Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Dec 22.
Published in final edited form as: Expert Opin Orphan Drugs. 2019 Nov 27;7(11):473–500. doi: 10.1080/21678707.2019.1684258

Advances in the Treatment of Neuronal Ceroid Lipofuscinosis

Jonathan B Rosenberg 1,*, Alvin Chen 1,*, Stephen M Kaminsky 1, Ronald G Crystal 1, Dolan Sondhi 1
PMCID: PMC7755158  NIHMSID: NIHMS1653596  PMID: 33365208

Abstract

Neuronal ceroid lipofuscinoses (NCL) represent a class of neurodegenerative disorders involving defective lysosomal processing enzymes or receptors, leading to lysosomal storage disorders, typically characterized by observation of cognitive and visual impairments, epileptic seizures, ataxia, and deterioration of motor skills. Recent success of a biologic (Brineura®) for the treatment of neurologic manifestations of the central nervous system (CNS) has led to renewed interest in therapeutics for NCL, with the goal of ablating or reversing the impact of these devastating disorders. Despite complex challenges associated with CNS therapy, many treatment modalities have been evaluated, including enzyme replacement therapy, gene therapy, stem cell therapy, and small molecule pharmacotherapy. Because the clinical endpoints for the evaluation of candidate therapies are complex and often reliant on subjective clinical scales, the development of quantitative biomarkers for NCLs has become an apparent necessity for the validation of potential treatments. We will discuss the latest findings in the search for relevant biomarkers for assessing disease progression. For this review, we will focus primarily on recent pre-clinical and clinical developments for treatments to halt or cure these NCL diseases. Continued development of current therapies and discovery of newer modalities will be essential for successful therapeutics for NCL.

Areas covered:

The reader will be introduced to the NCL subtypes, natural histories, experimental animal models, and biomarkers for NCL progression; challenges and different therapeutic approaches, and the latest pre-clinical and clinical research for therapeutic development for the various NCLs. This review corresponds to the literatures covering the years from 1968 to mid-2019, but primarily addresses pre-clinical and clinical developments for the treatment of NCL disease in the last decade and as a follow-up to our 2013 review of the same topic in this journal.

Expert opinion:

Much progress has been made in the treatment of neurologic diseases, such as the NCLs, including better animal models and improved therapeutics with better survival outcomes. Encouraging results are being reported at symposiums and in the literature, with multiple therapeutics reaching the clinical trial stage for the NCLs. The potential for a cure could be at hand after many years of trial and error in the preclinical studies. The clinical development of enzyme replacement therapy (Brineura® for CLN2), immunosuppression (CellCept® for CLN3), and gene therapy vectors (for CLN1, CLN2, CLN3, and CLN6) are providing encouragement to families that have a child afflicted with NCL. We believe that successful therapies in the future may involve the combination of two or more therapeutic modalities to provide therapeutic benefit especially as the patients grow older.

Keywords: Neuronal ceroid lipofuscinoses (NCL); ceroid lipofuscinosis, neuronal (CLN); palmitoyl protein thioesterase-1 (PPT1); tripeptidyl peptidase-1 (TPP1); enzyme replacement therapy (ERT); gene therapy; adeno-associated virus (AAV); clinical trials

1. Introduction to Neuronal Ceroid Lipofuscinoses

Neuronal ceroid lipofuscinoses (NCL) are a collection of 13 autosomal recessive and 1 autosomal dominant neurologic disorders involving defective lysosomal processing enzymes or receptors, leading to lysosomal storage disorders16. Similar to other neurologic diseases, the disruption of normal lysosomal function leads to a toxic buildup of protein aggregates within the central nervous system (CNS), and is a major contributor to NCL pathologies7. The unique feature of NCL is the aggregation of lipopigments (lipofuscin) in lysosomes5,8,9, which appear in histopathology as fine yellow-brown pigment autofluorescent granules, composed of a mixture of lipids and protein, including protein subunit c of mitochondrial ATP synthase (SCMAS) and sphingolipid activator proteins (SAP) A and D, among others (Table I)1014. While the CNS is dramatically affected in all NCL patients, many have additional eye pathology (retina and ganglion cells), and other NCL mutations result in systemic pathology of visceral organs such as liver, heart, spleen, adrenals, and kidney7,15,16. While the buildup of lipofuscin aggregates in neurons occurs slowly as a secondary consequence of the normal aging process5,14, it is rapidly advanced in NCL patients. As the lipofuscin aggregates in the lysosomes, it alters the neuronal cytoskeleton and cellular trafficking, creating a pathologic state resulting in neuronal loss, and glial proliferation and activation7.

Table I.

Identifying Features of Neuronal Ceroid Lipofuscinoses (NCL) 1

Neuronal ceroid lipofuscinoses type Gene/protein (disease variant) Age of onset Symptoms Protein localization2 Protein function/activity3 Abnormal lipid/protein composition
CLN1 PPT1 (INCL) 5 months to 2 years (infantile) Decreased head growth, blindness, seizures, hyperexcitability, slowed motor and mental development Lysosomal matrix, extralysosomal vesicles, extracellular Palmitoylthioesterase Phospholipids (lipofuscin)/SAPs
CLN2 TPP1 (LINCL) 2–4 years (late infantile) Progressive dementia, motor dysfunction, seizures, myoclonus, ataxia, loss of vision Lysosomal matrix Serine protease Unknown lipids/SCMAS
CLN3 CLN3 (JNCL) 4–10 years (juvenile) Visual failure from retinal degeneration, progressive cognitive decline, motor dysfunction, seizures, and behavior problems Late endosomal / lysosomal membrane, presynaptic vesicles Modulation of vesicular trafficking/fusion, pH regulation and osmoregulation284, resistance to endoplasmic reticulum stress56; regulation of ganglioside levels285 BMP, phospholipids, galactosyl-ceramide/SCMAS
CLN4 DNAJC5, CSPα, (Parry NCL) ~30 years (adult) Slow progressing, death 10–20 years after onset Cytosolic, associated-vesicular membranes Hsc70 co-chaperone (exo/endocytosis) Unknown lipids/SAPs
CLN5 CLN5 (Finnish variant) 4.5–6 years (late infantile, juvenile) Slight motor clumsiness, seizures, ataxia, myoclonus Lysosomal matrix Modulation of vesicular trafficking/fusion Sphingolipids/SCMAS
CLN6 CLN6 2–4 years (late infantile); and adult onset Resembles LINCL in clinical characteristics ER-lysosomal membrane Likely involved with degradation of post-translationally modified proteins in lysosomes286 Phospho- and glycosphingo-lipids, cholesterol/SCMAS
CLN7 MFSD8 2–4 years (late infantile) Seizures, poor mobility, visual impairment Lysosomal membrane Transmembrane transporter function predicted, regulation of soluble lysosomal proteins287 Unknown lipids/SCMAS
CLN8 CLN8 (EPMR) 5–10 years (juvenile) Increasing seizures until puberty, then decreasing frequency through adulthood ER/ERGIC membrane Regulation in lipid metabolism predicted, regulates lysosome biogenesis72, Endoplasmic reticulum-export recycling receptor71 Ceramides, phospholipids, sphingolipids, sulfatides/SCMAS
CLN9 Unknown Unknown unknown unknown UNK, role in ceramide synthesis postulated Ceramides, sphingomyelin, sphingolipids, globosides/protein n.d.
CLN10 CTSD/Cathepsin D (congenital) Congenital, neonatal, and late infantile variants288 Epilepsy, extreme brain atrophy, with death in a few days from cardiac failure Lysosomal matrix, extracellular Aspartyl endopeptidase BMP, cholesterol, phospho- and sphingo-lipids/SAPs
CLN11 GRN/Progranulin 22–23 years (adult) Seizures, visual deterioration, myoclonus Extracellular UNK, roles in inflammation, embryogenesis, cell motility and tumorigenesis postulated, non-enzyme lipids/protein n.d.
CLN12 ATPase 13A2, 13–16 years (adult) Poor mobility, myoclonus, deteriorated speech Lysosomal membrane, multivesicular bodies UNK, regulation of ion homeostasis postulated, secretion of exosomes and α-synuclein86, non-enzyme lipids/protein n.d.
CLN13 CTSF/Cathepsin F (Adult Kufs type B) 20–32 years (adult) Dementia, personality changes Lysosomal matrix Cysteine protease lipids/protein n.d.
CLN14 K+ channel protein (KCTD7) 8–24 months (infantile) Seizures, visual and verbal deterioration, brain atrophy Cytostolic, associated to membrane Transmembrane protein voltage-gate potasium channel complex, potassium and glutamine transport92, autophagy93 lipids/protein n.d.
1

Abbreviations: NCL (neuronal ceroid lipofuscinoses), CLN (ceroid lipofuscinosis neuronal), INCL (infantile NCL), LINCL (late infantile NCL), JNCL (juvenile NCL), PPT1 (palmitoyl protein thioesterase1), TTP1 (tripeptidyl peptidase1), MFSD8 (major facilitator superfamily domain‐containing protein 8), EPMR (progressive epilepsy with mental retardation), KCTD7 (potassium channel tetramerization domain containing protein 7), SCMAS (subunit c of mitochondrial ATP synthase), SAPs (sphingolipid activator proteins), BMP (bis(monoacylglycerol)phosphate), and n.d. (not determined).

2

Soluble proteins (CLN1, CLN2, CLN5, CLN10, CLN13); transmembrane proteins (CLN3, CLN6, CLN7, CLN8, CLN12, CLN14).

3

Functions are unknown (UNK) for most of the CLNs; however, investigations have determined possible function/activity.

As a collective group, NCL patients are typically defined by observation of cognitive and visual impairments, epileptic seizures, and deterioration of motor skills and balance issues12,17. The hallmarks of these diseases are similar to other cognitive disorders and therefore, families encounter difficulties and severe delays in obtaining a confirmed diagnosis of NCL from local pediatricians and neurologists, unless there is an older sibling with the same diagnosis18. Children with NCL, who initially exhibit normal development, suffer a regression in their ability to talk or walk and develop cognitive or learning disability. Each NCL type has a different age of onset and rate of decline of the observational hallmarks of language and motor skill development, with many becoming lethal as the children age14,19,20. The severity of progressive neuronal degeneration in the CNS leads to the overt neurologic phenotypic changes typically observed in NCL-affected children6. NCL is the most common cause of dementia in children1,3,6,21. Other than Brineura® therapy for CLN2, there are no effective therapies for NCL22. However, the last 5 years have seen significant progress in development of therapeutics to halt and/or reverse the progression of these diseases. There are new therapeutics on the horizon and some that have already gained approval. We will review these advances in this article.

Presently, the NCL family of disorders includes 14 separate subtypes. The majority are inherited as autosomal recessive disorders with a combined incidence worldwide of 1:12,500 to 1:100,000 (Table I)14,23,24. The NCL database maintained by University College in London, UK (https://www.ucl.ac.uk/ncl/mutation.shtml) lists 515 known genetic mutations from 1436 NCL patients, including missense, nonsense, deletions, and splicing defects, assigned to genes encoding the 14 ceroid lipofuscinosis neuronal (CLN) lysosomal proteins4. From this database, the 4 most common NCL types are CLN1 (also known as “Infantile NCL”, with 220 patients), CLN2 (“Late Infantile NCL”, 369 patients), CLN3 (“Juvenile NCL”, 423 patients), and CLN6 (“variant LINCL”, 132 patients). Although the disorders all dramatically affect the CNS, some NCL children are also afflicted by blindness (typical in CLN2, CLN3, CLN11, CLN14), sleep disorders, and language delay or language regression12,25,26. The natural history of each NCL subtype is reviewed in section 2. Similar to other lysosomal storage disorders, NCL disorders affect the lysosomal processing of cellular waste products, and include defects involving secreted lysosomal proteins (CLN1, CLN2, CLN5, CLN10, and CLN13) and lysosomal transmembrane proteins (CLN3, CLN4, CLN6, CLN7, CLN8, CLN12, and CLN14)2729. Over the past decade, investigators have deciphered the functions of many of the CLN proteins; however, several remain unknown with regards to function/activity (Table I)2932.

Since our 2013 review of the NCL family of diseases in this journal33, there have been several reviews focusing on the biology of various forms of NCL and the potential of different therapeutic interventions35,14,26,29,3440. Hence for this review, we will focus primarily on pre-clinical and clinical developments for the treatment of NCL disease since 2013, and demonstrate the exponential growth in the interest of academicians and industry to put forward resources to halt or cure these devastating rare diseases. This review will focus on the natural history for the different NCL subtypes (section 2); experimental animal models for development of NCL therapeutics (section 3); biomarkers reported for NCLs (section 4); challenges for development of NCL therapies (section 5); the different therapeutic approaches to NCL (section 6); the latest pre-clinical and clinical research for therapeutic development for the various NCLs (section 7); how all the promising academic preclinical work has resulted in many partnerships with foundation/industry (Section 8); and will end with conclusions (section 9) and our opinion on what we see as the breakthroughs and challenges in the field of therapeutic development for NCL (section 10). The sections on natural history, animal models, biomarkers and therapeutic approaches are subdivided by NCL subtype with the corresponding current findings.

2. Natural History of the NCLs

The natural history or progression of the various NCL types is of vital importance for identifying ideal treatment windows and milestones for assessment of improvements or of rate of decline. There have been many natural history studies to track the progression of NCL as the patients age (Table II)25,26. These observational studies did not involve therapy or treatments, and many of the early studies enrolled subjects with NCL of all subtypes. More recent observational studies have been completed for single NCL subtypes: CLN2, CLN3, and CLN6 (Table II).

Table II.

Human Clinical Trials for Neuronal Ceroid Lipofuscinoses (Observational clinical studies)1

NCL/Study Title Treatment Phase Number Enrolled Status NCT Number2
All CLN types
 Clinical and Neuropsychological Investigations in Batten Disease Natural history only, no treatment none (observational) 400 Recruiting NCT01873924
 Neuronal Ceroid Lipofuscinosis and Associated Sleep Abnormalities Natural history only, no treatment none (observational) 57 Completed NCT01966757
 Genetic Characterization of Movement Disorders and Dementias Natural history only, no treatment none (observational) 12,000 Recruiting NCT02014246
 Inherited Retinal Degenerative Disease Registry Natural history only, no treatment none (observational) 20,000 Recruiting NCT02435940
 Longitudinal Study of Neurodegenerative Disorders Hematopoietic Stem Cell Transplantation none (observational) 1,500 Recruiting NCT03333200
CLN2
 Genotype - Phenotype Correlations of LINCL Natural history only, no treatment none (observational) 18 Completed NCT00151268
 Genotype-Phenotype Correlations of Late Infantile Neuronal Ceroid Lipofuscinosis Natural history only, no treatment none (observational) 75 Active, not recruiting NCT01035424
 Collection of Cerebrospinal Fluid in Healthy Children none (normal controls) none (observational) 32 Terminated NCT01698229
CLN3
 Investigations of Juvenile Neuronal Ceroid Lipofuscinosis Natural history only, no treatment none (observational) 50 Recruiting NCT03307304
CLN6
 Natural History of Neuronal Ceroid Lipofuscinosis, Batten’s CLN6 Disease Natural history only, no treatment none (observational) 15 Recruiting NCT03285425
1

Abbreviations: neuronal ceroid lipofuscinosis (NCL); late infantile neuronal ceroid lipofuscinosis (LINCL)

2

Clinical identifier numbers obtained from clinicaltrials.gov website

2.1. CLN1.

CLN1 disease, also known as infantile Batten disease or infantile neuronal ceroid lipofuscinosis (INCL), is caused by mutations in the CLN1 gene, which encodes the secreted lysosomal protein palmitoyl-protein thioesterase 1 (PPT1)41. Early development appears normal in affected patients, but by 18 months of age, they typically begin to show signs of developmental regression42. Rapidly developing psychomotor issues include decreased muscle tone, ataxia, myoclonus, epilepsy, and loss of vision, mobility, and vocal abilities42. There is progressive brain atrophy, and microcephaly can result. As the condition progresses, a feeding tube is often required. Children with the disease do not usually survive past early childhood25,26,42. There are some CLN1 mutations that cause later-onset forms of the disease, but all forms are associated with severely reduced life expectancy4345.

