monitoring mass spectrometry (MRM-MS). Identical substrates suitable for (i) fluorimetric determination of single enzymes and (ii) for MRM-MS determination of multiple enzymes were synthesized by chemical coupling of alkyl-umbelliferone building blocks with the corresponding peptidyl-substrate groups recognized by the target enzyme. Enzymatic determinations were performed both by fluorimetry and MRM-MS in patients with NCL1, NCL2, and NCL10 and showed good agreement in single assays. Moreover, duplex and triplex determinations were successfully performed for NCL1, NCL2, and NCL10. Specific peptidyl-(4-alkyl-umbelliferone) substrates were also synthesized for mass spectrometric determinations of different cathepsins (cathepsins-D, -F, and -B), to provide a differentiation of proteolytic specificities.
Collapse
Affiliation(s)
- Stefan Maeser
- Steinbeis Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Mass Spectrometry Laboratory, Marktstrasse 29, Ruesselsheim am Main, D-65428, Germany
- Department of Chemistry, Universität Konstanz, Konstanz, 78457, Germany
| | - Brindusa-Alina Petre
- Steinbeis Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Mass Spectrometry Laboratory, Marktstrasse 29, Ruesselsheim am Main, D-65428, Germany
- Department of Chemistry, Universität Konstanz, Konstanz, 78457, Germany
- Department of Chemistry, A.I. Cuza University of Iasi, Iasi, Romania
| | - Laura Ion
- Steinbeis Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Mass Spectrometry Laboratory, Marktstrasse 29, Ruesselsheim am Main, D-65428, Germany
- Department of Chemistry, Universität Konstanz, Konstanz, 78457, Germany
- Department of Chemistry, A.I. Cuza University of Iasi, Iasi, Romania
| | - Stephan Rawer
- Steinbeis Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Mass Spectrometry Laboratory, Marktstrasse 29, Ruesselsheim am Main, D-65428, Germany
- Department of Chemistry, Universität Konstanz, Konstanz, 78457, Germany
| | - Alfried Kohlschütter
- Children's Hospital, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, D-20246, Germany
| | - Filippo M Santorelli
- Department of Molecular Medicine, IRCCS Foundation Stella Maris, I-56128 Calambrone, Pisa, Italy
| | - Alessandro Simonati
- Department of Neuroscience, Biomedicine, Movement-Neurology and Neuropathology, Policlinico GB Rossi, P.le LA Scuro, Verona, I-37134, Italy
| | - Angela Schulz
- Children's Hospital, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, D-20246, Germany
| | - Michael Przybylski
- Steinbeis Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Mass Spectrometry Laboratory, Marktstrasse 29, Ruesselsheim am Main, D-65428, Germany
- Department of Chemistry, Universität Konstanz, Konstanz, 78457, Germany
| |
Collapse
|
15
|
Aylward SC, Pindrik J, Abreu NJ, Cherny WB, O’Neal M, de Los Reyes E. Cerliponase alfa for CLN2 disease, a promising therapy. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1856654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Shawn C. Aylward
- Department of Pediatrics and Neurology, Nationwide Children‘s Hospital, Columbus, OH, USA
| | - Jonathan Pindrik
- Division of Pediatric Neurosurgery, Nationwide Children‘s Hospital, Columbus, OH, USA
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Nicolas J. Abreu
- Department of Pediatrics and Neurology, Nationwide Children‘s Hospital, Columbus, OH, USA
| | - W. Bruce Cherny
- Department of Pediatric Neurosurgery, St. Luke‘s Children‘s Hospital, Boise, ID, USA
| | - Matthew O’Neal
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Emily de Los Reyes
- Department of Pediatrics and Neurology, Nationwide Children‘s Hospital, Columbus, OH, USA
| |
Collapse
|
16
|
Blanset D, Hutt J, Morgan S. Current use of animal models of disease for nonclinical safety testing. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
17
|
Katz ML, Buckley RM, Biegen V, O'Brien DP, Johnson GC, Warren WC, Lyons LA. Neuronal Ceroid Lipofuscinosis in a Domestic Cat Associated with a DNA Sequence Variant That Creates a Premature Stop Codon in CLN6. G3 (BETHESDA, MD.) 2020; 10:2741-2751. [PMID: 32518081 PMCID: PMC7407459 DOI: 10.1534/g3.120.401407] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/06/2020] [Indexed: 01/04/2023]
Abstract
A neutered male domestic medium-haired cat presented at a veterinary neurology clinic at 20 months of age due to progressive neurological signs that included visual impairment, focal myoclonus, and frequent severe generalized seizures that were refractory to treatment with phenobarbital. Magnetic resonance imaging revealed diffuse global brain atrophy. Due to the severity and frequency of its seizures, the cat was euthanized at 22 months of age. Microscopic examination of the cerebellum, cerebral cortex and brainstem revealed pronounced intracellular accumulations of autofluorescent storage material and inflammation in all 3 brain regions. Ultrastructural examination of the storage material indicated that it consisted almost completely of tightly-packed membrane-like material. The clinical signs and neuropathology strongly suggested that the cat suffered from a form of neuronal ceroid lipofuscinosis (NCL). Whole exome sequence analysis was performed on genomic DNA from the affected cat. Comparison of the sequence data to whole exome sequence data from 39 unaffected cats and whole genome sequence data from an additional 195 unaffected cats revealed a homozygous variant in CLN6 that was unique to the affected cat. This variant was predicted to cause a stop gain in the transcript due to a guanine to adenine transition (ENSFCAT00000025909:c.668G > A; XM_003987007.5:c.668G > A) and was the sole loss of function variant detected. CLN6 variants in other species, including humans, dogs, and sheep, are associated with the CLN6 form of NCL. Based on the affected cat's clinical signs, neuropathology and molecular genetic analysis, we conclude that the cat's disorder resulted from the loss of function of CLN6. This study is only the second to identify the molecular genetic basis of a feline NCL. Other cats exhibiting similar signs can now be screened for the CLN6 variant. This could lead to establishment of a feline model of CLN6 disease that could be used in therapeutic intervention studies.
Collapse
Affiliation(s)
- Martin L Katz
- Neurodegenerative Diseases Research Laboratory and Department of Ophthalmology,
| | | | | | | | | | - Wesley C Warren
- Life Sciences Center, University of Missouri, Columbia, MO and
| | | |
Collapse
|
18
|
Specchio N, Pietrafusa N, Trivisano M. Changing Times for CLN2 Disease: The Era of Enzyme Replacement Therapy. Ther Clin Risk Manag 2020; 16:213-222. [PMID: 32280231 PMCID: PMC7127909 DOI: 10.2147/tcrm.s241048] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/18/2020] [Indexed: 01/23/2023] Open
Abstract
Neuronal ceroid lipofuscinosis type 2 (CLN2 disease) is a progressive neurodegenerative disease that results in early-onset, severe, progressive, neurological disabilities, leading to death in late childhood or early adolescence. Management has relied on symptomatic care, and supportive and palliative strategies, but the approval of the enzyme replacement therapy cerliponase alfa in the USA and Europe in 2017 brought different treatment opportunities. We describe the natural history of CLN2 disease, its diagnosis and management, and the preclinical and clinical development of cerliponase alfa. A PubMed search was undertaken for cerliponase alfa and rhTPP1 to identify preclinical and clinical studies. The hallmark-presenting symptoms of CLN2 disease are unprovoked seizures and a history of language delay, and progression involves motor dysfunction, and cognitive and visual decline. Cerliponase alfa has shown efficacy and tolerability in mouse and canine models of CLN2 disease when delivered intracerebroventricularly. Administration of cerliponase alfa in patients with CLN2 disease has led to significant reductions in the rate of decline of motor and language functions in comparison with a natural history population. The approval of cerliponase alfa has brought a new era for CLN2 disease, highlighting the need to understand different patterns of disease progression and clinical needs in treated patients.