2.2. CLN2.

CLN2 disease, also known as late infantile neuronal ceroid lipofuscinosis (LINCL), is caused by mutations in the CLN2 gene, which encodes the lysosomal protein tripeptidyl peptidase 1 (TPP1)46. Affected children usually display symptoms of the disease by ages 2 to 4, typically starting with recurrent seizures and difficulty in coordinating movements47,48. The disease progresses with developmental regression, affecting speech and motor skills. Actions such as sitting and walking become more difficult, as does swallowing. Affected children also develop myoclonus, vision loss, and intellectual disabilities, and typically do not survive past 8 to 12 years47. Natural history observational studies have linked genotype and the severity of the phenotype, and the rate of neurologic decline has also been determined47,49,50. In an MRI study analyzing CLN2 children (n=38) compared to healthy controls (n=52), global thinning of the cortical regions was noted across the brain49. In newer observational studies for the same set of CLN2 subjects and healthy controls, MRI was used to measure the regional cortical thickness in the brain, which appears to be 2X thicker in controls vs CLN2 subjects, demonstrating the overall declining neurologic health of the CLN2 children50. In a recent study, which used the same methods of assessment for patient cohorts in the US and Europe, the natural history of children afflicted with CLN2 was defined, characterizing a predictable time frame of decline of language and motor skills51.

2.3. CLN3.

CLN3 disease, also known as juvenile neuronal ceroid lipofuscinosis (JNCL), is the juvenile-onset form of the disease. It is caused by mutations to the CLN3 gene that encodes a lysosomal transmembrane protein, the function of which is unknown52. It is theorized to be involved with the modulation of vesicular trafficking and fusion, or alternatively with regulation of lysosomal pH or osmoregulation16,5356. Disease onset usually occurs between 4 and 10 years of age, starting with visual failure from rapidly progressing retinal degeneration, followed by progressive cognitive decline and motor dysfunction5759. Behavioral problems and seizures then develop. Life expectancy is in the early 20s14,59. A small scale natural history trial is currently recruiting CLN3 patients (Table II), with no results reported to date.

2.4. CLN4.

CLN4 disease, also referred to as Parry disease, is an adult NCL, with signs and symptoms typically appearing around age 306063. Affected individuals develop general seizures and myoclonic jerks, along with decline in intellectual and physical function. Dementia, ataxia, tremors, and speech problems worsen over time, leading to death about 15 years after the symptoms first appear25. Contrary to the previously described forms of the disease, CLN4 patients do not have deterioration of vision, and it is the only NCL with an autosomal dominant inheritance pattern61,63. CLN4 disease is caused by a mutations in the DNAJC5 gene, which encodes the pre-synaptic co-chaperone cysteine string protein (CSP)61,63. CLN4 disease is closely linked to CLN1 disease, and it has been theorized that CSP and PPT1 have a substrate-enzyme relationship63.

2.5. CLN5.

CLN5 disease is also known as the Finnish variant of late infantile NCL64. It is caused by mutations in the CLN5 gene, which produces a soluble lysosomal glycoprotein of unknown function. The age of onset is typically between 4 and 7 years of age65. Affected children usually have normal growth and development until the disease manifests with motor impairment or psychomotor regression. As the disease progresses, patients exhibit clumsiness, seizures, loss of vision, ataxia, and myoclonus. Life expectancy ranges from 13 to 30 years of age, with some individuals having a slower progressing form of the disease65.

2.6. CLN6.

CLN6 disease, also known as variant late-infantile/early juvenile NCL, is caused by a mutation in a transmembrane protein66,67. The disease manifests between 18 months and 8 years of age, and is highly variable. Affected children experience loss of vision and motor skills, epilepsy, dysarthria, and ataxia68. Deterioration is rapid and death usually occurs before the teenage years. Mutations in the CLN6 gene have also been shown to be responsible for Kufs disease Type A, which was formerly classified as CLN4 disease62. Kufs Type A disease presents with symptoms very similar to those of CLN4 disease, but it has an autosomal recessive pattern of inheritance62. A small-scale natural history trial is currently recruiting CLN6 patients (Table II), with no results reported yet.

2.7. CLN7.

CLN7 disease is caused by a mutation in a lysosomal membrane protein of unknown function69. Onset is typically detected between ages 2 and 7, with symptoms including epilepsy, developmental regression, myoclonus, ataxia, loss of vision, and speech impairment70. Motor function and cognitive ability progressively worsen until death in the teenage years.

2.8. CLN8.

CLN8 disease is caused by mutations in a gene encoding a transmembrane endoplasmic reticulum protein, which regulates lysosome biogenesis12,71,72. The disease presents in 2 different ways: the Turkish variant late-infantile NCL or Northern epilepsy. The severity of the CLN8 gene mutation is responsible for the differences. The Turkish variant is the more-severe form of the disease, with onset typically between ages 2 and 773. Affected patients develop myoclonic epilepsy and ataxia, along with developmental regression, which causes loss of the ability to walk and speak. Cognitive ability also declines progressively, and affected individuals rarely survive past late childhood or early adolescence. Northern epilepsy is the less-severe form of the disease and is characterized by recurrent seizures74. Age of onset is typically between ages 5 and 10. As the disease progresses, affected individuals develop ataxia and other motor dysfunctions, and experience a slow decline in cognitive abilities. In the less severe Northern variant, affected individuals develop vision problems later in life, and life expectancy is reduced, although most survive into their 50s74.

2.9. CLN9.

CLN9 disease was identified recently75, and is not well characterized. The underlying genetic cause remains unknown, but it is theorized to be a regulator of dihydroceramide synthase76. Symptoms typically appear in early childhood (average age 4 years) and include declining vision, seizures, ataxia, and myoclonus75. Cerebral atrophy and developmental regression also occur, with death usually between ages 15 and 20 years75.

2.10. CLN10.

CLN10 is very rare, and it is likely that most cases go undiagnosed7781. The disease can present in neonates with microencephaly, absence of reflexes, epilepsy, and respiratory problems7781. Seizures can start before birth. CLN10 has the earliest onset for the NCLs, with expected survival measured in days, and cardiac failure being the most common cause of death7781. There are also late infantile and juvenile variants of the disease, with later onset of symptoms and a slower disease progression82,83. These patients may have normal early psychomotor development, but then begin to exhibit neurodegenerative symptoms including ataxia, and cognitive decline, with loss of vision, speech and mobility82,83. CLN10 disease is caused by mutations in the gene encoding for the lysosomal aspartic protease cathepsin D (CTSD). Different mutations cause varying levels of cathepsin D functionality, leading to the differences in disease progression81.

2.11. CLN11.

Similar to CLN4, CLN11 disease has a relatively late onset of early- to mid-20s. The disease is characterized by rapidly progressive visual failure due to retinal dystrophy, seizures, myoclonus, cerebellar ataxia, and cerebellar atrophy84. To date, it has only been reported in two members of an Italian family84. It is caused by a homozygous mutation in the granulin precursor (GRN) gene84.

2.12. CLN12.

CLN12 disease, also known as Juvenile parkinsonism-neuronal ceroid lipofuscinosis, has an onset between ages 13 to 1685. This Parkinson-like disorder is caused by mutations in the ATP13A2 gene, which encodes an enzyme that is thought to regulate ion homeostasis86. First characterized in a canine model87,88, CLN12 disease has also been identified in four members of a Belgian family85. For these 4 subjects, learning difficulties were evident around the age of 8 years, followed by unsteady gait, myoclonus, and mood disturbance in the pre-teen years. As the disease progresses, the affected individuals lose speech and develop mobility issues. Over time, they exhibit a combination of progressive spinocerebellar ataxia, bulbar syndrome, extrapyramidal and pyramidal involvement, and intellectual deterioration85.

2.13. CLN13.

CLN13 disease, also known as Kufs disease type B, is characterized by adult onset dementia, often associated with movement and behavioral abnormalities89,90. The disease is caused by mutations in the gene encoding cathepsin-F, a lysosomal cysteine protease89,90. Symptoms usually present around 30 years of age, starting with memory deficits and behavioral disturbances. As the disease progresses, motor function deteriorates and brain function decreases. Affected individuals develop tremors, bradykinesia, and extrapyramidal type rigidity. CLN13 is rare among the NCL family of diseases, and unlike most NCLs there is no loss of vision89,90.

2.14. CLN14.

CLN14 disease, also known as progressive myoclonic epilepsy (EPM3), is caused by mutations in the KCTD7 gene, which codes for the potassium channel tetramerization domain-containing protein 791,92. The disease is infantile-onset, usually manifesting between 8 to 24 months of age. Patients present with myoclonic seizures, followed by neurodegeneration, developmental regression, and visual failure as they age. Intracellular accumulation of autofluorescent lipopigment storage material is evident in fibroblasts, neurons, and eccrine secretory epithelial cells obtained through skin biopsy93,94

3. Experimental Animal Models Used for Testing Therapeutics for NCLs

Over the course of the last decade, many animal species and strains have been identified, with naturally occurring neurologic deficits relevant to the NCLs, while other experimental animals have been genetically engineered to knock out or knock-in NCL specific genes34,9598. Breeders and farmers have identified quite a few mammals with NCL-like deficits and investigators have mapped the genomic mutations responsible for NCL subtypes and variants in mice, dogs, pigs, and sheep (Table III)97,99,100. While most of these mutations are not the same as the major human NCL mutations, the hallmarks of the specific NCL subtypes are present, such as neuronal apoptosis, gliosis, and autofluorescent lipofuscin aggregates, allowing investigations of the function of the NCL proteins in situ34,98,101. Animal disease models are a useful tool for the development of novel new therapeutics for the human NCL. Since pre-clinical studies involve delivering candidate drugs/viral vectors to complex organs such as the brain or liver, where most NCL variants have pathology, an animal model is helpful to evaluate proof of concept. Large animal models (dogs, pigs, and sheep) provide a surrogate for the therapeutic target with similar size and complexity closer to the human brain34,35,39,98. These experimental animals also allow for multiple assessments of safety along with comparative studies for drug distribution as a function of administration routes to the CNS. The following summarizes the current animal models available for each NCL with particular emphasis of those added to the literature since 2013 (Table III). While there are non-mammalian models (zebra fish, C. elegans, and yeast) being created for NCL drug development, they are not included here as they have been previously reviewed by Faller et al34.

Table III.

Neuronal Ceroid Lipofuscinoses Animal Models

Neuronal ceroid lipofuscinoses type1 Gene/protein (variant) Mouse models Large animal models with neuronal ceroid lipofuscinoses defects2
CLN1 PPT1 (INCL) Cln1−/−, Ppt1Δex4, Ppt1Δex9, Cln1R151X103 Miniature Dachshund
CLN2 TPP1 (LINCL) Tpp1−/−, Arg446His, Cln2R207X/R207X110, TgLSL-TPP1111 Long-haired Dachshund2
CLN3 CLN3 (JNCL) Cln3−/−, Cln3Δex1−6, Cln3Δex7−8 ExeGen mini pig
CLN4 DNAJC5, CSPα, (ANCL) None None
CLN5 CLN5 Cln5Δex3 Borderdale sheep119,120, Devon cattle, Border collie
CLN6 CLN6 nclf (c.307insC) South Hampshire sheep2, Merino sheep127, Australian shepherd
CLN7 MFSD8 Mfsd8tm1a/tm1128a Chihuahua131, Japanese Macaque132
CLN8 CLN8 Cln8/mnd (c.267–268insC) English setter2, Alpine Dachsbracke134, Saluki135
CLN9 unknown None None
CLN10 Cathepsin D (CTSD) CTSD −/−136 Swedish Landrace sheep, American bulldog
CLN11 Progranulin Grn−/−139, PGRN-deficient140
CLN12 ATPase 13A2, None Tibetan terrier
CLN13 Cathepsin F (CTSF) Ctsf−/− None
CLN14 K+ channel protein (KCTD7) None None
Unknown NCL genetic defect unknown Rambouillet sheep, Beefmaster & Holstein cattle, domestic ferret, short-haired Siamese & Japanese cats, Aegidienberger horse, Nubian goat, Vietnamese pot-bellied pig, Lovebird parrot, Cynomologous monkey, Mallet duck, and dogs such as the American Staffordshire terrier, Australian terrier, Polish lowland sheepdog, Miniature schnauzer, Cocker spaniel, Dalmatian, Japanese retriever, Welsh corgi, Labrador retriever, Australian cattle dog
1

Soluble proteins (CLN1, CLN2, CLN5, CLN10, CLN13); transmembrane proteins (CLN3, CLN6, CLN7, CLN8, CLN12, CLN14).

2

Active experimental populations available for study.

3.1. CLN1.

There are two different mouse models commonly used for CLN1 research. These have been genetically created by homozygous deletion of either the entire exon 4 (Ppt1Δex4) or the exon 9 (Ppt1Δex9) of the PPT1 genome102. These deletion mutations are not genetically analogous to human CLN1 mutations. Miller et al created the R151x mouse model, with a knock-in for the C>T genomic point mutation in exon 5 of the Cln1 gene, which mimics the human p.R151x mutation, the most common nonsense mutation in Cln1 (52.3%) based on genetic surveys31,103,104. The Cln1R151X mouse model was used to demonstrate that read-through drugs gentamicin and Ataluren (Translarna) increased PPT1 enzyme activity, a measure of therapeutic efficacy103. In addition to the engineered mouse models, a Dachshund and a Cane Corso dog have each been found to have CLN1 phenotype and genotype, with efforts underway to establish colonies105,106.

3.2. CLN2.

Pre-clinical studies with TPP1 deficient mice (Tpp1−/−), a model of CLN2 disease, were used to evaluate the potential for enzyme replacement and gene therapies107109. Two newly developed mouse models have been characterized. (1) CLN2R208X which carries a nonsense mutation that, unlike the original Cln2−/− mouse model, allows for mutation-guided therapies such as antisense oligonucleotides (ASO) and nonsense suppression which would be relevant to the human p.R208X mutation110. (2) A mouse model based on an inducible TPP1 transgene is still in development, which uses a stop cassette inserted ahead of the TgLSL-TPP1 transgene, thus disrupting TPP1 expression. The stop cassette is removable by tamoxifen-induced cre-mediated recombination; however, none of the four tested transgenes worked in the mice as hoped111. In addition, similar to CLN1, a Dachshund dog model has also been found with CLN2 phenotype and genotype101.

3.3. CLN3.

Multiple mouse models of CLN3 have been developed, each with large gene deletions (Cln3−/−, Cln3Δex1−6, Cln3Δex7−8) to mimic the human mutation. These mice display similar progression of CLN3 pathology including early glial activation followed by neuronal loss112. These mouse models have been used to develop anti-inflammatory and candidate gene therapies113118. Some phenotypes observed in CLN3 patients are not recapitulated in these mouse models6. A novel porcine model of CLN3 disease (CLN3Δex7−8/Δex7−8) is currently under development by Exemplar Genetics (ExeGen minipig)6, but no detailed publications are available. These CLN3 minipigs had decline in behavioral, pathological, and visual deficits similar to CLN3 children6. In contrast to mice, pigs are good animal models as they share with humans similar brain development and structure, anatomy, and physiology6. In addition, these pigs have a macula–like structure, area centralis, that has been shown to be valuable for studying macular-associated retinal diseases, a critical phenotype of the human disease which is missing in CLN3 mice6.

3.4. CLN4.

There are no animal models created to model CLN4 disease, and no naturally occurring CLN4 animals have been reported34,39,101.

3.5. CLN5.

In addition to the exon 3 deleted mouse model of CLN5 (Cln5Δex3), there are bovine, canine, and ovine large animal models with naturally occurring mutations homologous to that of human CLN5106,119124.

3.6. CLN6.

Unlike the other CLN mouse models created via homologous recombination, there is a naturally occurring mouse model of CLN6 disease (Cln6nclf), with a 1 bp insertion frameshift mutation similar to that detected in CLN6 patients of Pakistani origin68. The Cln6nclf mice present hallmarks of the CLN6 phenotype, including early loss of vision followed by severe CNS and behavioral deficits68. Recent developments for CLN6 gene therapy of the eye using CLN6 mice revealed that the retinal disease in Cln6nclf mice is predominantly characterized by photoreceptor degeneration125. There are also two breeds of sheep (South Hampshire and Merino) and one dog model of CLN6126,127.

3.7. CLN7.

A mouse model of CLN7 disease was created by inserting a lacZ gene-trap cassette and disrupting the murine CLN7 gene128, MFSD8. These Cln7-deficient mice (Mfsd8tm1a/tm1a) resemble the NCL-phenotype of human CLN7, including the accumulation of autofluorescent material in the brain and peripheral tissues, rapid progressive deterioration of photoreceptor cells in the retina, and generalized neuroinflammation128. Mutant CLN7 mice lose more than 70% of their rod photoreceptors by 4 months of age, and have accumulation of SCMAS and saposin D in the retinas129. The lifespan of these mice is shortened, and lysosomal dysfunction was shown by the storage of autofluorescent lipofuscin-like lipopigments: subunit c of mitochondrial ATP synthase, and saposin D in the CNS and peripheral organs; and increased expression of lysosomal cathepsins B, D, and Z in the brains of Cln7 knockout mice130. Neurodegeneration occurs in the olfactory bulb, cerebral cortex, and cerebellum130. A family of Chihuahua dogs has also been diagnosed with a naturally-occurring mutation in the MFSD8 gene, resulting in CLN7 disease131. The symptoms also match human cases131. There is also a suspected case of the same mutation in a Chinese crested dog, but there were no family ancestries available to confirm the genetic trait131. A naturally occurring primate model has been identified in Japanese macaques132 with a homozygous frame shift mutation in the MFSD8 gene. The affected monkeys display progressive neurological deficits and visual impairment over a disease course of 5 to 6 years. While the mutation is not homologous to any previously identified CLN7 mutations in humans, it is believed to result in the formation of a truncated, non-functional lysosomal protein132. The disease manifestations closely mirror those of human CLN7, as well as those of the MFSD8 dogs132.

3.8. CLN8.

In addition to a known English Setter model133, novel mutations in the CLN8 gene have been discovered in two other breeds of dogs. One mutation, observed in Alpenländische Dachsbracke dogs, consists of the entire deletion of the CLN8 gene134. Another novel mutation in the CLN8 gene was observed in the Salukis breed, consisting of a single base pair insertion in exon 2, producing a stop codon135. In affected dogs of both breeds, the clinical signs and histological findings mirrored those of human CLN8 patients.

3.9. CLN9.

No animal models have been reported for CLN934,39,101.

3.10. CLN10.

Mouse models are available to study the cathepsin D (CTSD) defect found in CLN10 patients, with knockouts of the gene by neomycin inactivation of exon 4 have been created136. Naturally occurring defects have also been found in sheep137 and in American bulldogs138.

3.11. CLN11.

For CLN11 mouse models, two strains of progranulin (PGRN)-deficient mice have been created. One strain (Grn−/−) consists of 129/SvJ mice that have the Pgrn gene knocked out through targeted disruption139, while the other is a conditional knockout created by crossing pgrn floxed mice with CAG-Cre transgenic mice140. The first strain was created to investigate male sexual behavior, aggression, and anxiety in mice, while the second strain was created to investigate frontotemporal dementia (FTD). Little is known about the function of progranulin in the CNS, but the available data supports its involvement in embryonic development, cell growth modulation, wound healing, inflammation, and tumorigenesis140. The Grn−/− mice were observed to have elevated aggression and anxiety levels compared to wild-type mice, as well as lower levels of mRNA expression of the serotonergic receptor 5-HT1A in the hippocampus139. The PGRN-deficient mice were observed to have an exaggerated inflammatory response to bacterial infection, and the brains showed greater activation of microglia and astrocytes140. When subjected to behavioral testing, PGRN-deficient mice demonstrated increased depression- and disinhibition-like behavior, as well as social recognition deficits141. Progressive development of neuropathology started at 12 months of age, and older mice demonstrated impaired spatial learning and memory in the Morris water maze141.

3.12. CLN12.

There is an mouse model for CLN12 (Atp13a2-deficient) available which was created by gene disruption, and a CLN12-like phenotype has been found in Tibetan terriers due to a deletion mutation in ATP13A2 gene87,101,142.

3.13. CLN13.

No new animal models since our 2013 review34,39,101.

3.14. CLN14.

No new animal models since our 2013 review34,39,101.

4. Biomarkers

Although there are many common clinical pathologies among the NCLs, the subset corresponding to any one NCL as well as the age of onset varies significantly between the NCLs and this may or may not correlate with genotype. Therefore, the determination of endpoints for the evaluation of candidate therapies is complex and often relies on complicated clinical scales. The development of quantitative biomarkers for the NCLs is thus an important component in the validation of potential treatments. Biomarkers that are specific biochemical and/or structural changes that represent a unique “fingerprint” of a particular NCL would be ideal as surrogates for assessing disease progression, particularly if the assessed clinical phenotypes, which are often non-quantitative and subjective, are highly variable among the affected population. The use of clinical grading scores of a patient’s performance on a language/motor based rating scale or on quality-of-life questionnaires is subjective by the nature of the observational assessments. Alternatively, the use of clinical imaging by MRI or CT to monitor the progression of the disease may be quantitative, providing a measure of structural changes that correspond to disease progression. Discovery of a validated biomarker in the blood or CSF would enable investigators to track the improvement or decline of their patients with a biochemical assay without subjective appraisals of the subject’s condition.

Recent investigations of NCL subjects and animal models have resulted in several potential clinical biomarkers. In the Hersrud et al143 study, plasma biomarkers were assessed in subjects exhibiting CLN3 (n=13) and other NCLs, 3 with CLN1, 1 with CLN2 and 2 with Finnish variant, CLN5. These investigations identified 7 potential biomarkers associated with neurodegenerative and inflammatory diseases that were significantly elevated in CLN3 plasma samples compared to normal controls, including: brain-derived neurotrophic factor (BDNF), cell adhesion molecules (NrCAM and VCAM1), clusterin, adiponectin, Apolipoprotein E (ApoE), and myoglobin. A subset of these biomarkers (BDNF, ApoE, and VCAM1) were significantly elevated in all NCL serum samples. These findings suggest that these potential biomarkers denote cellular decay in the CNS during late-stages of NCL, especially the neurotrophin BDNF, which is a compensatory response to brain injury and neuroinflammation143.

The study by Sleat et al., was the first biomarker study comparing NCL patients (CLN1, CLN2, CLN3) and healthy controls with mass spectrometry and proteomics144. Whereas the earlier Hersrud study tested plasma from NCL patients with immunoassays143, the Sleat study144 assessed biomarkers from NCL patient brain tissue and CSF derived from autopsy. Unfortunately neither study assessed more than a single time point in the same patient, leaving missing the critical time-dependent change in biomarkers. For example a promising candidate biomarker is myoglobin, where the levels of myoglobin were elevated in the CSF in CLN1, CLN2, and CLN3, and in plasma for CLN3143. As might be expected in the brain samples of those affected with an NCL, abnormal storage material components were significantly elevated, such as: (1) ATP5G1 (one of three human paralogs that encode membrane subunit c of the mitochondrial ATP synthase) in CLN2; and (2) PSAP (prosaposin, precursor of lysosomal saposin proteins, A, B, C and D) in CLN1 (Table I). Glial cell activation of GFAP (glial fibrillary acidic protein) in CLN1/CLN2 and STXBP1 and STX1B (syntaxin-binding proteins, both associated with recurrent seizures (epilepsy), abnormal brain function (encephalopathy), and neurodevelopmental disorders) in CLN1 were all decreased144,145. This study identified 18 potential CSF biomarkers in common for 3 NCLs that were significantly altered compared to the normal control. Specifically, proteins elevated in all NCL CSF included: cellular retinoic acid binding protein 1 (CRABP1), aldehyde dehydrogenase 1-A2 (ALDH1A2), and collagen type XIV α1 chain (COL14A1). Proteins consistently down-regulated proteins in all NCLs tested were myelin-associated glycoprotein (MAG) and carnosine dipeptidase 1144. Proteins that appear to be NCL-specific included REG3A (regenerating family member 3 alpha; elevated in CLN1 CSF), STXBP1 (reduced in CLN1 CSF and brain), and calbindin1 (elevated in CLN2 CSF)144.

A study published last year using mass spectrometry for untargeted metabolite profiling focused on identifying potential biomarkers from CSF samples from CLN2 patients (n=11) vs healthy controls (n=10)146. Among the CSF metabolites evaluated, potential biomarkers for tracking the progression of CLN2 disease included a dozen acetylated amino acids, such as N-acetylaspartylglutamic acid (NAAG), the most abundant excitatory peptide neurotransmitter in the CNS, glycerol-3-phosphoinositol, N-acetylneuraminic acid, N-acetylneuraminic acid dimer, N-acetylalanine, N-acetylserine and N-acetylthreonine, as well as glycero-3-phosphoinositol and sulfoacetic acid. All of the metabolites were significantly reduced in CSF samples from CLN2 affected subjects146. It is not known if this panel of metabolites is observed in other NCLs, or may offer a unique biomarker signature for CLN2 disease prognosis and the monitoring of efficacy for treatment modalities146.

There have been several studies evaluating serum biomarkers for CLN3 disease in affected individuals and mouse models. A clinical study of CLN3 analyzed 25 subjects (11 male and 14 females) by genome-wide expression profiling (GWAS), and found dysregulation of the dual specificity phosphatase 2 (DUSP2) gene in all CLN3 individuals, while 13 other genes including guanine nucleotide exchange factor 1 for small GTPases of the Ras family (RAPGEF1) and transcription factor Spi-B (SPIB) were found to be possible disease-modifier genes147. Validity of these potential biomarkers would be likely if these genes were found to play a critical role in CLN3 biosynthesis. There have been two studies in CLN3-deficient mice. An analysis of homozygous CLN3-deficient mice (Cln3Δex7/8) for hematological biomarkers found changes as early as 15 weeks of age in serum ferritin concentrations, mean corpuscular volume (MCV) of red blood cells, and reticulocyte counts compared to wild-type mice. Also, T-cell deficiencies were found in the male Cln3Δex7/8 mice100. In contrast, another recent hematological biomarker study in CLN3 knockout mice (Cln3Δex7/8) found no consistent changes148.

5. Challenges for Development of Therapies for Treatment of NCLs

Since the manifestations of the NCL family of diseases affect the CNS directly, a successful therapy must overcome a host of issues to become an approved therapeutic. The blood-brain barrier (BBB) is a major barrier to the systemic administration of most CNS therapeutics39,149. Only selective compounds like lipids are transported across the tight junctions forming the barrier149. Some research has focused on direct administration of therapeutics to the CNS to bypass this anatomical barrier. The challenges for effective CNS therapy include: (1) the short half-life of proteins used in enzyme replacement modalities, necessitating a repeated infusion of the recombinant proteins to be efficacious; (2) the need for widespread distribution of the therapeutic across the CNS; (3) avoiding immune response against the therapeutics; and (4) identifying the disease and treating early due to the rapid decline of NCL patients. In addition to these challenges, delivery of recombinant functional proteins or viral vectors to the CNS also have to overcome the dense cellular environment of specialized cell types in order to target the appropriate cell type. This is important, since different NCL subtypes affect different regions of the brain, ranging from widespread neurons across the cortex regions to focal cerebellar deficits3,39. When successful therapy requires the spread of therapeutics to cover the breadth of the CNS, the challenge is to assure that local administration of the therapeutic is sufficient. For example, while a reservoir of constitutive production of secreted proteins may cure CLN1 and CLN2, transmembrane NCL proteins like CLN3 that are not secreted and hence do not undergo receptor-mediated uptake by neighboring cells. For the NCLs with non-secreted gene products, larger numbers of transduced cells will be obligatory for a cure. A significant issue is the immune response against the therapeutic150. Antibody response evoked against the therapeutic protein or, in the case of gene therapy, the delivery vehicle, can lead to clearance of infused proteins and/or gene therapy vector. Finally, due to the rapid decline in health of NCL patients from the disease manifestations, it may not be possible to reverse or halt disease progression once the NCL is identified. This is especially critical in early onset NCL for infants. Successful modalities may require a combination of the therapies. Therapies like enzyme replacement therapy may provide coverage in the short term, while long-term therapies like gene therapy take time to develop in multiple regions of the CNS and body.

6. Therapeutic Approaches to the NCLs

There have been major advances in the treatment of NCLs, including identification of the genetic mutations and resulting protein defects, creation of animal models, enzyme replacement therapies, and the initiation of clinical trials for novel treatment modalities. Unlike other lysosomal storage disorders, the proteins involved in some of the NCLs are not secreted, making therapeutic attempts more difficult. Despite numerous challenges ranging from therapeutic strategy, to early identification of afflicted subjects and measurable endpoints, new research has been carried out in 2 categories: (1) Disease-specific therapies that are unique to a given subtype; and (2) therapies that target downstream pathology, which may be applicable to more than a single NCL2,6,33,37,151,152. The first category includes: enzyme replacement therapy, stem cell ex vivo therapy and gene therapy, and the second category includes pharmaceutical treatments involving small molecule drugs. The following describes each of these treatment options and the associated pros and cons. The current treatment modalities in development for each NCL type are discussed in section 7.

6.1. Enzyme replacement therapy (ERT)

Using ERT as a strategy to augment the decreased levels and/or non-functioning protein with a functional protein, treatment of many lysosomal storage disorders have been evaluated by systemic protein infusion7,153155. While this works for lysosomal storage disorders (LSD) that are non-neuropathic, they have limited success due to the blood-brain barrier (BBB) in disorders like the NCLs that have CNS involvement156. To circumvent the BBB, it is necessary to use extremely high doses of systemically-delivered enzyme during the neonatal period in order to get some penetrance into the CNS. An alternative is to use fusion proteins in which enzyme molecules are attached to a peptide moiety that is able to assist in delivery across the BBB by receptor-mediated endocytosis. Finally, another approach could be by direct infusion of the recombinant protein into the CSF via an intrathecal, intracerebroventricular, or intracisternal route149,154,156. Studies have shown that ERT products have varying degrees of success in treating lysosomal storage disorders, and are dependent on initiating therapy at an early stage of disease development as well as achieving widespread distribution. Early intervention is key to halting or slowing disease progression prior to irreversible pathology. Since several of the NCL defects arise in proteins that are secreted (CLN1, CLN2, CLN5, CLN10, CLN13), ERT could potentially provide functional versions of the relevant protein to the site of need in the CNS. The advantage of treatment of a disease in which the gene codes for a secreted protein is that the ERT protein uses the natural mechanism by which receptor-mediated uptake provides an efficient pathway for trafficking to the lysosome157,158. Importantly, only low amounts of the enzyme (~5–10% of the endogenous levels) are typically needed for therapeutic effects7,154,155. However, there are several disadvantages of ERT to treat the NCL7,154,155. First, the short half-lives of the NCL enzymes (days to weeks) require repeat administration of the therapeutic protein to the CNS. This requires frequent, specialized treatment. There may be the development of humoral immunity against repeat administered proteins that could limit efficacy, induce inflammatory responses, and reduce cost effectiveness. There are also the potential complications resulting from the use of delivery devices such as catheters or long-term placement of implanted reservoirs154. Finally, the manufacturing of the protein product requires extensive methods development and processes that include qualified host cell lines and validated purification schemes. Despite these challenges, one of the first successes in NCL therapeutic development is Brineura®, an FDA-approved intracerebroventricular administration of ERT for CLN2159.

6.2. Stem Cell Therapy

As an alternative to the requirement for a lifelong repeated dosing for ERT, ex vivo cell therapy could provide an in vivo source of the therapeutic protein via transplanting heterologous bone marrow or isolated hematopoietic stem cells with the normal gene coding for the functional protein, or alternatively, autologous marrow or stem cells that have been genetically modified with the normal gene.

6.2.1. Bone marrow transplant - unmodified.

One source of a therapeutic stem cell population for NCL therapy is the delivery of hematopoietic marrow cells from a donor to the affected patient. While this has been only modestly effective in clinical studies for several CNS disorders, such as cerebral adrenoleukodystrophy160, it may work for some of the NCLs. There are many disadvantages to using marrow transplants including the requirement of an HLA-matched donor to prevent graft rejection and the need for a lifelong immunosuppression regimen161. In addition, providing unprocessed marrow cells may fail to provide sufficient amounts of wild-type enzyme to the whole body; and critical for NCLs, the likelihood of impact on the CNS manifestations is low due to the BBB.