Collapse
Affiliation(s)
- Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Nicola Pietrafusa
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marina Trivisano
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
19
|
Story BD, Miller ME, Bradbury AM, Million ED, Duan D, Taghian T, Faissler D, Fernau D, Beecy SJ, Gray-Edwards HL. Canine Models of Inherited Musculoskeletal and Neurodegenerative Diseases. Front Vet Sci 2020; 7:80. [PMID: 32219101 PMCID: PMC7078110 DOI: 10.3389/fvets.2020.00080] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Mouse models of human disease remain the bread and butter of modern biology and therapeutic discovery. Nonetheless, more often than not mouse models do not reproduce the pathophysiology of the human conditions they are designed to mimic. Naturally occurring large animal models have predominantly been found in companion animals or livestock because of their emotional or economic value to modern society and, unlike mice, often recapitulate the human disease state. In particular, numerous models have been discovered in dogs and have a fundamental role in bridging proof of concept studies in mice to human clinical trials. The present article is a review that highlights current canine models of human diseases, including Alzheimer's disease, degenerative myelopathy, neuronal ceroid lipofuscinosis, globoid cell leukodystrophy, Duchenne muscular dystrophy, mucopolysaccharidosis, and fucosidosis. The goal of the review is to discuss canine and human neurodegenerative pathophysiologic similarities, introduce the animal models, and shed light on the ability of canine models to facilitate current and future treatment trials.
Collapse
Affiliation(s)
- Brett D. Story
- Auburn University College of Veterinary Medicine, Auburn, AL, United States
- University of Florida College of Veterinary Medicine, Gainesville, FL, United States
| | - Matthew E. Miller
- Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Allison M. Bradbury
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily D. Million
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Dominik Faissler
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, United States
| | - Deborah Fernau
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Sidney J. Beecy
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, United States
| | - Heather L. Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
20
|
Nelvagal HR, Cooper JD. An update on the progress of preclinical models for guiding therapeutic management of neuronal ceroid lipofuscinosis. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1703672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Hemanth Ramesh Nelvagal
- Department of Pediatrics, Division of genetics and genomics, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | - Jonathan D Cooper
- Department of Pediatrics, Division of genetics and genomics, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| |
Collapse
|
21
|
Huber RJ, Hughes SM, Liu W, Morgan A, Tuxworth RI, Russell C. The contribution of multicellular model organisms to neuronal ceroid lipofuscinosis research. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165614. [PMID: 31783156 DOI: 10.1016/j.bbadis.2019.165614] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023]
Abstract
The NCLs (neuronal ceroid lipofuscinosis) are forms of neurodegenerative disease that affect people of all ages and ethnicities but are most prevalent in children. Commonly known as Batten disease, this debilitating neurological disorder is comprised of 13 different subtypes that are categorized based on the particular gene that is mutated (CLN1-8, CLN10-14). The pathological mechanisms underlying the NCLs are not well understood due to our poor understanding of the functions of NCL proteins. Only one specific treatment (enzyme replacement therapy) is approved, which is for the treating the brain in CLN2 disease. Hence there remains a desperate need for further research into disease-modifying treatments. In this review, we present and evaluate the genes, proteins and studies performed in the social amoeba, nematode, fruit fly, zebrafish, mouse and large animals pertinent to NCL. In particular, we highlight the use of multicellular model organisms to study NCL protein function, pathology and pathomechanisms. Their use in testing novel therapeutic approaches is also presented. With this information, we highlight how future research in these systems may be able to provide new insight into NCL protein functions in human cells and aid in the development of new therapies.
Collapse
Affiliation(s)
- Robert J Huber
- Department of Biology, Trent University, Peterborough, Ontario K9L 0G2, Canada
| | - Stephanie M Hughes
- Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre and Genetics Otago, University of Otago, Dunedin, New Zealand
| | - Wenfei Liu
- School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Alan Morgan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown St., Liverpool L69 3BX, UK
| | - Richard I Tuxworth
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Claire Russell
- Dept. Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK.
| |
Collapse
|
22
|
Sleat DE, Wiseman JA, El-Banna M, Zheng H, Zhao C, Soherwardy A, Moore DF, Lobel P. Analysis of Brain and Cerebrospinal Fluid from Mouse Models of the Three Major Forms of Neuronal Ceroid Lipofuscinosis Reveals Changes in the Lysosomal Proteome. Mol Cell Proteomics 2019; 18:2244-2261. [PMID: 31501224 PMCID: PMC6823856 DOI: 10.1074/mcp.ra119.001587] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/06/2019] [Indexed: 01/06/2023] Open
Abstract
Treatments are emerging for the neuronal ceroid lipofuscinoses (NCLs), a group of similar but genetically distinct lysosomal storage diseases. Clinical ratings scales measure long-term disease progression and response to treatment but clinically useful biomarkers have yet to be identified in these diseases. We have conducted proteomic analyses of brain and cerebrospinal fluid (CSF) from mouse models of the most frequently diagnosed NCL diseases: CLN1 (infantile NCL), CLN2 (classical late infantile NCL) and CLN3 (juvenile NCL). Samples were obtained at different stages of disease progression and proteins quantified using isobaric labeling. In total, 8303 and 4905 proteins were identified from brain and CSF, respectively. We also conduced label-free analyses of brain proteins that contained the mannose 6-phosphate lysosomal targeting modification. In general, we detect few changes at presymptomatic timepoints but later in disease, we detect multiple proteins whose expression is significantly altered in both brain and CSF of CLN1 and CLN2 animals. Many of these proteins are lysosomal in origin or are markers of neuroinflammation, potentially providing clues to underlying pathogenesis and providing promising candidates for further validation.
Collapse
Affiliation(s)
- David E Sleat
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ 08854; Department of Biochemistry and Molecular Biology, Robert-Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Piscataway, NJ 08854.
| | | | - Mukarram El-Banna
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ 08854
| | - Haiyan Zheng
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ 08854
| | - Caifeng Zhao
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ 08854
| | - Amenah Soherwardy
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ 08854
| | - Dirk F Moore
- Department of Biostatistics, School of Public Health, Rutgers - The State University of New Jersey, Piscataway, NJ 08854
| | - Peter Lobel
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ 08854; Department of Biochemistry and Molecular Biology, Robert-Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Piscataway, NJ 08854.
| |
Collapse
|
23
|
Chakrabarti S, Chandra S, Roy A, Dasarathi S, Kundu M, Pahan K. Upregulation of tripeptidyl-peptidase 1 by 3-hydroxy-(2,2)-dimethyl butyrate, a brain endogenous ligand of PPARα: Implications for late-infantile Batten disease therapy. Neurobiol Dis 2019; 127:362-373. [PMID: 30928643 PMCID: PMC6588492 DOI: 10.1016/j.nbd.2019.03.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/11/2019] [Accepted: 03/24/2019] [Indexed: 11/24/2022] Open
Abstract
The late-infantile Batten disease or late-infantile neuronal ceroid lipofuscinosis (LINCL) is an autosomal recessive lysosomal storage disorder caused by mutations in the Cln2 gene leading to deficiency of lysosomal enzyme tripeptidyl peptidase 1 (TPP1). At present, available options for this fatal disorder are enzyme replacement therapy and gene therapy, which are extensively invasive and expensive. Our study demonstrates that 3-hydroxy-(2,2)-dimethyl butyrate (HDMB), a brain endogenous molecule, is capable of stimulating TPP1 expression and activity in mouse primary astrocytes and a neuronal cell line. HDMB activated peroxisome proliferator-activated receptor-α (PPARα), which, by forming heterodimer with Retinoid X receptor-α (RXRα), transcriptionally upregulated the Cln2 gene. Moreover, by using primary astrocytes from wild type, PPARα-/- and PPARβ-/- mice, we demonstrated that HDMB specifically required PPARα for inducing TPP1 expression. Finally, oral administration of HDMB to Cln2 heterozygous (Cln2+/-) mice led to a marked upregulation of TPP1 expression in the motor cortex and striatum in a PPARα-dependent fashion. Our study suggests that HDMB, a brain endogenous ligand of PPARα, might have therapeutic importance for LINCL treatment.