6.2.2. Stem cell transplant – local or systemic delivery.

To improve the potency of transplanted cells, most research has targeted the use of a subpopulation of the blood, marrow, or umbilical cord blood – hematopoietic stem cells (HSC)161. Isolation of HSC from blood or bone marrow or umbilical cord blood can be easily accomplished with antibody sorting columns or with fluorescent antibody cell sorting (FACS)162,163. Isolated processed HSC can be obtained from the donor’s blood or marrow following cytokine immobilization to boost the HSC populations. Infusion of the stem cells can be either systemic or localized to the CNS. Use of systemic delivery of cells may correct the disease in peripheral organs, but the BBB presents a challenge to effective CNS therapy36. Clinical trials for metachromatic leukodystrophy (MLD), unrelated to the NCLs, have shown that some of the transplanted stem cells do manage to migrate into the CNS, becoming glial cells164169. Other clinical studies have looked at direct localized delivery of cells to CNS to correct neurodegeneration170,171. There are several stem cell types (hematopoietic, mesenchymal, neurogenic) available for transplant trials168,171. Several clinical trials with HSC have been completed for CNS disorders unrelated to NCLs (globoid cell leukodystrophy, adrenoleukodystrophy, MLD, and Hurler syndrome) and show safety and limited efficacy165,172, suggesting that this approach could offer a solution. But, there are distinct disadvantages to using HSC for NCL therapy. The requirement of a matched donor to avoid graft rejection, and the continued immunosuppression remain as significant challenges. While systemic cell therapy involves intravenous infusions, direct delivery to the CNS involves an invasive procedure with safety risks. In addition, use of HSC therapy requires log expansion of stem cells in culture prior to stem cell transplant, requiring a wait period before the administration procedure. Based on previous human clinical data, the human stem cell transplant may correct only a small subset of the CNS, and thus fail to provide sufficient amounts of functional enzyme to the target CNS cells173. A critical hurdle here is the extent to which transplanted cells can safely and effectively repopulate the CNS.

6.3. Gene Therapy

In contrast to transplanting unmodified cells, gene therapy involves use of viral vectors (e.g., AAV, lentivirus) to deliver genes that encode functional proteins in vivo. If safe and efficacious, gene therapy has the potential for lifelong cure150. Gene therapy applications have been evaluated by direct administration of the viral vectors to the patient or by augmenting pre-treated isolated cells ex vivo for autologous transplant to patients. The HSC isolated from patients can be grown in culture and reinfused back into the patients (ex vivo) following viral transduction and sorting for the optimal producing cells. The advantages and disadvantages of these strategies are described below.

The NCL family of diseases have features that facilitate a gene therapy strategy. First, each of the diseases is caused by mutations in a single gene, they are primarily autosomal recessive disorders, and the missing/deficient protein has been well characterized for most of the 14 NCLs13,15,16. For instance, with CLN2, comparisons of genotype and phenotype suggest that only 5 to 10% of normal levels of the functional protein are associated with a less severe form of the disease with delayed age of onset and mortality. Thus, the severe phenotype should be ameliorated if the functional TPP1 protein is at levels that are 5 to 10% that of normal. Second, several of the NCL proteins are secreted and are capable of cross-correcting neighboring cells. This precludes the need to transfer the therapeutic gene to all of the cells in the CNS. Cross-correction occurs by the uptake of the TPP1 protein precursor (the “pro” form secreted by the gene therapy-corrected cells) is mediated by mannose 6-phosphate (M6P) receptors174176. The M6P moiety, added to the TPP1 protein precursor during post-translational processing, binds to cell membrane receptors followed by internalization and sorting of the TPP1 precursor to the lysosomal compartment. The implication of this biology for gene therapy is that the cells transduced by the viral vector become factories for the corrective enzyme, decreasing the challenge of extensive cell transduction and enhancing the potential for successful therapy177.

6.3.1. Ex vivo gene therapy, systemic.

The use of gene therapy vectors to modify stem cells has an extensive history and has been optimized over time to ensure a high rate of recovery of transduced cells162,178. Transplanting these modified cells to a patient with NCL has several potential advantages. Systemic delivery via an intravenous infusion catheter would spare the need for invasive surgical administration into the CNS and would simultaneously treat non-CNS disease manifestations162. Transduced stem cell progenitors are expected to replicate for the long term, thereby requiring a single dose for life-long therapy. If autologous patient cells are used, then immunosuppression regimens may not be required179. The disadvantages of ex vivo therapies for NCL include the challenge of assuring the appropriate trafficking of progenitor cells to the brain (if inadequate, direct administration to the CNS may be necessary) and the need to select for efficiently transduced HSC in vitro prior to re-administration162,167.

6.3.2. Ex vivo gene therapy, CNS.

As with systemically-delivered genetically-modified cells, local delivery to the CNS of an NCL patient would have several advantages: (1) a single dose of treated cells may be sufficient to stem or ablate the disease progression and no immunosuppression may be required if autologous cells are used; (2) there are neurogenic stem cells which have been approved for clinical transplant trials171; and (3) the expressed therapeutic protein would likely be translationally modified in an identical fashion to the normal protein. The disadvantages are similar to the systemic infusion route: manipulations of the HSC requiring complex ex vivo culture processing, and a potential lack of a selective advantage mechanism for efficiently transduced stem cells. The likely requirement for intraparenchymal or intrathecal administration of the genetically-modified stem cells is potentially risky with a need to carefully monitor safety for possible rogue differentiation or tumor formation. Further, an inflammatory response and/or cell-mediated immunity directed toward the administered cells may result in loss of therapy.

6.3.3. In vivo gene therapy, AAV vectors.

The most commonly used gene therapy transfer vector for therapeutic development for NCLs is the adeno-associated virus (AAV)152,180. It has the advantage of high efficiency in vivo cell transduction and persistent transgene expression. There are many AAV serotypes with tropism for cells in the CNS and the retina, thus providing numerous options with some selectivity for specific cell type180183. There is extensive safety data for the use of AAV for CNS gene transfer as a therapeutic modality to address Parkinson’s disease, Alzheimer’s disease, Canavan’s disease, Pompe, MPS IIIA, MLD, and LINCL (CLN2) in human clinical trials89,184196. There remain several challenges for AAV gene therapy to the CNS. Although systemic AAV therapy has been shown to be efficacious in mouse models of NCL, the blood-brain barrier appears to be a greater hurdle in humans and therefore direct CNS administration may be required180. Pre-existing anti-AAV seropositivity in humans may lead to complications of inflammatory response, and/or cellular immunity-mediated loss of transgene-expressing cells197,198. Careful choice of AAV serotype can reduce this concern, but it is not practical for selecting specific serotypes to address the needs of individual subjects197,199.

6.3.4. In vivo gene therapy, lentivirus vectors.

While the majority of the gene therapy investigations have tested AAV vectors, one group has been testing lentiviruses to deliver the genes for CLN5 and CLN6 to cell cultures and large animal models (NCL-afflicted sheep)200,201. While lentivirus vectors can support larger transgenes and can insert the payload into the host cell genome to ensure passage of the therapeutic genes to daughter cells, neither of these advantages are particularly relevant to NCLs, where genes are not large and cells are mostly non-(or slowly-) dividing cells. However, most of the lentiviral vectors have been used with ex vivo stem cells and there is limited safety and clinical data on the use of lentiviruses for direct CNS gene therapy.

6.4. Pharmacological Treatments

Various small molecule treatments have been evaluated for treating neurological disorders such as the NCLs. These small molecules include lysosomotropic agents, glutamate receptor antagonists, anti-apoptotic drugs, and immunosuppressants21,202. A number of these agents have been evaluated in clinical trials for NCLs, as described below and separately for each NCL in section 7. The advantages of small molecule therapies include the ability to manufacture large and easy-to-characterize batches of the drugs for clinical testing of safety and efficacy. Disadvantages include the potential of the drugs to have numerous off-target effects, toxicology related to unanticipated reactions, and unpredictable outcomes of metabolites. Importantly, these drugs represent an orthogonal approach and can be used either with existing therapies or may be combined with other genetic-based therapies. It is important to note though, that the disadvantages of combining treatments would be that it makes it difficult to discern individual degrees of efficacy. The small populations of those afflicted with an NCL would preclude carefully controlled comparative studies with individual and combined drugs. Finally, animal studies for other diseases have shown that doses in small animal models may not be scalable for human usage.

6.4.1. Lysosomotropic agents.

The first class of drugs being tested as NCL therapeutics are termed lysosomotropic drugs (includes cysteamine, phosphocysteamine, cysteamine bitartrate (Cystagon) and N-acetylcysteine). These compounds are transported to lysosomes where they function by inhibiting formation of cysteine thioesters and blocking ceroid accumulation, thereby reducing storage material and improving lifespan of treated animals203. Investigational drugs that mimic the thioesterase activity of PPT1 may have therapeutic potential for CLN1 by providing an alternative enzymatic means to hydrolyze the thioester bonds in the lysosomal ceroids203,204.

6.4.2. AMPA receptor antagonists.

These novel candidate therapeutics are antagonists designed to bind the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA receptor; AMPAR), a CNS transmembrane receptor for glutamate and the glutamate analog, AMPA36. AMPA receptors are continuously being trafficked into and out of the neuronal plasma membranes205. The noncompetitive AMPA receptor antagonists (talampanel and perampanel) have been demonstrated to have activity in the treatment of adults with partial seizures, suggesting a potential target for the treatment of NCLs with epilepsy205,206. These drugs treat the symptoms rather than the underlying disease. Several mouse studies have indicated potential usage for AMPA receptor attenuator in NCLs36,207209.

6.4.3. Anti-apoptotic drugs.

Anti-apoptotic drugs, such as flupirtine (ethyl-2 amino-6 (fluorobenzyl)-amino-3-pyridine carbate), which is a non-steroidal analgesic, are being tested to arrest neurodegenerative progression in CNS neurons prominent in some NCLs210,211. Another example is anti-caspase-4 inhibitor (such as Z-LEVD-FMK), which has also been shown to inhibit the apoptotic effects212.

6.4.4. Other interventions.

There are multiple small molecule drugs being tested for NCLs that have shown promising results in other neurologic disorders. Immunosuppressive drugs such as CellCept® (mycophenolic mofetil) and lipid regulating agents such as LOPID® (gemfibrozil) are currently being tested in clinical trials. Gemfibrozil, which has been shown to reduce inflammation, glial activation, lipofuscin in the brain, and apoptosis, is being studied for delay of motor deficits in mouse models116,213. Finally, immunomodulatory drugs (such as fingolimod and teriflunomide) have been shown to reverse neurodegeneration in a mouse model116. Some of these drugs have been combined with diet modifications to improve the current therapies, which will be discussed in the individual subtype sections.

7. Pre-clinical and Clinical Trials for the NCLs

Even though the most prevalent and most studied NCLs are CLN1, CLN2, and CLN3, many of the other NCL subtypes have also been the target of development of novel therapeutics in the last few years. The most promising and latest efforts are discussed below. We have separate sections for natural history studies (Table II) and interventional NCL clinical trials (Table IV). Treatment of NCL subtypes that are affected via soluble secreted lysosomal proteins (such as CLN1, CLN2, and CLN10) have had the most success with ERT and gene therapy either in animal models or human clinical trials. Receptor and membrane-bound CLNs (CLN3 and CLN6) have also had some success in treating the disease phnotypes in mice and large animals. As a general principal, candidate therapies require proof-of-concept animal studies with some measure of efficacy typically in one or more of the following parameters: CNS pathology, behavior, survival, and distribution of morphological or histological measures as a function of time. The candidate therapy also requires demonstration of general safety prior to commencing with a human study. The following sub-sections detail the progress made for the various therapeutic modalities for each NCL subtype since our last review on this topic33. There have been no publications for therapeutics that address CLN4, CLN9, CLN12, CLN13, and CLN14 and thus these are not included in the subheading reviews below.

Table IV.

Human Clinical Trials for Neuronal Ceroid Lipofuscinoses (Clinical studies, Phases 1–4)1

NCL/Study Title Treatment Phase Number enrolled Status Number2
All CLN types
 Stem Cell Transplant for Inborn Errors of Metabolism Stem Cell Transplant Phase 2/3 135 Completed (Has Results) NCT00176904
 Human Placental-Derived Stem Cell Transplantation Human Placental Derived Stem Cell Phase 1 43 Active, not recruiting NCT01586455
 UCB Transplant of Inherited Metabolic Diseases With Administration of Intrathecal UCB Derived Oligodendrocyte-Like Cells UCB-derived oligodendrocyte-like cells Phase 1 12 Recruiting NCT02254863
CLN1
 Cystagon to Treat Infantile Neuronal Ceroid Lipofuscinosis Cystagon and N-acetylcysteine (oral, daily) Phase 4 10 Completed227 NCT00028262
 Study of HuCNS-SC Cells in Patients With Infantile or Late Infantile Neuronal Ceroid Lipofuscinosis (NCL) Human CNS stem cells (HuCNS-SC) Phase 1 6 Completed NCT00337636
 Safety and Efficacy Study of HuCNS-SC in Subjects With Neuronal Ceroid Lipofuscinosis Human CNS stem cells (HuCNS-SC) Phase 1b 0 Withdrawn NCT01238315
CLN2
 Safety Study of a Gene Transfer Vector for Children With Late Infantile Neuronal Ceroid Lipofuscinosis AAV2CUhCLN2 (3×1012 particle units) Phase 1 10 Active, not recruiting NCT00151216
 Study of HuCNS-SC Cells in Patients With Infantile or Late Infantile Neuronal Ceroid Lipofuscinosis (NCL) Human CNS stem cells (HuCNS-SC) Phase 1 6 Completed NCT00337636
 Safety Study of a Gene Transfer Vector (Rh.10) for Children With Late Infantile Neuronal Ceroid Lipofuscinosis (LINCL) AAVrh.10CUhCLN2 (9.0×1011 or 2.85×1011 genome copies) Phase 1 25 Active, not recruiting NCT01161576
 Safety and Efficacy Study of HuCNS-SC in Subjects With Neuronal Ceroid Lipofuscinosis Human CNS stem cells (HuCNS-SC) Phase 1b 0 Withdrawn NCT01238315
 AAVRh.10 Administered to Children With Late Infantile Neuronal Ceroid Lipofuscinosis AAVrh.10CUhCLN2 Phase 1/2 8 Active, not recruiting NCT01414985
 A Phase 1/2 Open-Label Dose-Escalation Study to Evaluate Safety, Tolerability, Pharmacokinetics, and Efficacy of Intracerebroventricular BMN 190 in Patients With Late-Infantile Neuronal Ceroid Lipofuscinosis (CLN2) Disease BMN 190 (rhTPP1); ERT via ICV infusion Phase 1/2 24 Completed (Has Results) NCT01907087
 A Multicenter, Multinational, Extension Study to Evaluate the Long-Term Efficacy and Safety of BMN 190 in Patients With CLN2 Disease BMN 190 (rhTPP1); ERT via ICV infusion Phase 1/2 23 Active, not recruiting NCT02485899
 A Safety, Tolerability, and Efficacy Study of Intracerebroventricular BMN 190 in Pediatric Patients < 18 Years of Age With CLN2 Disease BMN 190 (rhTPP1); ERT via ICV infusion Phase 2 15 Enrolling by invitation NCT02678689
CLN3
 CellCept® for Treatment of Juvenile Neuronal Ceroid Lipofuscinosis Mycophenolate mofetil (CellCept®) Phase 2 19 Completed NCT01399047
 Phase I/IIa Gene Transfer Clinical Trial for Juvenile Neuronal Ceroid Lipofuscinosis, Delivering the CLN3 Gene by Self-Complementary AAV9 scAAV9-CLN3 Phase 1/2 7 Recruiting NCT03770572
CLN6
 Batten CLN6 Gene Therapy scAAV9.CB.CLN6 Phase 1/2 12 Recruiting NCT02725580
1

Abbreviations: enzyme replacement therapy (ERT); recombinant human tripeptidyl peptidase-1 (rhTPP1); adeno-associated virus (AAV); umbilical cord blood (UCB); neuronal ceroid lipofuscinosis (NCL); late infantile neuronal ceroid lipofuscinosis (LINCL).

2

Clinical identifier numbers obtained from ClinicalTrials.gov website.

7.1. Therapies for CLN1

7.1.1. ERT.

Pre-clinical studies have demonstrated recombinant PPT1 to be efficacious and safe214,215. Human PPT1 enzyme administered intrathecally on 3 consecutive days significantly extended the lifespan of the Ppt1 knockout mouse model when treated at 6 wk age215. Motor deterioration was delayed, and neuropathology was improved based on autofluorescence (associated with lysosomal storage), CD68, and GFAP immunostaining215.

7.1.2. Stem cell therapy.

Ppt1−/− mice implanted with human CNS stem cell neurospheres (hCNS-SCns) resulted in increased PPT1 production in the CNS and reduced lipofuscin storage216. Two Phase I clinical trials in CLN1 and CLN2 subjects, sponsored by StemCells, Inc (NCT00337636, NCT01238315, Table IV) have investigated the use of cultured human CNS stem cells implanted into the CNS for safety assessment217. The feasibility of neurosphere implantation and absence of transplantation-related serious adverse events warrants further study of HuCNS-SC transplantation as a potential treatment for CLN1. However, a second trial (NCT01238315) was withdrawn due to lack of patient accrual (clinicaltrial.gov).

7.1.3. Gene therapy.

Gene therapy studies in the CLN1 mouse model have demonstrated that a treatment with dual routes of administration appears to delay disease progression218,219. Dual intraparenchymal and intrathecal administration of an AAV9-hPPT1 vector into the CNS showed synergistic improvements in median lifespan and motor function, compared to treatment groups receiving only one route of administration220. The conclusion of these studies was that targeting the brain may not be sufficient and that targeting the spinal cord in addition leads to an improved outcome. A study (ABO-202), with orphan drug designation from the FDA and the European Medicines Agency (EMA), is evaluating self-complementary adeno-associated virus 9 (scAAV9) expressing the human CLN1 gene both systemically (intravenous) and to the CNS (intrathecal) in human clinical trial221. A third approach tested the AAV2 gene transfer vector to deliver the human PPT1 gene to the eye of Ppt1-deficient mice to correct CLN1-mediated vision degeneration and demonstrated a significant increase in retinal function and reduced loss of photoreceptors218,222. To treat both the CNS and systemic effects of CLN1 disease in the CLN1−/− mouse, CNS-directed AAV5-mediated gene therapy was combined with either bone marrow transplantation or systemic delivery of phosphocysteamine. Both were equally efficacious223,224.

7.1.4. Pharmacological treatments.

Numerous investigations have been evaluated for the capacity to reduce or ablate the storage defects of CLN1203,224226. Lysosomotropic drugs (such as phosphocysteamine) showed promise in blocking ceroid accumulation, reducing storage material, and improving lifespan of treated CLN1 model mice, cultured CLN1 cells, and human CLN1 patients203,224,226. A pilot clinical trial in CLN1 patients using a combination of cysteamine bitartrate (Cystagon®) and N-acetylcysteine (NCT00028262) resulted in modest improvement in patients carrying severe CLN1 mutations227. Anti-apoptotic drugs like flupirtine, retigabine, and anti-caspase-4 inhibitor (Z-LEVD-FMK) have been tested to inhibit the apoptotic effects and arrest neurodegenerative progression in CLN1 cultured patient lymphoblasts202,210,211.

Immunomodulatory compounds with acceptable tolerability profiles are being designed to modify the immune response of NCLs116. For example, Fingolimod works by sequestering lymphocytes in the lymph nodes and prevents them from being transported to the CNS, while teriflunomide works by inhibiting the production and function of activated T cells116. Pre-symptomatic mouse models of CLN1 disease were treated over a period of 5 months with the oral administration of one of two immunomodulatory compounds, fingolimod or teriflunomide116. At the end of the treatment period, both forms of immunomodulation reduced secondary neuroinflammation, the T cell count (in optic nerve and CNS), and microgliosis in the CNS. The treatments also reduced axonal damage, neuronal loss, retinal thinning, frequency of myoclonic jerks, and total brain atrophy116. These treatments are non-invasive, and might work in conjunction with gene therapy or ERT.

7.2. Therapies for CLN2

7.2.1. ERT.

ERT has been shown to have therapeutic effects in murine and canine CLN2 models. Weekly intrathecal injections of TPP1 recombinant protein in a TPP1-deficient mouse model resulted in a significantly improved median lifespan228. Even when mice were treated after the onset of disease symptoms (late-stage), ERT reversed the decline in motor function. In mice, the intrathecal route of administration resulted in an uneven distribution of TPP1 within the brains of treated animals, and repeated intrathecal administrations also became more difficult as the study progressed, due to scarring228. In contrast, in a canine model, direct administration of TPP1 via infusion into the CSF, repeated every other week, delayed the onset and progression of the disease without significant adverse effects229. Survival was prolonged, and cognitive function was preserved. The ERT approach requires a lifelong commitment of regular infusions. One issue with ERT therapy directly to the CNS is the general concern that once the CNS pathology is cured, other effects of the disease will become apparent. For instance, Katz et. al., observed elevated serum troponin levels in all ERT-treated CLN2 dogs, suggesting that the TPP1 deficiency in the heart was not corrected by the ERT administered to the CNS229. A related study looked at intravitreal (IVT) administration in the canine CLN2 model, with biweekly hTPP1 treatments starting at 3 months of age230. The treatment was well tolerated and was found to slow or prevent disease progression based on electroretinography (ERG) and post-mortem histopathology of the retinas230.

In a recently concluded clinical trial (NCT01907087) based on canine preclinical proof of concept data, repeat intraventricular administration of Cerliponase alfa (a recombinant proenzyme form of TPP1) slowed disease progression in treated CLN2 patients compared to untreated historical controls231. One challenge of this therapy is that 50% of the study participants experienced some adverse events relating to the delivery devices including device-related infections, device leakage, and needle displacement231. None of these adverse events were severe enough to warrant withdrawal from the trial. There was no evidence that cerliponase alfa evoked an immune response that impacted safety or efficacy232. Importantly, significant slowing of disease-related clinical decline was observed. Based on this study the FDA approved Brineura® (cerliponase alfa) in 2017231. Patients are given a 300 mg dose every other week, with drug delivery accomplished using a surgically-implanted ventricular reservoir and catheter231.

7.2.2. Stem cell therapy.

Preclinical studies have been undertaken to examine the effects of stem cell therapy of the retina pathology in a canine model of CLN2233. A single dose of autologous bone marrow-derived stem cells were transduced with an AAV2 expressing TPP1 construct, and implanted intravitreally into one eye of a group of miniature long-haired Dachshunds with a frame shift mutation causing a complete absence of TPP1 enzymatic activity. The treated eyes had a significant reduction in both the prevalence of retinal detachments and rate of progression of retinal degeneration when compared to control eyes of the same dogs implanted with control GFP-expressing stem cells233. As mentioned for CLN1, there have been 2 clinical trials studying the use of human CNS stem cells involving CLN1 and CLN2 patients (NCT00337636, NCT01238315). However, although a safe and tolerable outcome was reported, there were no clinical improvements217. See 7.1.2. for details of the trials.

7.2.3. Gene therapy.

Gene therapy is less invasive than ERT as it is a one-time administration in contrast to the lifelong repeated treatments. Numerous murine pre-clinical studies addressing CLN2 deficiency by treatment with various AAV serotypes have demonstrated high levels of TPP1 enzyme and reduction of storage material107,234237. Direct intraparenchymal administration of several AAV serotypes expressing CLN2 demonstrated that AAVrh.10hCLN2 gene therapy provided the best distribution in the CNS and reduction of disease phenotype in mice and widespread TPP1 distribution in nonhuman primates236. In a canine model of CLN2 disease, a single intracerebroventricular injection of rAAV2caTPP1 (AAV2 expressing the canine form of TPP1) was found to delay disease onset and progression, protect from cognitive decline, and extend the lifespan238. AAV-derived TPP1 expression in ependymal cells, which line the CSF-filled brain ventricles and the central canal of the spinal cord, was robust and widely distributed in brain parenchyma. Significantly, elevated AAV-derived TPP1 levels were also detected in peripheral organs, including the spleen and heart238. However, these levels of TPP1 expression were not enough to ameliorate non-neuronal pathology and the dogs still exhibited progressive cardiac, liver, and muscle pathology and impaired cardiac function, suggesting that the optimal treatment regimen may require systemic and CNS delivery of TPP1 genes238. Three clinical studies have assessed the potential for gene therapy for CLN2 using AAV2 (NCT00151216) and AAVrh.10 (NCT01161576, NCT01414985; Table IV). All of these studies used direct intraparenchymal delivery to the CNS2,190,239. Results for the AAV2 study demonstrated a statistically significant slowing of the neurologic decline in treated subjects vs published natural history controls190. The results for the AAVrh.10 study have not yet been published.

7.2.4. Pharmacological treatments.

Anti-apoptotic drugs like flupirtine have been tested to arrest neurodegenerative progression in CLN2-cultured lymphoblasts and neurons210. Retigabine, analogous to flupirtine, was shown to rescue CLN2 patient-derived lymphoblasts from accelerated apoptosis, and decreased ceramide accumulation211. Gemfibrozil has been shown to reduce inflammation and increase survival in treated Cln2 knockout mouse models213, and has been shown to be safe in children240.

7.3. Therapies for CLN3

7.3.1. ERT.

No published studies to date.

7.3.2. Stem cell therapy.

No published studies to date.

7.3.3. Gene therapy.

Preclinical testing of gene therapy using AAV vectors to treat CLN3 disease have shown efficacy in both cell culture and mouse models. AAV2-CLN3 was successful in restoring full-length CLN3 transcript and protein in iPSC-derived retinal neurons115, and CRISPR-Cas9 plasmids have been used to correct CLN3 patient-derived iPSCs241. Preclinical studies with intraparenchymally-administered AAVrh.10 or intravenously-administered scAAV9 vectors expressing the CLN3 gene partially correct mouse models of the disease113,114. These results have prompted a clinical trial with AAV9 for CLN3 patients (Amicus Therapeutics, NCT03770572), which is recruiting at Nationwide Children’s Hospital (Columbus, OH, USA) (Table IV). This non-randomized, open-label, dose-escalation clinical trial for efficacy and safety of a single administration of AAV9-CLN3 via the intrathecal route in CLN3 subjects (3 to 10 yr old children), will begin with a low-dose derived from the minimum effective dose (MED) as determined in non-clinical studies (NCT03770572). Upon demonstration of safety in this first cohort, dose escalation to a high-dose (2x MED) will proceed. Publications do not provide the starting dose.

7.3.4. Pharmacological treatments.

Three phosphodiesterase-4 (PDE4) inhibitors (rolipram, roflumilast, and PF-06266047) have been tested in Cln3Δex7/8 knockout mice to determine whether the resulting increase in cyclic adenosine monophosphate (cAMP) levels would have a beneficial neurological effect242. Cln3Δex7/8 mice have reduced brain cAMP levels, which could be corrected by inhibiting phosphodiesterases, which mediate cAMP degradation, and PDE4 is most prominently expressed in the CNS242. The use of PDE inhibitors limits neuronal apoptosis and neuroinflammation, as well as augment glutamate transporter expression242. Treatment with PDE4 inhibitors resulted in significant improvements in motor function; decreased neuroinflammation, glial activation, and lysosomal pathology; and restored glutamate transporter expression to levels comparable to wild-type animals242. These results support their potential use in treating CLN3. In another study, AMPA receptor antagonists/attenuators (EGIS-8332) delivered in low doses significantly improved motor skills of CLN3 mice207. Retigabine, was observed to be neuroprotective in CLN3-defective cultured cells225 and rescued CLN3 patient-derived lymphoblasts from accelerated apoptosis as well as decreasing ceramide accumulation, demonstrating potential for anti-apoptotic drugs as treatment of NCL disease211. A survey was conducted among parents of CLN3 patients undergoing treatment with the anti-apoptotic drug flupirtine. However, the statistical analysis of the reported beneficial effect from the treatment, based on the Unified Batten disease rating scale, showed no difference between treatment and control groups243.

Another suitable target for pharmaceutical treatment is the transcription factor EB (TFEB) which serves to regulate cellular clearance by enhancing processes such as lysosomal proliferation, expression of degradative enzymes, autophagy, lysosomal exocytosis and lysosomal proteostasis21,244. Both trehalose, the non-reducing disaccharide of glucose, and the drug MK2206 have been shown to inhibit the serine/threonine kinase Akt (protein kinase B), which has in turn been shown to control TFEB activity through phosphorylation21,244. Cln3Δex7/8 mice that were given oral trehalose treatments had extended lifespans, clearance of lysosomal storage, and reduced neuroinflammation and neurodegeneration244. Intraperitoneal administration of MK2206 resulted in nuclear translocation of TFEB and upregulation of lysosomal and autophagy genes in the brains of Cln3Δex7/8 mice244. When tested on fibroblasts derived from patients with CLN1, CLN2, CLN3, and CLN7 disease, MK2206 enhanced cellular clearance of ceroid lipopigment deposits244.

CLN3-affected individuals have increased autoantibody formation against CNS antigens245. Preclinical studies in the CLN3-deficient mice have found elevated autoantibody titers246,247, leading to studies to test immunosuppressive agents in these mice248. Immunosuppression with mycophenolate mofetil administered by gavage for 30, 70, or 150 consecutive days was found to alter the disease severity in CLN3 mice with a significant improvement in motor skills248. Further treatment with the immunomodulators (section 7.1.4) fingolimod and teriflunomide on a CLN3 mouse model were found to have similar outcomes in pre-symptomatic mice116. The results suggest that secondary neuroinflammation is a relevant mechanism for CLN3 disease progression. Additional pharmacological experiments with an in vitro cell culture model of CLN3 disease found that treatment with lithium improved defective autophagy249. Autofluorescence and accumulation of mitochondrial ATP synthase subunit c were both reduced, and the lithium-treated cells were more resistant to autophagic stress-triggered cell death250. The mechanism of action of the neuroprotective effect of lithium was proposed to be through inhibition of inositol monophosphatase, which improves the defective autophagy250.

The immunosuppressant mycophenolate mofetil (CellCept®), approved by the FDA as an anti-rejection medication following organ transplantation, was tested in CLN3 patients for controlling secondary autoimmunity and neuroinflammation (NCT01399047)251. The trial (double-blind, placebo-controlled), though limited to the short-term (8 wk), found that mycophenolate was well-tolerated. The next step is to examine clinical impact with a longer-term treatment regimen. A similar therapeutic strategy tested a regimen of prednisolone eye drops, triamcinolone (intravitreal), mycophenolate mofetil (oral), along with brinzolamide and nepafenac anti-inflammatory medications over a 2 yr period to a CLN3 patient (7.5 yo) with decreased visual acuity252. The patient retained ambulatory vision after 2 yr, which is normally absent in CLN3 subjects of comparable age, suggesting that inflammation of the retina plays a role in vision loss in CLN3 subjects252.

7.4. Therapies for CLN5

7.4.1. ERT.

No published studies to date.

7.4.2. Stem cell therapy.

No published studies to date.

7.4.3. Gene therapy.

Neural cells cultured from prenatal CLN5-deficient sheep brains rapidly accumulate storage bodies in culture, mainly in microglial cells and also in neurons and astrocytes200. Transduction of these cells with a recombinant lentiviral vector expressing ovine CLN5 rapidly reversed accumulation of storage material with a 50% reduction of cell-contained storage bodies 6 days after treatment200. Direct intraparenchymal and intracerebroventricular injections of lentiviral vectors in juvenile CLN5 sheep successfully delivered the transgene, but the doses appeared to be too low for therapeutic effect as there was no improvement in disease progression201. Juvenile CLN5 sheep (2–3 months of age before the onset of clinical symptoms) receiving higher doses of lentiviral vectors as well as AAV9 vectors have remained disease-free 4 to 5 months beyond the typical age of end-stage clinical disease122. All AAV9-CLN5 or lentivirus-CLN5 treated animals appeared phenotypically normal, with delayed-onset visual loss being the only clinical symptom122. In a cohort of the same study in which a self-complementary AAV9 vector was administered to older CLN5 sheep (at 7 months, the early stage of clinical disease), there was attenuation of disease progression and extension of lifespan but visual loss remained122.

7.4.4. Pharmacological treatments.

No published studies to date.

7.5. Therapies for CLN6

7.5.1. ERT.

No published studies to date.

7.5.2. Stem cell therapy.

No published studies to date.

7.5.3. Gene therapy.

One of the hallmarks of CLN6 disease is the rapid loss of vision which provides a relatively good phenotype for evaluating efficacy253. Early studies with AAV8-CLN6 failed to modulate vision loss40, but a current study of AAV2–7m8 to target the inner retina bipolar cells significantly slowed loss of photoreceptor cells and function in Cln6nclf mice125. Combination gene therapy targeting the CNS and the eye may be necessary to treat all aspects of the CLN6 disease.