Collapse
Affiliation(s)
- Sudipta Chakrabarti
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Sujyoti Chandra
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA; Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| | - Sridevi Dasarathi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Madhuchhanda Kundu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA; Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA.
| |
Collapse
|
24
|
Lu L, Michelena TM, Wong A, Zhang CJ, Meng Y. The inhibition of acetylcholinesterase by a brain-targeting polylysine-ApoE peptide: biochemical and structural characterizations. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2018:155-158. [PMID: 30440362 DOI: 10.1109/embc.2018.8512202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The in-trans delivery of protein therapeutics across the blood-brain barrier by K16ApoE peptide carrier has been demonstrated to improve the neurological symptoms and increase the life-span of late-infantile neuronal ceroid lipofuscinosis (LINCL) mice. However, acute toxicity of K16ApoE was observed in LINCL mice resulting in a narrow therapeutic index, limiting the potential of translating the K16ApoE into a viable drug delivery system. This study aims to unravel the toxic mechanism of action. We hypothesized that the toxic response towards the peptide was induced by inhibition of acetylcholinesterase (AChE) activity at neuro-muscular junction. Here, results from the dose-response study suggested that AChE activity was inhibited by K16ApoE at either low or high doses but not at the mid-dose where a significant increase in AChE activity was observed. Meanwhile, molecular docking simulations showed that the N-terminus of K16ApoE is capable of binding to the active site gorge of AChE. In addition to a favorable spatial orientation, this docking pose also revealed strong surface charge interactions which may account for the observed inhibitory effect. While statistical analysis of the dose response and survival ratio suggested that AChE is not the primary mechanism of action for the acute toxicity of K16ApoE, both biochemical evidence and structural analysis have assigned indirect but critical roles for AChE in the overall toxicity mechanism of this peptide carrier.
Collapse
|
25
|
Katz N, Goode T, Hinderer C, Hordeaux J, Wilson JM. Standardized Method for Intra-Cisterna Magna Delivery Under Fluoroscopic Guidance in Nonhuman Primates. Hum Gene Ther Methods 2018; 29:212-219. [PMID: 30032644 DOI: 10.1089/hgtb.2018.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Intrathecal delivery of adeno-associated virus vectors and other therapeutics are currently being evaluated for the treatment of central nervous system sequelae of lysosomal storage diseases, motor neuron diseases, and neurodegenerative diseases. As products transition from preclinical to clinical studies, a standardized and clinically relevant method of intrathecal delivery is increasingly germane. Here, we describe a method of intrathecal delivery via suboccipital puncture into the cisterna magna under fluoroscopic guidance in nonhuman primates. This procedure is suitable for use in good laboratory practice compliant studies, has an excellent safety profile, and is highly similar to the procedure currently being explored for use in humans.
Collapse
Affiliation(s)
- Nathan Katz
- Gene Therapy Program, Department of Medicine, University of Pennsylvania , Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Tamara Goode
- Gene Therapy Program, Department of Medicine, University of Pennsylvania , Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Christian Hinderer
- Gene Therapy Program, Department of Medicine, University of Pennsylvania , Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Juliette Hordeaux
- Gene Therapy Program, Department of Medicine, University of Pennsylvania , Perelman School of Medicine, Philadelphia, Pennsylvania
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, University of Pennsylvania , Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
26
|
Schulz A, Ajayi T, Specchio N, de Los Reyes E, Gissen P, Ballon D, Dyke JP, Cahan H, Slasor P, Jacoby D, Kohlschütter A. Study of Intraventricular Cerliponase Alfa for CLN2 Disease. N Engl J Med 2018; 378:1898-1907. [PMID: 29688815 DOI: 10.1056/nejmoa1712649] [Citation(s) in RCA: 299] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Recombinant human tripeptidyl peptidase 1 (cerliponase alfa) is an enzyme-replacement therapy that has been developed to treat neuronal ceroid lipofuscinosis type 2 (CLN2) disease, a rare lysosomal disorder that causes progressive dementia in children. METHODS In a multicenter, open-label study, we evaluated the effect of intraventricular infusion of cerliponase alfa every 2 weeks in children with CLN2 disease who were between the ages of 3 and 16 years. Treatment was initiated at a dose of 30 mg, 100 mg, or 300 mg; all the patients then received the 300-mg dose for at least 96 weeks. The primary outcome was the time until a 2-point decline in the score on the motor and language domains of the CLN2 Clinical Rating Scale (which ranges from 0 to 6, with 0 representing no function and 3 representing normal function in each of the two domains), which was compared with the time until a 2-point decline in 42 historical controls. We also compared the rate of decline in the motor-language score between the two groups, using data from baseline to the last assessment with a score of more than 0, divided by the length of follow-up (in units of 48 weeks). RESULTS Twenty-four patients were enrolled, 23 of whom constituted the efficacy population. The median time until a 2-point decline in the motor-language score was not reached for treated patients and was 345 days for historical controls. The mean (±SD) unadjusted rate of decline in the motor-language score per 48-week period was 0.27±0.35 points in treated patients and 2.12±0.98 points in 42 historical controls (mean difference, 1.85; P<0.001). Common adverse events included convulsions, pyrexia, vomiting, hypersensitivity reactions, and failure of the intraventricular device. In 2 patients, infections developed in the intraventricular device that was used to administer the infusion, which required antibiotic treatment and device replacement. CONCLUSIONS Intraventricular infusion of cerliponase alfa in patients with CLN2 disease resulted in less decline in motor and language function than that in historical controls. Serious adverse events included failure of the intraventricular device and device-related infections. (Funded by BioMarin Pharmaceutical and others; CLN2 ClinicalTrials.gov numbers, NCT01907087 and NCT02485899 .).