Using the self-complementary AAV9 (scAAV9), significant reversal of phenotypic defect in Cln6nclf mice has been achieved40. Cain et al254 delivered scAAV9-hCLN6 to the cerebral ventricles of Cln6nclf mice resulting in improved behavioral motor skills and decreased autofluorescent lysosomal storage material at up to 3 months of age254. Longer-term studies with the same vector and route resulted in reduction of autofluorescent substrate material throughout the brain and spinal cord, and improved motor performance and cognitive behavior out to 22 months with median survival extended to 22 months, significantly greater than the 12 to 14 months for untreated mice255. There were significant reductions in astrocyte and microglial activation in the same model at 18 months256. A follow-up safety study looked at 3 Cynomolgus macaques treated with scAAV9-hCLN6 by intrathecal lumbar injection. Over the 6 month course of the study, there were no adverse events or apparent pathology observed. High levels of transgene expression were observed in both the brain and the spinal cord255. The successful proof of concept in the mouse and the distribution and safety data from the non-human primate has been used to support a clinical phase I/II trial for CLN6 patients21,40. Using 1.5 × 1013 vector genomes (vg) of a self-complementary AAV serotype 9 vector to deliver CLN6 cDNA (scAAV9.CB.CLN6) intrathecally (lumbar puncture) to patients (n=12 proposed, 10 treated currently), an open-label, single-dose clinical trial is in progress at Nationwide Children’s Hospital (NCT02725580, Table IV)257,258. No results have been published for this trial, but Amicus Therapeutics, who has licensed this product, has shown encouraging interim data on 2 treated subjects at NCL meetings259,260. Two siblings with the same genotype were treated with a single intrathecal administration of AAV9-CLN6, at ages 2.8 and 5.3 years of age. Two years post-treatment, the younger sibling’s motor and language scores showed no evidence of disease progression, while the older sibling’s scores indicated stabilization of disease progression259.

In studies in a large animal model, CLN6−/− South Hampshire sheep were treated with either lentiviral or AAV9 viral vectors expressing CLN6. The ongoing trial seeks to assess improvements via CT scans and behavioral analyses (including vision, cognitive score, and maze navigation) to determine if the expression is sufficient to delay or halt disease progression261. Direct intraparenchymal and intracerebroventricular injections of lentiviral vectors in juvenile CLN6 sheep successfully delivered the transgene, but the doses appeared to be too low and there was no improvement in disease progression201. Higher doses of lentiviral vectors as well as AAV9 vectors in juvenile CLN6 sheep demonstrated only one of six treated animals with clinical benefit. This is in contrast to the CLN5 study in sheep in which all treated animals showed great improvement comparable to normal controls201. An in vitro study with mixed neural cultures isolated from the CLN6 sheep, found treatment with a lentiviral vector delivering the wild-type CLN6 gene was able to repair disease-related changes in lysosomal acidity, autophagic flux, and synaptic endocytosis121.

7.5.4. Pharmacological treatments.

Anti-apoptotic drugs like flupirtine have been tested to prevent apoptosis in CLN6-deficient lymphoblast cultured cells210,225. Retigabine, was observed to be neuroprotective and rescued CLN6 patient-derived lymphoblasts from accelerated apoptosis, and decreased ceramide accumulation211. Dietary supplementation of CLN6nclf mice with naturally-occurring immunomodulators curcumin and docosahexaenoic acid (DHA) was found to result in significantly higher visual acuity and higher ERG amplitudes compared to untreated control CLN6nclf mice. Retinal morphology in DHA-supplemented mice was significantly preserved compared to wild-type mice, curcumin-treated CLN6nclf mice, and untreated CLN6nclf253. Oral administration of the anti-inflammatory drug minocycline was evaluated in CLN6 NCL South Hampshire lambs, but chronic drug administration over the course of 3 to 14 months did not change disease progression201,262

7.6. Therapies for CLN7

7.6.1. ERT.

No published studies to date.

7.6.2. Stem cell therapy.

No published studies to date.

7.6.3. Gene therapy.

While no traditional gene therapy studies have been published for CLN7, a study with an antisense oligonucleotide therapy was used to treat one individual with CLN7 disease. The antisense oligonucleotide drug was specially tailored for this individual’s mutation, and preliminary results suggest that the treatment has appeared to halt further deterioration, with reduced seizure intensity and frequency263.

7.6.4. Pharmacological treatments.

No published studies to date.

7.7. Therapies for CLN8

7.7.1. ERT.

No published studies to date.

7.7.2. Stem cell therapy.

Bone marrow transplants in CLN8 mouse and dog models led to CLN8 expression in marrow but not in CNS264.

7.7.3. Gene therapy.

No published studies to date.

7.7.4. Pharmacological treatments.

Several early studies in CLN8-deficient mice showed improvements with AMPA receptor attenuator (ZK-187638) therapy, including improvements in motor coordination; however, nothing new has been published since the 2006 study265. Retigabine, was observed to be neuroprotective and rescued CLN8 patient-derived lymphoblasts from accelerated apoptosis, and decreased ceramide accumulation211. Other interventions tested for CLN8 include mice administered IGF-1 (insulin-like growth factor 1) by infusion into brain ventricles, resulting in neuroprotection266. β2-adrenergic agonists have mediated motor neuron improvements in CLN8 mice267.

7.8. Therapies for CLN10

7.8.1. ERT.

No published studies to date.

7.8.2. Stem cell therapy.

No published studies to date.

7.8.3. Gene therapy.

In a studies published in 2010 and 2011, AAV therapy with mCathD transgene administered intraparenchymally or intraventricularly in CLN10 mice demonstrated cathepsin D expression and enhanced lifespan268,269. No further studies have been reported.

7.8.4. Pharmacological treatments.

No published studies to date.

7.9. Therapies for CLN11

7.9.1. ERT.

No published studies to date.

7.9.2. Stem cell therapy.

No published studies to date.

7.9.3. Gene therapy.

AAV1 vectors that mediate progranulin were administered to the medial prefrontal cortex (mPFC) in Grn−/− mice with a resulting reduction in lipofuscinosis and microgliosis, even in regions where progranulin expression was low, suggesting that only a small number of cells needs to be transduced for efficacy. Progranulin expression was detectable in neurons but not in microglia, suggesting that microgliosis is partly due to neuronal dysfunction and this can be at least partially repaired270. Intracerebroventricular delivery of AAV9-GRN mediated high levels of broad and sustained GRN expression, but GRN overexpression led to cell toxicity and hippocampal neurodegeneration preceded by T cell infiltration271. When repeated with an AAV4 vector, severe ependymal and choroidal hypertrophy was observed with corresponding T cell infiltration271. Control mice injected with EGFP-expressing vectors did not experience the same issues, suggesting that the overexpression of GRN, not the AAV vector, may be harmful. While a localized immune response was seen in the AAV1 study, the AAV9 and AAV4 study used higher doses and followed the treatment for a longer study period. The higher expression levels may have caused toxicity or the earlier study was not long enough to observe toxicity271. Before clinical translation of the therapy can occur, a safe level of expression has to be determined which maintains efficacy.

7.9.4. Pharmacological treatments.

No published studies to date.

8. Academic and Industry Partnerships

The most promising treatment modalities have been successfully translated from animal work into clinical trials for CLNs 1, 2, 3, and 6 [such as NCT00028262 (ERT, CLN1), NCT00151216 (AAV2, CLN2), NCT00337636 (stem cells, CLN1), NCT01161576 (AAVrh.10, CLN2), NCT01238315 (stem cells, CLN1), NCT01399047 (Cellcept®, CLN3), NCT01414985 (AAVrh.10, CLN2), NCT01907087 (ERT, CLN2), and NCT02725580 (AAV9, CLN6); ClinicalTrials.gov]37. More recently, the FDA approved the first ever treatment for CLN2 disease, an enzyme replacement therapy called Brineura®231. The partnership between investigators and Biotech startups have resulted in numerous companies involved in NCL research and clinical trials. Based on a survey of the field, we have summarized the companies (listed alphabetically) that show an NCL disease as part of their product pipeline. This is not an exhaustive list, but provides insight to the progress that has been made in the development of therapeutics for these rare and devastating diseases.

Abeona Therapeutics (Dallas, TX, USA) – CLN1 and CLN3 gene therapy clinical trials with AAV9 (Phase 1/2) in 2019272.

Amicus Therapeutics (Cranbury, NJ, USA) – CLN3, CLN6, and CLN8 – gene therapy applications with clinical trials (Phase 1/2) in 2019273.

BioMarin (Novato, CA, USA) – ERT for CLN2 disease (with clinical trials NCT02485899, NCT02678689)274.

Circumvent Pharmaceuticals (Portland, OR, USA) – CLN1, CLN3, CLN4, CLN10 – developing small molecule thioesterase mimetics275.

Genentech (South San Francisco, CA, USA) – CLN3 with CellCept® (mycophenolic mofetil) immunosuppressive drug (with clinical trial NCT01399047).

Polaryx Therapeutics (Paramus, NJ, USA) – CLN2 and CLN3 – developing small molecule therapy (PLX-200, Gemfibrozil), an FDA-approved lipid lowering agent that increases lysosome biogenesis, anti-inflammatory mediators, remyelination, and neurotrophins, with clinical trials expected in 3Q 2019 in the US276.

REGENXBIO (Rockville, MD, USA) – CLN2 – developing gene therapy for the treatment of CLN2 using RGX-181 (AAV9-hTPP1) by direct CNS injections to augment TPP1 in children with CLN2277.

Spark Therapeutics (Philadelphia, PA, USA) – developing gene therapy for CLN2 using SPK-TPP1 vector278.

9. Conclusions

These are exciting times for treatment of NCLs, with multiple therapeutics reaching the clinical trial stage for several CLN subtypes. The potential for cures could be at hand after many years of trial and error in the preclinical studies. The clinical development of ERT for CLN2 (Brineura® by BioMarin); immunosuppression for CLN3 (CellCept® by Genentech); and gene therapy vectors for CLN1 (ABO-202 by Abeona), CLN2 (RGX-181 by REGENXBIO, SPK-1001 by Spark Therapeutics), CLN3 (ABO-201 by Abeona, AAV9-CLN3 by Amicus), CLN6 (AAV9-CLN6 by Amicus), and CLN8 (AAV9-CLN8 by Amicus) have given NCL patients and families hope that the neurodgerenative pathology can be abated or reversed and clinical outcomes can yield improved quality of life152,279. With the milestone approval of the two gene therapy drugs, Luxturna (AAV2-RPE65) by both the FDA and European Commission and Zolgensma (AAV9-SMN) by the FDA280,281, gene therapy is finally becoming a reality for other orphan diseases such as the NCLs. However, much work remains to be done before a cure for any of the NCLs is widely available. As seen with the Luxturna and Zolgensma trials, the cost of the drug and benefits for all patients must be weighed for each treatment modality282.

10. Expert Opinion

The NCL family of diseases, like many other lysosomal storage disorders, have neurological involvement, making them a challenging target for treatment. However, emerging therapies, especially the progress made in the last decade, are overcoming these barriers and providing hope for patients and their families. With increased interest by both academic scientists and biotechnology companies, and with support from patient advocacy groups and disease-specific foundations, progress has been made in defining the natural history of these diseases, providing a critical standard by which to compare the outcomes of candidate therapies. NCL clinical research teams from all over the world are also working together to maintain and provide access to patient registries. Also important are the newly-identified biomarkers of disease progression that are unique to a given NCL and enable in-life measures of the impact of a therapeutic modality. Well-defined natural history studies and unique biomarkers of disease progression will be increasingly important as they help identify the milestones for outcomes in clinical trials that aid in the assessment of efficaciousness and comparisons of new therapies.

When considering different therapeutic approaches, ERT may conceptually be the simplest, but the blood-brain barrier is a major hurdle to the therapeutic enzyme accessing the CNS, the most important target in all NCLs. The recent approval for intraventricular administration of Cerliponase Alfa for the treatment of CLN2 disease is a significant step forward in the field and opens up the possibility of expanding ERT not just to other NCLs but also to other LSDs with neurological involvement231. While this is a huge breakthrough, it must be remembered that ERT requires lifelong repeated administrations, a significant confounding factor for reasons of cost and access to therapy.

In theory, cell therapies offer promise for the treatment of these diseases, but preclinical studies to date have been confounded by limited migration from the sites of administration to encompass all the affected areas of the brain.

Gene therapies on the other hand, could be the best solution. Once administered, a gene therapy expresses the gene of interest on a persistent bases (AAV or lentivirus vectors) with the potential to be a “one and done” treatment. In the last decade, many gene therapy studies outlined in this review have demonstrated success in animal models (small and large) of NCLs. Improved vector serotypes, and less invasive delivery methods leading to widespread distribution of the therapeutic product in the CNS, have been key to these successes. Many of these preclinical studies originating in academic labs have translated to early-phase clinical studies, and numerous industry partners facilitating clinical development projects with anticipated rapid regulatory approval.

Pharmacological approaches, in addition to providing palliative care for patients with these diseases, have the potential to target processes that have an impact on the disease. Examples of these approaches are anti-apoptotic drugs, immunosuppressants, and specific receptor antagonists, but none have yet led to definitive clinical benefits. The search for these small molecule drugs with efficacy continues as partnerships between pharmaceutical companies with access to large libraries of novel and repurposed agents, and scientists who provide the screening tools and animal models, is growing. This increased interest in the field indicates that new therapies may be on the horizon.

It is important to note that most of treatments being developed for NCLs target the CNS, but it is becoming clear that treating the peripheral manifestations will also be essential. For example, studies done in the dog model of CLN2 disease, where AAV-mediated delivery of CLN2 via intracerebroventricular administration showed the delay in onset of neurological symptoms, and increased survival was observed in the context of progressive cardiac disease as well as liver and muscle pathology101. Studies such as this point to the fact that when the more severe and fatal neurological pathologies are resolved, the current non-life threatening peripheral symptoms come to the forefront. Therefore, there may be a need for combinatorial approaches that deliver the same therapeutic to both the CNS and the viscera. Further, the eye disease associated with many NCLs needs to be addressed in parallel to prevent the blindness.

Finally, and critically important, is the early diagnosis of an NCL. Future newborn screening programs will enable diagnosis shortly after birth, allowing the initiation of therapy, when indicated, during asymptomatic stages. There are many therapies in the pipeline for the treatment of NCLs. Most of these may halt or slow the progression of disease but are unlikely to completely or even partially reverse the disease, therefore early diagnosis is more important than ever. In the US, many states have included newborn screening for some lysosomal disorders in their screening panel283, and with the new therapeutics, both approved (Brineura®) and clinical development of others, it will not be long before various NCL diseases have therapies available. In the next decade, the possibility of early diagnosis combined with effective therapies should change the outlook for patients with these fatal diseases.

Article Highlights.

  • Neuronal ceroid lipofuscinoses (NCL) are a collection of neurologic disorders involving defective lysosomal processing enzymes or receptors that lead to cognitive and visual impairments, seizures, deterioration of motor/language skills, and premature death.

  • Many experimental animal models (mice, dogs, pigs, and sheep) for the NCLs have been created or identified, which possess genomic mutations responsible for NCL subtypes, allowing investigations of safety and efficacy of new therapeutics in preclinical studies.

  • Development of quantitative biomarkers in the blood or CSF for NCLs, enabling investigators to track biochemically the improvement or decline of disease-related phenotypes, is an important component in the validation of potential treatments.

  • Review of the challenges of delivery of treatments, primarily due to CNS involvement and the blood-brain barrier, and possible therapeutic approaches for the development of treatment modalities for NCL, such as enzyme replacement therapy, cell therapy, gene therapy, and pharmacological drugs.

  • Current therapies for each of the NCL subtypes are discussed with emphasis on the most promising ones being translated to clinical trials, such as clinical development of ERT for CLN2 (Brineura®), immunosuppression for CLN3 (CellCept®), and gene therapy vectors for CLN1, CLN2, CLN3 and CLN6.

  • Gene therapy clinical trials using adeno-associated virus (AAV) to deliver the normal NCL-relevant gene to the CNS are underway and are being evaluated for safety and efficacy.

  • A combination approach using multiple treatment modalities may be the key to developing the most promising therapeutic approaches for NCLs.

Acknowledgments.

These studies were supported, in part, by 1R01NS061848, U01NS047458, and Foundation for Life, Chicago, IL.

Abbreviations used:

NCL

neuronal ceroid lipofuscinoses

CLN

ceroid lipofuscinosis, neuronal

LSD

lysosomal storage disorders

CNS

central nervous system

BBB

blood-brain barrier

PPT1

palmitoyl protein thioesterase-1

TPP1

tripeptidyl peptidase-1

ERT

enzyme replacement therapy

AAV

adeno-associated virus

INCL

infantile NCL

LINCL

late infantile NCL

JNCL

juvenile NCL

SCMAS

subunit c of mitochondrial ATP synthase

SAP

sphingolipid activator proteins

BMP

bis(monoacylglycero)phosphate

HSC

hematopoietic stem cells

HuCNS-SC

human central nervous system stem cells

CTSD

cathepsin D

GRN

granulin precursor

PGRN

progranulin

BDNF

brain-derived neurotrophic factor

MLD

metachromatic leukodystrophy

M6P

mannose 6-phosphate

AMPA

α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid

FDA

Food and drug administration

MED

minimum effective dose

n.d.

not determined

Footnotes

Declaration of Interest. RGC is a consultant to BioMarin. All other authors declare no conflict of interest.