Collapse
Affiliation(s)
- Angela Schulz
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - Temitayo Ajayi
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - Nicola Specchio
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - Emily de Los Reyes
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - Paul Gissen
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - Douglas Ballon
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - Jonathan P Dyke
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - Heather Cahan
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - Peter Slasor
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - David Jacoby
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| | - Alfried Kohlschütter
- From the Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.S., A.K.); BioMarin Pharmaceutical, Novato, CA (T.A., H.C., P.S., D.J.); the Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome (N.S.); Nationwide Children's Hospital and Ohio State University, Columbus (E.L.R.); UCL Great Ormond Street Institute of Child Health, London (P.G.); and the Citigroup Biomedical Imaging Center, Departments of Radiology and Genetic Medicine, Weill Cornell Medical College, New York (D.B., J.P.D.)
| |
Collapse
|
27
|
Vuillemenot BR, Korte S, Wright TL, Adams EL, Boyd RB, Butt MT. Safety Evaluation of CNS Administered Biologics-Study Design, Data Interpretation, and Translation to the Clinic. Toxicol Sci 2018; 152:3-9. [PMID: 27354708 DOI: 10.1093/toxsci/kfw072] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Many central nervous system (CNS) diseases are inadequately treated by systemically administered therapies due to the blood brain barrier (BBB), which prevents achieving adequate drug concentrations at sites of action. Due to the increasing prevalence of neurodegenerative diseases and the inability of most systemically administered therapies to cross the BBB, direct CNS delivery will likely play an increasing role in treatment. Administration of large molecules, cells, viral vectors, oligonucleotides, and other novel therapies directly to the CNS via the subarachnoid space, ventricular system, or parenchyma overcomes this obstacle. Clinical experience with direct CNS administration of small molecule therapies suggests that this approach may be efficacious for the treatment of neurodegenerative disorders using biological therapies. Risks of administration into the brain tissue or cerebrospinal fluid include local damage from implantation of the delivery system and/or administration of the therapeutic and reactions affecting the CNS. Preclinical safety studies on CNS administered compounds must differentiate between the effects of the test article, the delivery device, and/or the vehicle, and assess exacerbations of reactions due to combinations of effects. Animal models characterized for safety assessment of CNS administered therapeutics have enabled human trials, but interpretation can be challenging. This manuscript outlines the challenges of preclinical intrathecal/intracerebroventricular/intraparenchymal studies, evaluation of results, considerations for special endpoints, and translation of preclinical findings to enable first-in-human trials. Recommendations will be made based on the authors' collective experience with conducting these studies to enable clinical development of CNS-administered biologics.
Collapse
Affiliation(s)
| | - Sven Korte
- Covance Laboratories GmbH, Münster, Germany
| | | | - Eric L Adams
- Northern Biomedical Research, Muskegon, Michigan
| | | | | |
Collapse
|
28
|
Katz ML, Rustad E, Robinson GO, Whiting REH, Student JT, Coates JR, Narfstrom K. Canine neuronal ceroid lipofuscinoses: Promising models for preclinical testing of therapeutic interventions. Neurobiol Dis 2017; 108:277-287. [PMID: 28860089 DOI: 10.1016/j.nbd.2017.08.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/26/2017] [Indexed: 10/19/2022] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are devastating inherited progressive neurodegenerative diseases, with most forms having a childhood onset of clinical signs. The NCLs are characterized by progressive cognitive and motor decline, vision loss, seizures, respiratory and swallowing impairment, and ultimately premature death. Different forms of NCL result from mutations in at least 13 genes. The clinical signs of some forms overlap significantly, so genetic testing is the only way to definitively determine which form an individual patient suffers from. At present, an effective treatment is available for only one form of NCL. Evidence of NCL has been documented in over 20 canine breeds and in mixed-breed dogs. To date, 12 mutations in 8 different genes orthologous to the human NCL genes have been found to underlie NCL in a variety of dog breeds. A Dachshund model with a null mutation in one of these genes is being utilized to investigate potential therapeutic interventions, including enzyme replacement and gene therapies. Demonstration of the efficacy of enzyme replacement therapy in this model led to successful completion of human clinical trials of this treatment. Further research into the other canine NCLs, with in-depth characterization and understanding of the disease processes, will likely lead to the development of successful therapeutic interventions for additional forms of NCL, for both human patients and animals with these disorders.
Collapse
Affiliation(s)
- Martin L Katz
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| | - Eline Rustad
- Blue Star Animal Hospital, Göteborg 417 07, Sweden
| | - Grace O Robinson
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Rebecca E H Whiting
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Jeffrey T Student
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Joan R Coates
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Kristina Narfstrom
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
29
|
Nelvagal HR, Cooper JD. Translating preclinical models of neuronal ceroid lipofuscinosis: progress and prospects. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1360182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Hemanth R. Nelvagal
- Pediatric Storage Disorders Laboratory, Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, David Geffen School of Medicine, UCLA, Torrance, CA, USA
| | - Jonathan D. Cooper
- Pediatric Storage Disorders Laboratory, Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, David Geffen School of Medicine, UCLA, Torrance, CA, USA
| |
Collapse
|
30
|
Specchio N, Bellusci M, Pietrafusa N, Trivisano M, de Palma L, Vigevano F. Photosensitivity is an early marker of neuronal ceroid lipofuscinosis type 2 disease. Epilepsia 2017. [DOI: 10.1111/epi.13820] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nicola Specchio
- Department of Neuroscience; Bambino Gesù Children's Hospital; IRCCS (Istituto di Ricerca e Cura a Carattere Scientifico); Rome Italy
| | - Marcello Bellusci
- Department of Neuroscience; Bambino Gesù Children's Hospital; IRCCS (Istituto di Ricerca e Cura a Carattere Scientifico); Rome Italy
- Pediatric Neurology; 12 October University Hospital; Madrid Spain
| | - Nicola Pietrafusa
- Department of Neuroscience; Bambino Gesù Children's Hospital; IRCCS (Istituto di Ricerca e Cura a Carattere Scientifico); Rome Italy
| | - Marina Trivisano
- Department of Neuroscience; Bambino Gesù Children's Hospital; IRCCS (Istituto di Ricerca e Cura a Carattere Scientifico); Rome Italy
| | - Luca de Palma
- Department of Neuroscience; Bambino Gesù Children's Hospital; IRCCS (Istituto di Ricerca e Cura a Carattere Scientifico); Rome Italy
| | - Federico Vigevano
- Department of Neuroscience; Bambino Gesù Children's Hospital; IRCCS (Istituto di Ricerca e Cura a Carattere Scientifico); Rome Italy
| |
Collapse
|
31
|
Morgan SJ, Couch J, Guzzie-Peck P, Keller DA, Kemper R, Otieno MA, Schulingkamp RJ, Jones TW. Regulatory Forum Opinion Piece *: Use and Utility of Animal Models of Disease for Nonclinical Safety Assessment: A Pharmaceutical Industry Survey. Toxicol Pathol 2017; 45:372-380. [PMID: 28351296 DOI: 10.1177/0192623317701004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
An Innovation and Quality (IQ) Consortium focus group conducted a cross-company survey to evaluate current practices and perceptions around the use of animal models of disease (AMDs) in nonclinical safety assessment of molecules in clinical development. The IQ Consortium group is an organization of pharmaceutical and biotechnology companies with the mission of advancing science and technology. The survey queried the utilization of AMDs during drug discovery in which drug candidates are evaluated in efficacy models and limited short-duration non-Good Laboratory Practices (GLP) toxicology testing and during drug development in which drug candidates are evaluated in GLP toxicology studies. The survey determined that the majority of companies used AMDs during drug discovery primarily as a means for proactively assessing potential nonclinical safety issues prior to the conduct of toxicology studies, followed closely by the use of AMDs to better understand toxicities associated with exaggerated pharmacology in traditional toxicology models or to derisk issues when the target is only expressed in the disease state. In contrast, the survey results indicated that the use of AMDs in development is infrequent, being used primarily to investigate nonclinical safety issues associated with targets expressed only in disease states and/or in response to requests from global regulatory authorities.
Collapse
Affiliation(s)
- Sherry J Morgan
- 1 AbbVie, Inc., Preclinical Safety, North Chicago, Illinois, USA
| | - Jessica Couch
- 2 Genentech, Inc., Department of Safety Assessment, South San Francisco, California, USA
| | - Peggy Guzzie-Peck
- 3 Janssen Research and Development, Preclinical Development and Safety, Spring House, Pennsylvania, USA
| | | | - Ray Kemper
- 5 Vertex Pharmaceuticals, Inc., Preclinical Safety Assessment, Boston, Massachusetts, USA
| | - Monicah A Otieno
- 3 Janssen Research and Development, Preclinical Development and Safety, Spring House, Pennsylvania, USA
| | | | - Thomas W Jones
- 7 Eli Lilly and Company, Toxicology and Pathology, Indianapolis, Indiana, USA
| |
Collapse
|
32
|
Wiseman JA, Meng Y, Nemtsova Y, Matteson PG, Millonig JH, Moore DF, Sleat DE, Lobel P. Chronic Enzyme Replacement to the Brain of a Late Infantile Neuronal Ceroid Lipofuscinosis Mouse Has Differential Effects on Phenotypes of Disease. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 4:204-212. [PMID: 28345005 PMCID: PMC5363315 DOI: 10.1016/j.omtm.2017.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/23/2017] [Indexed: 12/12/2022]
Abstract
Late infantile neuronal ceroid lipofuscinosis (LINCL) is a fatal inherited neurodegenerative disease caused by loss of lysosomal protease tripeptidyl peptidase 1 (TPP1). We have investigated the effects of chronic intrathecal (IT) administration using enzyme replacement therapy (ERT) to the brain of an LINCL mouse model, in which locomotor function declines dramatically prior to early death. Median lifespan was significantly extended from 126 days to >259 days when chronic IT treatment was initiated before the onset of disease. While treated animals lived longer and showed little sign of locomotor dysfunction as measured by stride length, some or all (depending on regimen) still died prematurely. One explanation is that cerebrospinal fluid (CSF)-mediated delivery may not deliver TPP1 to all brain regions. Morphological studies support this, showing delivery of TPP1 to ventral, but not deeper and dorsal regions. When IT treatment is initiated in severely affected LINCL mice, lifespan was extended modestly in most but dramatically extended in approximately one-third of the cohort. Treatment improved locomotor function in these severely compromised animals after it had declined to the point at which animals normally die. This indicates that some pathology in LINCL is reversible and does not simply reflect neuronal death.
Collapse
Affiliation(s)
- Jennifer A Wiseman
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Yu Meng
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Yuliya Nemtsova
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Paul G Matteson
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - James H Millonig
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Neuroscience & Cell Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Dirk F Moore
- School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - David E Sleat
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Peter Lobel
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
33
|
Extraneuronal pathology in a canine model of CLN2 neuronal ceroid lipofuscinosis after intracerebroventricular gene therapy that delays neurological disease progression. Gene Ther 2017; 24:215-223. [PMID: 28079862 PMCID: PMC5398942 DOI: 10.1038/gt.2017.4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/23/2016] [Accepted: 01/03/2017] [Indexed: 01/04/2023]
Abstract
CLN2 neuronal ceroid lipofuscinosis is a hereditary lysosomal storage disease with primarily neurological signs that results from mutations in TPP1, which encodes the lysosomal enzyme tripeptidyl peptidase-1 (TPP1). Studies using a canine model for this disorder demonstrated that delivery of TPP1 enzyme to the cerebrospinal fluid (CSF) by intracerebroventricular administration of an AAV-TPP1 vector resulted in substantial delays in the onset and progression of neurological signs and prolongation of life span. We hypothesized that the treatment may not deliver therapeutic levels of this protein to tissues outside the central nervous system that also require TPP1 for normal lysosomal function. To test this hypothesis, dogs treated with CSF administration of AAV-TPP1 were evaluated for the development of non-neuronal pathology. Affected treated dogs exhibited progressive cardiac pathology reflected by elevated plasma cardiac troponin-1, impaired cardiac function and development of histopathological myocardial lesions. Progressive increases in the plasma activity levels of alanine aminotransferase and creatine kinase indicated development of pathology in the liver and muscles. The treatment also did not prevent disease-related accumulation of lysosomal storage bodies in the heart or liver. These studies indicate that optimal treatment outcomes for CLN2 disease may require delivery of TPP1 systemically as well as directly to the central nervous system.
Collapse
|
34
|
Cohen-Pfeffer JL, Gururangan S, Lester T, Lim DA, Shaywitz AJ, Westphal M, Slavc I. Intracerebroventricular Delivery as a Safe, Long-Term Route of Drug Administration. Pediatr Neurol 2017; 67:23-35. [PMID: 28089765 DOI: 10.1016/j.pediatrneurol.2016.10.022] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/07/2016] [Accepted: 10/30/2016] [Indexed: 01/19/2023]
Abstract
Intrathecal delivery methods have been used for many decades to treat a broad range of central nervous system disorders. A literature review demonstrated that intracerebroventricular route is an established and well-tolerated method for prolonged central nervous system drug delivery in pediatric and adult populations. Intracerebroventricular devices were present in patients from one to 7156 days. The number of punctures per device ranged from 2 to 280. Noninfectious complication rates per patient (range, 1.0% to 33.0%) were similar to infectious complication rates (0.0% to 27.0%). Clinician experience and training and the use of strict aseptic techniques have been shown to reduce the frequency of complications.
Collapse
Affiliation(s)
| | | | | | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | | | - Manfred Westphal
- Department of Neurosurgery, University Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Irene Slavc
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
35
|
Current Strategies for the Delivery of Therapeutic Proteins and Enzymes to Treat Brain Disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 137:1-28. [DOI: 10.1016/bs.irn.2017.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
36
|
Intravitreal implantation of TPP1-transduced stem cells delays retinal degeneration in canine CLN2 neuronal ceroid lipofuscinosis. Exp Eye Res 2016; 152:77-87. [DOI: 10.1016/j.exer.2016.09.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 11/19/2022]
|
37
|
Dawson G. Quantum dots and potential therapy for Krabbe's disease. J Neurosci Res 2016; 94:1293-303. [PMID: 27638611 PMCID: PMC5027984 DOI: 10.1002/jnr.23805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/27/2022]
Abstract
Enzyme replacement therapy and substrate reduction therapy have proved useful in reversing many pathological consequences of many nonneural lysosomal storage diseases but have not yet reversed pathology or influenced disease outcome in Krabbe's disease (KD). This Review discusses the relative merits of stem cell therapy, molecular chaperone therapy, gene therapy, substrate reduction therapy, enzyme replacement therapy, and combination therapy. Given the limitations of these approaches, this Review introduces the idea of using tiny, 6-nm, intensely fluorescent quantum dots (QDs) to deliver a cell-penetrating peptide and 6 histidine residue-tagged β-D-galactocerebrosidase across the blood-brain barrier. We can therefore follow the fate of injected material and ensure that all targets are reached and that accumulated material is degraded. Uptake of lysosomal hydrolases is a complex process, and the cell-penetrating peptide JB577 is uniquely able to promote endosomal egress of the QD cargo. This Review further shows that uptake may depend on the charge of the coating of the QD, specifically, that negative charge directs the cargo to neurons. Because KD involves primarily glia, specifically oligodendroglia, we experiment with many coatings and discover a coating (polyethylene glycol 600 amino) that has a positive charge and targets oligodendrocytes. A similar effect is achieved by treating with chondroitinase ABC to degrade the extracellular matrix, indicating that enzyme replacement has several hurdles to overcome before it can become a routine CNS therapy. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Glyn Dawson
- Department of Pediatrics, University of Chicago, Chicago, Illinois.