References.

  • 1.Schulz A, Kohlschutter A, Mink J, Simonati A, Williams R. NCL diseases - clinical perspectives. Biochim Biophys Acta. 2013; 1832:1801–1806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Sondhi D, Crystal RG, Kaminsky SM. Gene Therapy for Inborn Errors of Metabolism: Batten Disease In: Tuszynski MH, editor. Translational Neuroscience: Fundamental Approaches for Neurological Disorders. Boston, MA: Springer US; 2016. p. 111–129. [Google Scholar]
  • 3.Donsante A, Boulis NM. Progress in gene and cell therapies for the neuronal ceroid lipofuscinoses. Expert Opin Biol Ther. 2018; 18:755–764. [DOI] [PubMed] [Google Scholar]
  • 4.Mole SE, Anderson G, Band HA, Berkovic SF, Cooper JD, Kleine Holthaus SM, McKay TR, Medina DL, Rahim AA, Schulz A, Smith AJ. Clinical challenges and future therapeutic approaches for neuronal ceroid lipofuscinosis. Lancet Neurol. 2019; 18:107–116. [DOI] [PubMed] [Google Scholar]
  • 5.Rakheja D, Bennett MJ. Neuronal ceroid-lipofuscinoses. Transl Sci Rare Dis. 2018; 3:83–95. [Google Scholar]
  • 6.Johnson TB, Cain JT, White KA, Ramirez-Montealegre D, Pearce DA, Weimer JM. Therapeutic landscape for Batten disease: current treatments and future prospects. Nat Rev Neurol. 2019; 15:161–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Platt FM. Emptying the stores: lysosomal diseases and therapeutic strategies. Nat Rev Drug Discov. 2018; 17:133–150. [DOI] [PubMed] [Google Scholar]
  • 8.Glees P, Hasan M. Lipofuscin in neuronal aging and diseases. Norm Pathol Anat (Stuttg). 1976; 32:1–68. [PubMed] [Google Scholar]
  • 9.Cooper JD. The neuronal ceroid lipofuscinoses: the same, but different? Biochem Soc Trans. 2010; 38:1448–1452. [DOI] [PubMed] [Google Scholar]
  • 10.Palmer DN, Martinus RD, Cooper SM, Midwinter GG, Reid JC, Jolly RD. Ovine ceroid lipofuscinosis. The major lipopigment protein and the lipid-binding subunit of mitochondrial ATP synthase have the same NH2-terminal sequence. J Biol Chem. 1989; 264:5736–5740. [PubMed] [Google Scholar]
  • 11.Tyynela J, Palmer DN, Baumann M, Haltia M. Storage of saposins A and D in infantile neuronal ceroid-lipofuscinosis. FEBS Lett. 1993; 330:8–12. [DOI] [PubMed] [Google Scholar]
  • 12.Jalanko A, Braulke T. Neuronal ceroid lipofuscinoses. Biochim Biophys Acta. 2009; 1793:697–709. [DOI] [PubMed] [Google Scholar]
  • 13.Anderson GW, Goebel HH, Simonati A. Human pathology in NCL. Biochim Biophys Acta. 2013; 1832:1807–1826. [DOI] [PubMed] [Google Scholar]
  • 14.Mole SE, Haltia M. Chapter 70 - The Neuronal Ceroid-Lipofuscinoses (Batten Disease) In: Rosenberg RN, Pascual JM, editors. Rosenberg’s Molecular and Genetic Basis of Neurological and Psychiatric Disease (Fifth Edition). Boston: Academic Press; 2015. p. 793–808. [Google Scholar]
  • 15.Levine AS, Lemieux B, Brunning R, White JG, Sharp HL, Stadlan E, Krivit W. Ceroid accumulation in a patient with progressive neurological disease. Pediatrics. 1968; 42:583–591. [PubMed] [Google Scholar]
  • 16.Cotman SL, Staropoli JF. The juvenile Batten disease protein, CLN3, and its role in regulating anterograde and retrograde post-Golgi trafficking. Clin Lipidol. 2012; 7:79–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Santavuori P Neuronal ceroid-lipofuscinoses in childhood. Brain Dev. 1988; 10:80–83. [DOI] [PubMed] [Google Scholar]
  • 18.Fietz M, AlSayed M, Burke D, Cohen-Pfeffer J, Cooper JD, Dvorakova L, Giugliani R, Izzo E, Jahnova H, Lukacs Z, Mole SE, Noher de Halac I, Pearce DA, Poupetova H, Schulz A, Specchio N, Xin W, Miller N. Diagnosis of neuronal ceroid lipofuscinosis type 2 (CLN2 disease): Expert recommendations for early detection and laboratory diagnosis. Mol Genet Metab. 2016; 119:160–167. [DOI] [PubMed] [Google Scholar]
  • 19.Palmer DN, Barry LA, Tyynela J, Cooper JD. NCL disease mechanisms. Biochim Biophys Acta. 2013; 1832:1882–1893. [DOI] [PubMed] [Google Scholar]
  • 20.Haltia M The neuronal ceroid-lipofuscinoses. J Neuropathol Exp Neurol. 2003; 62:1–13. [DOI] [PubMed] [Google Scholar]
  • 21.Kohlschutter A, Schulz A, Bartsch U, Storch S. Current and Emerging Treatment Strategies for Neuronal Ceroid Lipofuscinoses. CNS Drugs. 2019; 33:315–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Markham A Cerliponase Alfa: First Global Approval. Drugs. 2017; 77:1247–1249. [DOI] [PubMed] [Google Scholar]
  • 23.Cardona F, Rosati E. Neuronal ceroid-lipofuscinoses in Italy: an epidemiological study. Am J Med Genet. 1995; 57:142–143. [DOI] [PubMed] [Google Scholar]
  • 24.Augestad LB, Diderichsen J. [Neuronal ceroid lipofuscinoses]. Tidsskr Nor Laegeforen. 2006; 126:1908–1910. [PubMed] [Google Scholar]
  • 25.Mink JW, Augustine EF, Adams HR, Marshall FJ, Kwon JM. Classification and natural history of the neuronal ceroid lipofuscinoses. J Child Neurol. 2013; 28:1101–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Nita DA, Mole SE, Minassian BA. Neuronal ceroid lipofuscinoses. Epileptic Disord. 2016; 18:73–88. [DOI] [PubMed] [Google Scholar]
  • 27.Mole SE, Williams RE, Goebel HH. Correlations between genotype, ultrastructural morphology and clinical phenotype in the neuronal ceroid lipofuscinoses. Neurogenetics. 2005; 6:107–126. [DOI] [PubMed] [Google Scholar]
  • 28.Getty AL, Pearce DA. Interactions of the proteins of neuronal ceroid lipofuscinosis: clues to function. Cell Mol Life Sci. 2011; 68:453–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Carcel-Trullols J, Kovacs AD, Pearce DA. Cell biology of the NCL proteins: What they do and don’t do. Biochim Biophys Acta. 2015; 1852:2242–2255. [DOI] [PubMed] [Google Scholar]
  • 30.Kyttala A, Lahtinen U, Braulke T, Hofmann SL. Functional biology of the neuronal ceroid lipofuscinoses (NCL) proteins. Biochim Biophys Acta. 2006; 1762:920–933. [DOI] [PubMed] [Google Scholar]
  • 31.Kousi M, Lehesjoki AE, Mole SE. Update of the mutation spectrum and clinical correlations of over 360 mutations in eight genes that underlie the neuronal ceroid lipofuscinoses. Hum Mutat. 2012; 33:42–63. [DOI] [PubMed] [Google Scholar]
  • 32.Kmoch S, Stranecky V, Emes RD, Mitchison HM. Bioinformatic perspectives in the neuronal ceroid lipofuscinoses. Biochim Biophys Acta. 2013; 1832:1831–1841. [DOI] [PubMed] [Google Scholar]
  • 33.Sondhi D, Rosenberg JB, Van de Graaf BG, Kaminsky SM, Crystal RG. Advances in the treatment of neuronal ceroid lipofuscinosis. Expert Opinion on Orphan Drugs. 2013; 1:951–975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Faller KM, Gutierrez-Quintana R, Mohammed A, Rahim AA, Tuxworth RI, Wager K, Bond M. The neuronal ceroid lipofuscinoses: Opportunities from model systems. Biochim Biophys Acta. 2015; 1852:2267–2278. [DOI] [PubMed] [Google Scholar]
  • 35.Neverman NJ, Best HL, Hofmann SL, Hughes SM. Experimental therapies in the neuronal ceroid lipofuscinoses. Biochim Biophys Acta. 2015; 1852:2292–2300. [DOI] [PubMed] [Google Scholar]
  • 36.Tarczyluk MA, Cooper JD. Investigative and emerging treatments for Batten disease. Expert Opinion on Orphan Drugs. 2015; 3:1031–1045. [Google Scholar]
  • 37.Geraets RD, Koh S, Hastings ML, Kielian T, Pearce DA, Weimer JM. Moving towards effective therapeutic strategies for Neuronal Ceroid Lipofuscinosis. Orphanet J Rare Dis. 2016; 11:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Grisolia M, Sestito S, Ceravolo F, Invernizzi F, Salpietro V, Polizzi A, Ruggieri M, Garavaglia B, Concolino D. The Neuronal Ceroid Lipofuscinoses: A Case-Based Overview. J Pediatr Biochem 2016; 06:60–65. [Google Scholar]
  • 39.Nelvagal HR, Cooper JD. Translating preclinical models of neuronal ceroid lipofuscinosis: progress and prospects. Expert Opinion on Orphan Drugs. 2017; 5:727–740. [Google Scholar]
  • 40.Kleine Holthaus S-M, Smith AJ, Mole SE, Ali RR, editors. Gene Therapy Approaches to Treat the Neurodegeneration and Visual Failure in Neuronal Ceroid Lipofuscinoses. 2018; Cham: Springer International Publishing. [DOI] [PubMed] [Google Scholar]
  • 41.Kim SJ, Zhang Z, Sarkar C, Tsai PC, Lee YC, Dye L, Mukherjee AB. Palmitoyl protein thioesterase-1 deficiency impairs synaptic vesicle recycling at nerve terminals, contributing to neuropathology in humans and mice. J Clin Invest. 2008; 118:3075–3086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Santavuori P, Haltia M, Rapola J, Raitta C. Infantile type of so-called neuronal ceroid-lipofuscinosis. 1. A clinical study of 15 patients. J Neurol Sci. 1973; 18:257–267. [DOI] [PubMed] [Google Scholar]
  • 43.Ramadan H, Al-Din AS, Ismail A, Balen F, Varma A, Twomey A, Watts R, Jackson M, Anderson G, Green E, Mole SE. Adult neuronal ceroid lipofuscinosis caused by deficiency in palmitoyl protein thioesterase 1. Neurology. 2007; 68:387–388. [DOI] [PubMed] [Google Scholar]
  • 44.Jeung H, Thomann PA, Wolf RC. Novel gene variations in early-onset frontotemporal dementia with positive family history of neural ceroid lipofuscinosis-1. Neurol Clin Pract. 2015; 5:484–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Delague V, Bareil C, Bouvagnet P, Salem N, Chouery E, Loiselet J, Megarbane A, Claustres M. Nonprogressive autosomal recessive ataxia maps to chromosome 9q34–9qter in a large consanguineous Lebanese family. Ann Neurol. 2001; 50:250–253. [PubMed] [Google Scholar]
  • 46.Sleat DE, Donnelly RJ, Lackland H, Liu CG, Sohar I, Pullarkat RK, Lobel P. Association of mutations in a lysosomal protein with classical late-infantile neuronal ceroid lipofuscinosis. Science. 1997; 277:1802–1805. [DOI] [PubMed] [Google Scholar]
  • 47.Steinfeld R, Heim P, von Gregory H, Meyer K, Ullrich K, Goebel HH, Kohlschutter A. Late infantile neuronal ceroid lipofuscinosis: quantitative description of the clinical course in patients with CLN2 mutations. Am J Med Genet. 2002; 112:347–354. [DOI] [PubMed] [Google Scholar]
  • 48.Williams RE, Adams HR, Blohm M, Cohen-Pfeffer JL, de Los Reyes E, Denecke J, Drago K, Fairhurst C, Frazier M, Guelbert N, Kiss S, Kofler A, Lawson JA, Lehwald L, Leung MA, Mikhaylova S, Mink JW, Nickel M, Shediac R, Sims K, Specchio N, Topcu M, von Lobbecke I, West A, Zernikow B, Schulz A. Management Strategies for CLN2 Disease. Pediatr Neurol. 2017; 69:102–112. [DOI] [PubMed] [Google Scholar]
  • 49.Dyke JP, Sondhi D, Voss HU, Yohay K, Hollmann C, Mancenido D, Kaminsky SM, Heier LA, Rudser KD, Kosofsky B, Casey BJ, Crystal RG, Ballon D. Brain Region-Specific Degeneration with Disease Progression in Late Infantile Neuronal Ceroid Lipofuscinosis (CLN2 Disease). AJNR Am J Neuroradiol. 2016; 37:1160–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Dyke JP, Voss HU, Sondhi D, Kaminsky SM, Blatteis J, Heier LA, Kosofsky B, Casey BJ, Crystal RG, Ballon D. Asymptotic neurodegeneration in CLN2 disease assessed by MRI cortical thickness histograms. . Mol Genet Metab. 2018; 123:S41. [Google Scholar]
  • 51.Nickel M, Simonati A, Jacoby D, Lezius S, Kilian D, Van de Graaf B, Pagovich OE, Kosofsky B, Yohay K, Downs M, Slasor P, Ajayi T, Crystal RG, Kohlschutter A, Sondhi D, Schulz A. Disease characteristics and progression in patients with late-infantile neuronal ceroid lipofuscinosis type 2 (CLN2) disease: an observational cohort study. Lancet Child Adolesc Health. 2018; 2:582–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ezaki J, Takeda-Ezaki M, Koike M, Ohsawa Y, Taka H, Mineki R, Murayama K, Uchiyama Y, Ueno T, Kominami E. Characterization of Cln3p, the gene product responsible for juvenile neuronal ceroid lipofuscinosis, as a lysosomal integral membrane glycoprotein. J Neurochem. 2003; 87:1296–1308. [DOI] [PubMed] [Google Scholar]
  • 53.Golabek AA, Kida E, Walus M, Kaczmarski W, Michalewski M, Wisniewski KE. CLN3 protein regulates lysosomal pH and alters intracellular processing of Alzheimer’s amyloid-beta protein precursor and cathepsin D in human cells. Mol Genet Metab. 2000; 70:203–213. [DOI] [PubMed] [Google Scholar]
  • 54.Holopainen JM, Saarikoski J, Kinnunen PK, Jarvela I. Elevated lysosomal pH in neuronal ceroid lipofuscinoses (NCLs). Eur J Biochem. 2001; 268:5851–5856. [DOI] [PubMed] [Google Scholar]
  • 55.Ramirez-Montealegre D, Pearce DA. Defective lysosomal arginine transport in juvenile Batten disease. Hum Mol Genet. 2005; 14:3759–3773. [DOI] [PubMed] [Google Scholar]
  • 56.Wu D, Liu J, Wu B, Tu B, Zhu W, Luo J. The Batten disease gene CLN3 confers resistance to endoplasmic reticulum stress induced by tunicamycin. Biochem Biophys Res Commun. 2014; 447:115–120. [DOI] [PubMed] [Google Scholar]
  • 57.Marshall FJ, de Blieck EA, Mink JW, Dure L, Adams H, Messing S, Rothberg PG, Levy E, McDonough T, DeYoung J, Wang M, Ramirez-Montealegre D, Kwon JM, Pearce DA. A clinical rating scale for Batten disease: reliable and relevant for clinical trials. Neurology. 2005; 65:275–279. [DOI] [PubMed] [Google Scholar]
  • 58.Perez-Poyato MS, Mila Recansens M, Ferrer Abizanda I, Montero Sanchez R, Rodriguez-Revenga L, Cusi Sanchez V, Garcia Gonzalez MM, Domingo Jimenez R, Camino Leon R, Velazquez Fragua R, Martinez-Bermejo A, Pineda Marfa M. Juvenile neuronal ceroid lipofuscinosis: clinical course and genetic studies in Spanish patients. J Inherit Metab Dis. 2011; 34:1083–1093. [DOI] [PubMed] [Google Scholar]
  • 59.Ostergaard JR. Juvenile neuronal ceroid lipofuscinosis (Batten disease): current insights. Degener Neurol Neuromuscul Dis. 2016; 6:73–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Berkovic SF, Carpenter S, Andermann F, Andermann E, Wolfe LS. Kufs’ disease: a critical reappraisal. Brain. 1988; 111 (Pt 1):27–62. [DOI] [PubMed] [Google Scholar]