| |
Collapse
|
38
|
Katz ML, Tecedor L, Chen Y, Williamson BG, Lysenko E, Wininger FA, Young WM, Johnson GC, Whiting REH, Coates JR, Davidson BL. AAV gene transfer delays disease onset in a TPP1-deficient canine model of the late infantile form of Batten disease. Sci Transl Med 2016; 7:313ra180. [PMID: 26560358 DOI: 10.1126/scitranslmed.aac6191] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The most common form of the childhood neurodegenerative disease late infantile neuronal ceroid lipofuscinosis (also called Batten disease) is caused by deficiency of the soluble lysosomal enzyme tripeptidyl peptidase 1 (TPP1) resulting from mutations in the TPP1 gene. We tested whether TPP1 gene transfer to the ependyma, the epithelial lining of the brain ventricular system, in TPP1-deficient dogs would be therapeutically beneficial. A one-time administration of recombinant adeno-associated virus (rAAV) expressing canine TPP1 (rAAV.caTPP1) resulted in high expression of TPP1 predominantly in ependymal cells and secretion of the enzyme into the cerebrospinal fluid leading to clinical benefit. Diseased dogs treated with rAAV.caTPP1 showed delays in onset of clinical signs and disease progression, protection from cognitive decline, and extension of life span. By immunostaining and enzyme assay, recombinant protein was evident throughout the brain and spinal cord, with correction of the neuropathology characteristic of the disease. This study in a naturally occurring canine model of TPP1 deficiency highlights the utility of AAV transduction of ventricular lining cells to accomplish stable secretion of recombinant protein for broad distribution in the central nervous system and therapeutic benefit.
Collapse
Affiliation(s)
- Martin L Katz
- Department of Ophthalmology, University of Missouri, Columbia, MO 65212, USA
| | - Luis Tecedor
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yonghong Chen
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Baye G Williamson
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Elena Lysenko
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Fred A Wininger
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Whitney M Young
- Department of Ophthalmology, University of Missouri, Columbia, MO 65212, USA
| | - Gayle C Johnson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Rebecca E H Whiting
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Joan R Coates
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Beverly L Davidson
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA. Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Lu JY, Nelvagal HR, Wang L, Birnbaum SG, Cooper JD, Hofmann SL. Intrathecal enzyme replacement therapy improves motor function and survival in a preclinical mouse model of infantile neuronal ceroid lipofuscinosis. Mol Genet Metab 2015; 116:98-105. [PMID: 25982063 DOI: 10.1016/j.ymgme.2015.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/09/2015] [Accepted: 05/10/2015] [Indexed: 11/29/2022]
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are a group of related hereditary lysosomal storage disorders characterized by progressive loss of neurons in the central nervous system resulting in dementia, loss of motor skills, seizures and blindness. A characteristic intralysosomal accumulation of autofluorescent storage material occurs in the brain and other tissues. Three major forms and nearly a dozen minor forms of NCL are recognized. Infantile-onset NCL (CLN1 disease) is caused by severe deficiency in a soluble lysosomal enzyme, palmitoyl-protein thioesterase-1 (PPT1) and no therapy beyond supportive care is available. Homozygous Ppt1 knockout mice reproduce the known features of the disease, developing signs of motor dysfunction at 5 months of age and death around 8 months. Direct delivery of lysosomal enzymes to the cerebrospinal fluid is an approach that has gained traction in small and large animal models of several other neuropathic lysosomal storage diseases, and has advanced to clinical trials. In the current study, Ppt1 knockout mice were treated with purified recombinant human PPT1 enzyme delivered to the lumbar intrathecal space on each of three consecutive days at 6 weeks of age. Untreated PPT1 knockout mice and wild-type mice served as additional controls. Four enzyme concentration levels (0, 2.6, 5.3 and 10.6 mg/ml of specific activity 20 U/mg) were administered in a volume of 80 μl infused over 8 min. Each group consisted of 16-20 mice. The treatment was well tolerated. Disease-specific survival was 233, 267, 272, and 284days for each of the four treatment groups, respectively, and the effect of treatment was highly significant (p<0.0001). The timing of motor deterioration was also delayed. Neuropathology was improved as evidenced by decreased autofluorescent storage material in the spinal cord and a decrease in CD68 staining in the cortex and spinal cord. The improvements in motor function and survival are similar to results reported for preclinical studies involving other lysosomal storage disorders, such as CLN2/TPP1 deficiency, for which intraventricular ERT is being offered in clinical trials. If ERT delivery to the CSF proves to be efficacious in these disorders, PPT1 deficiency may also be amenable to this approach.
Collapse
Affiliation(s)
- Jui-Yun Lu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8593, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-8593, USA
| | - Hemanth R Nelvagal
- Pediatric Storage Disorders Laboratory, Department of Basic and Clinical Neuroscience, King's Health Partners Centre for Neurodegeneration, James Black Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Lingling Wang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8593, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-8593, USA
| | - Shari G Birnbaum
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390-8593, USA
| | - Jonathan D Cooper
- Pediatric Storage Disorders Laboratory, Department of Basic and Clinical Neuroscience, King's Health Partners Centre for Neurodegeneration, James Black Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Sandra L Hofmann
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8593, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-8593, USA.
| |
Collapse
|
40
|
|
41
|
Abstract
Developing therapies for the brain is perhaps the greatest challenge facing modern medicine today. While a great many potential therapies show promise in animal models, precious few make it to approval or are even studied in human patients. The particular challenges to the translation of neurotherapeutics to the clinic are many, but a major barrier is difficulty in delivering therapeutics into the brain. The goal of this workshop was to present ways to deliver therapeutics to the brain, including the limitations of each method, and describe ways to track their delivery, safety, and efficacy. Solving the problem of delivery will aid translation of therapeutics for patients suffering from neurodegeneration and other disorders of the brain.
Collapse
Affiliation(s)
- Patricia I Dickson
- Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, 1124 W. Carson Street, HH1, Torrance, CA, 90502, USA,
| |
Collapse
|
42
|
King B, Marshall N, Beard H, Hassiotis S, Trim PJ, Snel MF, Rozaklis T, Jolly RD, Hopwood JJ, Hemsley KM. Evaluation of enzyme dose and dose-frequency in ameliorating substrate accumulation in MPS IIIA Huntaway dog brain. J Inherit Metab Dis 2015; 38:341-50. [PMID: 25421091 DOI: 10.1007/s10545-014-9790-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 10/08/2014] [Accepted: 10/29/2014] [Indexed: 12/18/2022]
Abstract
Intracerebrospinal fluid (CSF) infusion of replacement enzyme is under evaluation for amelioration of disease-related symptoms and biomarker changes in patients with the lysosomal storage disorder mucopolysaccharidosis type IIIA (MPS IIIA; www.clinicaltrials.gov ; NCT#01155778; #01299727). Determining the optimal dose/dose-frequency is important, given the invasive method for chronically supplying recombinant protein to the brain, the main site of symptom generation. To examine these variables, we utilised MPS IIIA Huntaway dogs, providing recombinant human sulphamidase (rhSGSH) to young pre-symptomatic dogs from an age when MPS IIIA dog brain exhibits significant accumulation of primary (heparan sulphate) and secondary (glycolipid) substrates. Enzyme was infused into CSF via the cisterna magna at one of two doses (3 mg or 15 mg/infusion), with the higher dose supplied at two different intervals; fortnightly or monthly. Euthanasia was carried out 24 h after the final injection. Dose- and frequency-dependent reductions in heparan sulphate were observed in CSF and deeper layers of cerebral cortex. When we examined the amount of immunostaining of the general endo/lysosomal marker, LIMP-2, or quantified activated microglia, the higher fortnightly dose resulted in superior outcomes in affected dogs. Secondary lesions such as accumulation of GM3 ganglioside and development of GAD-reactive axonal spheroids were treated to a similar degree by both rhSGSH doses and dose frequencies. Our findings indicate that the lower fortnightly dose is sub-optimal for ameliorating existing and preventing further development of disease-related pathology in young MPS IIIA dog brain; however, increasing the dose fivefold but halving the frequency of administration enabled near normalisation of disease-related biomarkers.
Collapse
Affiliation(s)
- Barbara King
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, SA, 5001, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Multifocal retinopathy in Dachshunds with CLN2 neuronal ceroid lipofuscinosis. Exp Eye Res 2015; 134:123-32. [PMID: 25697710 DOI: 10.1016/j.exer.2015.02.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 02/10/2015] [Accepted: 02/12/2015] [Indexed: 11/24/2022]
Abstract
The CLN2 form of neuronal ceroid lipofuscinosis is an autosomal recessively inherited lysosomal storage disease that is characterized by progressive vision loss culminating in blindness, cognitive and motor decline, neurodegeneration, and premature death. CLN2 disease results from mutations in the gene that encodes the soluble lysosomal enzyme tripeptidyl peptidase-1. A null mutation in the TPP1 gene encoding this enzyme causes a CLN2-like disease in Dachshunds. Dachshunds that are homozygous for this mutation serve as a model for human CLN2 disease, exhibiting clinical signs and neuropathology similar to those of children with this disorder. Affected dogs reach end-stage terminal disease status at 10-11 months of age. In addition to retinal changes typical of CLN2 disease, a retinopathy consisting of multifocal, bullous retinal detachment lesions was identified in 65% of (TPP1-/-) dogs in an established research colony. These lesions did not occur in littermates that were heterozygous or homozygous for the normal TPP1 allele. Retinal changes and the functional effects of this multifocal retinopathy were examined objectively over time using ophthalmic examinations, fundus photography, electroretinography (ERG), quantitative pupillary light response (PLR) recording, fluorescein angiography, optical coherence tomography (OCT) and histopathology. The retinopathy consisted of progressive multifocal serous retinal detachments. The severity of the disease-related retinal thinning was no more serious in most detached areas than in adjacent areas of the retina that remained in close apposition to the retinal pigment epithelium. The retinopathy observed in these dogs was somewhat similar to canine multifocal retinopathy (CMR), a disease caused by a mutation of the bestrophin gene BEST1. ERG a-wave amplitudes were relatively preserved in the Dachshunds with CLN2 disease, whether or not they developed the multifocal retinopathy. The retinopathy also had minimal effects on the PLR. Histological evaluation indicated that the CLN2 disease-related retinal degeneration was not exacerbated in areas where the retina was detached except where the detached areas were very large. DNA sequence analysis ruled out a mutation in the BEST1 exons or splice junctions as a cause for the retinopathy. Perfect concordance between the TPP1 mutation and the retinopathy in the large number of dogs examined indicates that the retinopathy most likely occurs as a direct result of the TPP1 mutation. Therefore, inhibition of the development and progression of these lesions can be used as an indicator of the efficacy of therapeutic interventions currently under investigation for the treatment of CLN2 disease in the Dachshund model. In addition, these findings suggest that TPP1 mutations may underlie multifocal retinopathies of unknown cause in animals and humans.
Collapse
|
44
|
Vuillemenot BR, Kennedy D, Cooper JD, Wong AMS, Sri S, Doeleman T, Katz ML, Coates JR, Johnson GC, Reed RP, Adams EL, Butt MT, Musson DG, Henshaw J, Keve S, Cahayag R, Tsuruda LS, O'Neill CA. Nonclinical evaluation of CNS-administered TPP1 enzyme replacement in canine CLN2 neuronal ceroid lipofuscinosis. Mol Genet Metab 2015; 114:281-93. [PMID: 25257657 DOI: 10.1016/j.ymgme.2014.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/02/2014] [Accepted: 09/03/2014] [Indexed: 11/16/2022]
Abstract
The CLN2 form of neuronal ceroid lipofuscinosis, a type of Batten disease, is a lysosomal storage disorder caused by a deficiency of the enzyme tripeptidyl peptidase-1 (TPP1). Patients exhibit progressive neurodegeneration and loss of motor, cognitive, and visual functions, leading to death by the early teenage years. TPP1-null Dachshunds recapitulate human CLN2 disease. To characterize the safety and pharmacology of recombinant human (rh) TPP1 administration to the cerebrospinal fluid (CSF) as a potential enzyme replacement therapy (ERT) for CLN2 disease, TPP1-null and wild-type (WT) Dachshunds were given repeated intracerebroventricular (ICV) infusions and the pharmacokinetic (PK) profile, central nervous system (CNS) distribution, and safety were evaluated. TPP1-null animals and WT controls received 4 or 16mg of rhTPP1 or artificial cerebrospinal fluid (aCSF) vehicle every other week. Elevated CSF TPP1 concentrations were observed for 2-3 days after the first ICV infusion and were approximately 1000-fold higher than plasma levels at the same time points. Anti-rhTPP1 antibodies were detected in CSF and plasma after repeat rhTPP1 administration, with titers generally higher in TPP1-null than in WT animals. Widespread brain distribution of rhTPP1 was observed after chronic administration. Expected histological changes were present due to the CNS delivery catheters and were similar in rhTPP1 and vehicle-treated animals, regardless of genotype. Neuropathological evaluation demonstrated the clearance of lysosomal storage, preservation of neuronal morphology, and reduction in brain inflammation with treatment. This study demonstrates the favorable safety and pharmacology profile of rhTPP1 ERT administered directly to the CNS and supports clinical evaluation in patients with CLN2 disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Steve Keve
- BioMarin Pharmaceutical Inc., Novato, CA, USA
| | | | | | | |
Collapse
|
45
|
Guo J, O'Brien DP, Mhlanga-Mutangadura T, Olby NJ, Taylor JF, Schnabel RD, Katz ML, Johnson GS. A rare homozygous MFSD8 single-base-pair deletion and frameshift in the whole genome sequence of a Chinese Crested dog with neuronal ceroid lipofuscinosis. BMC Vet Res 2015; 10:960. [PMID: 25551667 PMCID: PMC4298050 DOI: 10.1186/s12917-014-0181-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 08/06/2014] [Indexed: 02/02/2023] Open
Abstract
Background The neuronal ceroid lipofuscinoses are heritable lysosomal storage diseases characterized by progressive neurological impairment and the accumulation of autofluorescent storage granules in neurons and other cell types. Various forms of human neuronal ceroid lipofuscinosis have been attributed to mutations in at least 13 different genes. So far, mutations in the canine orthologs of 7 of these genes have been identified in DNA from dogs with neuronal ceroid lipofuscinosis. The identification of new causal mutations could lead to the establishment of canine models to investigate the pathogenesis of the corresponding human neuronal ceroid lipofuscinoses and to evaluate and optimize therapeutic interventions for these fatal human diseases. Case presentation We obtained blood and formalin-fixed paraffin-embedded brain sections from a rescue dog that was reported to be a young adult Chinese Crested. The dog was euthanized at approximately 19 months of age as a consequence of progressive neurological decline that included blindness, anxiety, and cognitive impairment. A diagnosis of neuronal ceroid lipofuscinosis was made based on neurological signs, magnetic resonance imaging of the brain, and fluorescence microscopic and electron microscopic examination of brain sections. We isolated DNA from the blood and used it to generate a whole genome sequence with 33-fold average coverage. Among the 7.2 million potential sequence variants revealed by aligning the sequence reads to the canine genome reference sequence was a homozygous single base pair deletion in the canine ortholog of one of 13 known human NCL genes: MFSD8:c.843delT. MFSD8:c.843delT is predicted to cause a frame shift and premature stop codon resulting in a truncated protein, MFSD8:p.F282Lfs13*, missing its 239 C-terminal amino acids. The MFSD8:c.843delT allele is absent from the whole genome sequences of 101 healthy canids or dogs with other diseases. The genotyping of archived DNA from 1478 Chinese Cresteds did not identify any additional MFSD8:c.843delT homozygotes and found only one heterozygote. Conclusion We conclude that the neurodegenerative disease of the Chinese Crested rescue dog was neuronal ceroid lipofuscinosis and that homozygosity for the MFSD8:c.843delT sequence variant was very likely to be the molecular-genetic cause of the disease. Electronic supplementary material The online version of this article (doi:10.1186/s12917-014-0181-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juyuan Guo
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| | - Dennis P O'Brien
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| | - Tendai Mhlanga-Mutangadura
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| | - Natasha J Olby
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| | - Jeremy F Taylor
- Division of Animal Science, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, MO, USA.
| | - Robert D Schnabel
- Division of Animal Science, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, MO, USA.
| | - Martin L Katz
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO, USA.
| | - Gary S Johnson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
46
|
A Hitchhiker's guide to the blood-brain barrier: in trans delivery of a therapeutic enzyme. Mol Ther 2014; 22:483-484. [PMID: 24584077 DOI: 10.1038/mt.2014.12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
47
|
Abstract
Aging dogs and cats show neurodegenerative features that are similar to human aging and Alzheimer disease. Neuropathologic changes with age may be linked to signs of cognitive dysfunction both in the laboratory and in a clinic setting. Less is known about cat brain aging and cognition and this represents an area for further study. Neurodegenerative diseases such as lysosomal storage diseases in dogs and cats also show similar features of human aging, suggesting some common underlying pathogenic mechanisms and also suggesting pathways that can be modified to promote healthy brain aging.
Collapse
Affiliation(s)
- Charles H Vite
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Section of Neurology & Neurosurgery, Department of Clinical Studies - Philadelphia, 3900 Delancey Street, Philadelphia, PA 19104, USA
| | - Elizabeth Head
- Department of Pharmacology & Nutritional Sciences, Sanders-Brown Center on Aging, University of Kentucky, 800 South Limestone Street, 203 Sanders Brown Building, Lexington, KY 40515, USA.
| |
Collapse
|
48
|
Guo J, Johnson GS, Brown HA, Provencher ML, da Costa RC, Mhlanga-Mutangadura T, Taylor JF, Schnabel RD, O'Brien DP, Katz ML. A CLN8 nonsense mutation in the whole genome sequence of a mixed breed dog with neuronal ceroid lipofuscinosis and Australian Shepherd ancestry. Mol Genet Metab 2014; 112:302-9. [PMID: 24953404 DOI: 10.1016/j.ymgme.2014.05.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 05/28/2014] [Accepted: 05/28/2014] [Indexed: 01/03/2023]
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are hereditary neurodegenerative diseases characterized by seizures and progressive cognitive decline, motor impairment, and vision loss accompanied by accumulation of autofluorescent lysosomal storage bodies in the central nervous system and elsewhere in the body. Mutations in at least 14 genes underlie the various forms of NCL. One of these genes, CLN8, encodes an intrinsic membrane protein of unknown function that appears to be localized primarily to the endoplasmic reticulum. Most CLN8 mutations in people result in a form of NCL with a late infantile onset and relatively rapid progression. A mixed breed dog with Australian Shepherd and Blue Heeler ancestry developed neurological signs characteristic of NCL starting at about 8months of age. The signs became progressively worse and the dog was euthanized at 21months of age due to seizures of increasing frequency and severity. Postmortem examination of the brain and retinas identified massive accumulations of intracellular autofluorescent inclusions characteristic of the NCLs. Whole genome sequencing of DNA from this dog identified a CLN8:c.585G>A transition that predicts a CLN8:p.Trp195* nonsense mutation. This mutation appears to be rare in both ancestral breeds. All of our 133 archived DNA samples from Blue Heelers, and 1481 of our 1488 archived Australian Shepherd DNA samples tested homozygous for the reference CLN8:c.585G allele. Four of the Australian Shepherd samples tested heterozygous and 3 tested homozygous for the mutant CLN8:c.585A allele. All 3 dogs homozygous for the A allele exhibited clinical signs of NCL and in 2 of them NCL was confirmed by postmortem evaluation of brain tissue. The occurrence of confirmed NCL in 3 of 4 CLN8:c.585A homozygous dogs, plus the occurrence of clinical signs consistent with NCL in the fourth homozygote strongly suggests that this rare truncating mutation causes NCL. Identification of this NCL-causing mutation provides the opportunity for identifying dogs that can be used to establish a canine model for the CLN8 disease (also known as late infantile variant or late infantile CLN8 disease).
Collapse
Affiliation(s)
- Juyuan Guo
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Gary S Johnson
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Holly A Brown
- Metz Petz Veterinary Clinic at Shawnee, Lima, OH, USA
| | - Michele L Provencher
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, USA
| | - Ronaldo C da Costa
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, USA
| | - Tendai Mhlanga-Mutangadura
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Jeremy F Taylor
- Division of Animal Science, University of Missouri College of Agriculture, Food and Natural Resources, Columbia, MO, USA
| | - Robert D Schnabel
- Division of Animal Science, University of Missouri College of Agriculture, Food and Natural Resources, Columbia, MO, USA
| | - Dennis P O'Brien
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Martin L Katz
- Mason Eye Institute, University of Missouri School of Medicine, Columbia, MO, USA.
| |
Collapse
|
49
|
Whiting RE, Narfström K, Yao G, Pearce JW, Coates JR, Castaner LJ, Jensen CA, Dougherty BN, Vuillemenot BR, Kennedy D, O'Neill CA, Katz ML. Enzyme replacement therapy delays pupillary light reflex deficits in a canine model of late infantile neuronal ceroid lipofuscinosis. Exp Eye Res 2014; 125:164-72. [DOI: 10.1016/j.exer.2014.06.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/05/2014] [Accepted: 06/09/2014] [Indexed: 12/12/2022]
|
50
|
Agile delivery of protein therapeutics to CNS. J Control Release 2014; 190:637-63. [PMID: 24956489 DOI: 10.1016/j.jconrel.2014.06.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/10/2014] [Accepted: 06/13/2014] [Indexed: 12/11/2022]
Abstract
A variety of therapeutic proteins have shown potential to treat central nervous system (CNS) disorders. Challenge to deliver these protein molecules to the brain is well known. Proteins administered through parenteral routes are often excluded from the brain because of their poor bioavailability and the existence of the blood-brain barrier (BBB). Barriers also exist to proteins administered through non-parenteral routes that bypass the BBB. Several strategies have shown promise in delivering proteins to the brain. This review, first, describes the physiology and pathology of the BBB that underscore the rationale and needs of each strategy to be applied. Second, major classes of protein therapeutics along with some key factors that affect their delivery outcomes are presented. Third, different routes of protein administration (parenteral, central intracerebroventricular and intraparenchymal, intranasal and intrathecal) are discussed along with key barriers to CNS delivery associated with each route. Finally, current delivery strategies involving chemical modification of proteins and use of particle-based carriers are overviewed using examples from literature and our own work. Whereas most of these studies are in the early stage, some provide proof of mechanism of increased protein delivery to the brain in relevant models of CNS diseases, while in few cases proof of concept had been attained in clinical studies. This review will be useful to broad audience of students, academicians and industry professionals who consider critical issues of protein delivery to the brain and aim developing and studying effective brain delivery systems for protein therapeutics.
Collapse
|