  • 61.Nijssen PC, Ceuterick C, van Diggelen OP, Elleder M, Martin JJ, Teepen JL, Tyynela J, Roos RA. Autosomal dominant adult neuronal ceroid lipofuscinosis: a novel form of NCL with granular osmiophilic deposits without palmitoyl protein thioesterase 1 deficiency. Brain Pathol. 2003; 13:574–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Arsov T, Smith KR, Damiano J, Franceschetti S, Canafoglia L, Bromhead CJ, Andermann E, Vears DF, Cossette P, Rajagopalan S, McDougall A, Sofia V, Farrell M, Aguglia U, Zini A, Meletti S, Morbin M, Mullen S, Andermann F, Mole SE, Bahlo M, Berkovic SF. Kufs disease, the major adult form of neuronal ceroid lipofuscinosis, caused by mutations in CLN6. Am J Hum Genet. 2011; 88:566–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Henderson MX, Wirak GS, Zhang YQ, Dai F, Ginsberg SD, Dolzhanskaya N, Staropoli JF, Nijssen PC, Lam TT, Roth AF, Davis NG, Dawson G, Velinov M, Chandra SS. Neuronal ceroid lipofuscinosis with DNAJC5/CSPalpha mutation has PPT1 pathology and exhibit aberrant protein palmitoylation. Acta Neuropathol. 2016; 131:621–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Santavuori P, Rapola J, Sainio K, Raitta C. A variant of Jansky-Bielschowsky disease. Neuropediatrics. 1982; 13:135–141. [DOI] [PubMed] [Google Scholar]
  • 65.Simonati A, Williams RE, Nardocci N, Laine M, Battini R, Schulz A, Garavaglia B, Moro F, Pezzini F, Santorelli FM. Phenotype and natural history of variant late infantile ceroid-lipofuscinosis 5. Dev Med Child Neurol. 2017; 59:815–821. [DOI] [PubMed] [Google Scholar]
  • 66.Sharp JD, Wheeler RB, Parker KA, Gardiner RM, Williams RE, Mole SE. Spectrum of CLN6 mutations in variant late infantile neuronal ceroid lipofuscinosis. Hum Mutat. 2003; 22:35–42. [DOI] [PubMed] [Google Scholar]
  • 67.Heine C, Koch B, Storch S, Kohlschutter A, Palmer DN, Braulke T. Defective endoplasmic reticulum-resident membrane protein CLN6 affects lysosomal degradation of endocytosed arylsulfatase A. J Biol Chem. 2004; 279:22347–22352. [DOI] [PubMed] [Google Scholar]
  • 68.Gao H, Boustany RM, Espinola JA, Cotman SL, Srinidhi L, Antonellis KA, Gillis T, Qin X, Liu S, Donahue LR, Bronson RT, Faust JR, Stout D, Haines JL, Lerner TJ, MacDonald ME. Mutations in a novel CLN6-encoded transmembrane protein cause variant neuronal ceroid lipofuscinosis in man and mouse. Am J Hum Genet. 2002; 70:324–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kousi M, Siintola E, Dvorakova L, Vlaskova H, Turnbull J, Topcu M, Yuksel D, Gokben S, Minassian BA, Elleder M, Mole SE, Lehesjoki AE. Mutations in CLN7/MFSD8 are a common cause of variant late-infantile neuronal ceroid lipofuscinosis. Brain. 2009; 132:810–819. [DOI] [PubMed] [Google Scholar]
  • 70.Topcu M, Tan H, Yalnizoglu D, Usubutun A, Saatci I, Aynaci M, Anlar B, Topaloglu H, Turanli G, Kose G, Aysun S. Evaluation of 36 patients from Turkey with neuronal ceroid lipofuscinosis: clinical, neurophysiological, neuroradiological and histopathologic studies. Turk J Pediatr. 2004; 46:1–10. [PubMed] [Google Scholar]
  • 71.Luzio JP. CLN8 safeguards lysosome biogenesis. Nat Cell Biol. 2018; 20:1333–1335. [DOI] [PubMed] [Google Scholar]
  • 72.di Ronza A, Bajaj L, Sharma J, Sanagasetti D, Lotfi P, Adamski CJ, Collette J, Palmieri M, Amawi A, Popp L, Chang KT, Meschini MC, Leung HE, Segatori L, Simonati A, Sifers RN, Santorelli FM, Sardiello M. CLN8 is an endoplasmic reticulum cargo receptor that regulates lysosome biogenesis. Nat Cell Biol. 2018; 20:1370–1377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mitchell WA, Wheeler RB, Sharp JD, Bate SL, Gardiner RM, Ranta US, Lonka L, Williams RE, Lehesjoki AE, Mole SE. Turkish variant late infantile neuronal ceroid lipofuscinosis (CLN7) may be allelic to CLN8. Eur J Paediatr Neurol. 2001; 5 Suppl A:21–27. [DOI] [PubMed] [Google Scholar]
  • 74.Ranta S, Lehesjoki AE. Northern epilepsy, a new member of the NCL family. Neurol Sci. 2000; 21:S43–47. [DOI] [PubMed] [Google Scholar]
  • 75.Schulz A, Dhar S, Rylova S, Dbaibo G, Alroy J, Hagel C, Artacho I, Kohlschutter A, Lin S, Boustany RM. Impaired cell adhesion and apoptosis in a novel CLN9 Batten disease variant. Ann Neurol. 2004; 56:342–350. [DOI] [PubMed] [Google Scholar]
  • 76.Schulz A, Mousallem T, Venkataramani M, Persaud-Sawin DA, Zucker A, Luberto C, Bielawska A, Bielawski J, Holthuis JC, Jazwinski SM, Kozhaya L, Dbaibo GS, Boustany RM. The CLN9 protein, a regulator of dihydroceramide synthase. J Biol Chem. 2006; 281:2784–2794. [DOI] [PubMed] [Google Scholar]
  • 77.Norman RM, Wood N. A Congenital Form of Amaurotic Family Idiocy. J Neurol Psychiatry. 1941; 4:175–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Brown NJ, Corner BD, Dodgson MC. A second case in the same family of congenital familial cerebral lipoidosis resembling amaurotic family idiocy. Arch Dis Child. 1954; 29:48–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Humphreys S, Lake BD, Scholtz CL. Congenital amaurotic idiocy--a pathological, histochemical, biochemical and ultrastructural study. Neuropathol Appl Neurobiol. 1985; 11:475–484. [DOI] [PubMed] [Google Scholar]
  • 80.Barohn RJ, Dowd DC, Kagan-Hallet KS. Congenital ceroid-lipofuscinosis. Pediatr Neurol. 1992; 8:54–59. [DOI] [PubMed] [Google Scholar]
  • 81.Siintola E, Partanen S, Stromme P, Haapanen A, Haltia M, Maehlen J, Lehesjoki AE, Tyynela J. Cathepsin D deficiency underlies congenital human neuronal ceroid-lipofuscinosis. Brain. 2006; 129:1438–1445. [DOI] [PubMed] [Google Scholar]
  • 82.Steinfeld R, Reinhardt K, Schreiber K, Hillebrand M, Kraetzner R, Bruck W, Saftig P, Gartner J. Cathepsin D deficiency is associated with a human neurodegenerative disorder. Am J Hum Genet. 2006; 78:988–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hersheson J, Burke D, Clayton R, Anderson G, Jacques TS, Mills P, Wood NW, Gissen P, Clayton P, Fearnley J, Mole SE, Houlden H. Cathepsin D deficiency causes juvenile-onset ataxia and distinctive muscle pathology. Neurology. 2014; 83:1873–1875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Smith KR, Damiano J, Franceschetti S, Carpenter S, Canafoglia L, Morbin M, Rossi G, Pareyson D, Mole SE, Staropoli JF, Sims KB, Lewis J, Lin WL, Dickson DW, Dahl HH, Bahlo M, Berkovic SF. Strikingly different clinicopathological phenotypes determined by progranulin-mutation dosage. Am J Hum Genet. 2012; 90:1102–1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Bras J, Verloes A, Schneider SA, Mole SE, Guerreiro RJ. Mutation of the parkinsonism gene ATP13A2 causes neuronal ceroid-lipofuscinosis. Hum Mol Genet. 2012; 21:2646–2650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Tsunemi T, Hamada K, Krainc D. ATP13A2/PARK9 regulates secretion of exosomes and alpha-synuclein. J Neurosci. 2014; 34:15281–15287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Farias FH, Zeng R, Johnson GS, Wininger FA, Taylor JF, Schnabel RD, McKay SD, Sanders DN, Lohi H, Seppala EH, Wade CM, Lindblad-Toh K, O’Brien DP, Katz ML. A truncating mutation in ATP13A2 is responsible for adult-onset neuronal ceroid lipofuscinosis in Tibetan terriers. Neurobiol Dis. 2011; 42:468–474. [DOI] [PubMed] [Google Scholar]
  • 88.Wohlke A, Philipp U, Bock P, Beineke A, Lichtner P, Meitinger T, Distl O. A one base pair deletion in the canine ATP13A2 gene causes exon skipping and late-onset neuronal ceroid lipofuscinosis in the Tibetan terrier. PLoS Genet. 2011; 7:e1002304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Smith KR, Dahl HH, Canafoglia L, Andermann E, Damiano J, Morbin M, Bruni AC, Giaccone G, Cossette P, Saftig P, Grotzinger J, Schwake M, Andermann F, Staropoli JF, Sims KB, Mole SE, Franceschetti S, Alexander NA, Cooper JD, Chapman HA, Carpenter S, Berkovic SF, Bahlo M. Cathepsin F mutations cause Type B Kufs disease, an adult-onset neuronal ceroid lipofuscinosis. Hum Mol Genet. 2013; 22:1417–1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wang C, Xu H, Yuan Y, Lian Y, Xie N, Ming L. Novel compound heterozygous mutations causing Kufs disease type B. Int J Neurosci. 2018; 128:573–576. [DOI] [PubMed] [Google Scholar]
  • 91.Van Bogaert P, Azizieh R, Desir J, Aeby A, De Meirleir L, Laes JF, Christiaens F, Abramowicz MJ. Mutation of a potassium channel-related gene in progressive myoclonic epilepsy. Ann Neurol. 2007; 61:579–586. [DOI] [PubMed] [Google Scholar]
  • 92.Moen MN, Fjaer R, Hamdani EH, Laerdahl JK, Menchini RJ, Vigeland MD, Sheng Y, Undlien DE, Hassel B, Salih MA, El Khashab HY, Selmer KK, Chaudhry FA. Pathogenic variants in KCTD7 perturb neuronal K+ fluxes and glutamine transport. Brain. 2016; 139:3109–3120. [DOI] [PubMed] [Google Scholar]
  • 93.Metz KA, Teng X, Coppens I, Lamb HM, Wagner BE, Rosenfeld JA, Chen X, Zhang Y, Kim HJ, Meadow ME, Wang TS, Haberlandt ED, Anderson GW, Leshinsky-Silver E, Bi W, Markello TC, Pratt M, Makhseed N, Garnica A, Danylchuk NR, Burrow TA, Jayakar P, McKnight D, Agadi S, Gbedawo H, Stanley C, Alber M, Prehl I, Peariso K, Ong MT, Mordekar SR, Parker MJ, Crooks D, Agrawal PB, Berry GT, Loddenkemper T, Yang Y, Maegawa GHB, Aouacheria A, Markle JG, Wohlschlegel JA, Hartman AL, Hardwick JM. KCTD7 deficiency defines a distinct neurodegenerative disorder with a conserved autophagy-lysosome defect. Ann Neurol. 2018; 84:766–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Staropoli JF, Karaa A, Lim ET, Kirby A, Elbalalesy N, Romansky SG, Leydiker KB, Coppel SH, Barone R, Xin W, MacDonald ME, Abdenur JE, Daly MJ, Sims KB, Cotman SL. A homozygous mutation in KCTD7 links neuronal ceroid lipofuscinosis to the ubiquitin-proteasome system. Am J Hum Genet. 2012; 91:202–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Cotman SL, Vrbanac V, Lebel LA, Lee RL, Johnson KA, Donahue LR, Teed AM, Antonellis K, Bronson RT, Lerner TJ, MacDonald ME. Cln3(Deltaex7/8) knock-in mice with the common JNCL mutation exhibit progressive neurologic disease that begins before birth. Hum Mol Genet. 2002; 11:2709–2721. [DOI] [PubMed] [Google Scholar]
  • 96.Sleat DE, Wiseman JA, El-Banna M, Kim KH, Mao Q, Price S, Macauley SL, Sidman RL, Shen MM, Zhao Q, Passini MA, Davidson BL, Stewart GR, Lobel P. A mouse model of classical late-infantile neuronal ceroid lipofuscinosis based on targeted disruption of the CLN2 gene results in a loss of tripeptidyl-peptidase I activity and progressive neurodegeneration. J Neurosci. 2004; 24:9117–9126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Shacka JJ. Mouse models of neuronal ceroid lipofuscinoses: useful pre-clinical tools to delineate disease pathophysiology and validate therapeutics. Brain Res Bull. 2012; 88:43–57. [DOI] [PubMed] [Google Scholar]
  • 98.Bond M, Holthaus SM, Tammen I, Tear G, Russell C. Use of model organisms for the study of neuronal ceroid lipofuscinosis. Biochim Biophys Acta. 2013; 1832:1842–1865. [DOI] [PubMed] [Google Scholar]
  • 99.Vaquer G, Riviere F, Mavris M, Bignami F, Llinares-Garcia J, Westermark K, Sepodes B. Animal models for metabolic, neuromuscular and ophthalmological rare diseases. Nat Rev Drug Discov. 2013; 12:287–305. [DOI] [PubMed] [Google Scholar]
  • 100.Staropoli JF, Haliw L, Biswas S, Garrett L, Holter SM, Becker L, Skosyrski S, Da Silva-Buttkus P, Calzada-Wack J, Neff F, Rathkolb B, Rozman J, Schrewe A, Adler T, Puk O, Sun M, Favor J, Racz I, Bekeredjian R, Busch DH, Graw J, Klingenspor M, Klopstock T, Wolf E, Wurst W, Zimmer A, Lopez E, Harati H, Hill E, Krause DS, Guide J, Dragileva E, Gale E, Wheeler VC, Boustany RM, Brown DE, Breton S, Ruether K, Gailus-Durner V, Fuchs H, de Angelis MH, Cotman SL. Large-scale phenotyping of an accurate genetic mouse model of JNCL identifies novel early pathology outside the central nervous system. PLoS One. 2012; 7:e38310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Katz ML, Johnson GC, Leach SB, Williamson BG, Coates JR, Whiting REH, Vansteenkiste DP, Whitney MS. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Jalanko A, Vesa J, Manninen T, von Schantz C, Minye H, Fabritius AL, Salonen T, Rapola J, Gentile M, Kopra O, Peltonen L. Mice with Ppt1Deltaex4 mutation replicate the INCL phenotype and show an inflammation-associated loss of interneurons. Neurobiol Dis. 2005; 18:226–241. [DOI] [PubMed] [Google Scholar]
  • 103.Miller JN, Kovacs AD, Pearce DA. The novel Cln1(R151X) mouse model of infantile neuronal ceroid lipofuscinosis (INCL) for testing nonsense suppression therapy. Hum Mol Genet. 2015; 24:185–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Brooks DA, Muller VJ, Hopwood JJ. Stop-codon read-through for patients affected by a lysosomal storage disorder. Trends Mol Med. 2006; 12:367–373. [DOI] [PubMed] [Google Scholar]
  • 105.Sanders DN, Farias FH, Johnson GS, Chiang V, Cook JR, O’Brien DP, Hofmann SL, Lu JY, Katz ML. A mutation in canine PPT1 causes early onset neuronal ceroid lipofuscinosis in a Dachshund. Mol Genet Metab. 2010; 100:349–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kolicheski A, Barnes Heller HL, Arnold S, Schnabel RD, Taylor JF, Knox CA, Mhlanga-Mutangadura T, O’Brien DP, Johnson GS, Dreyfus J, Katz ML. Homozygous PPT1 Splice Donor Mutation in a Cane Corso Dog With Neuronal Ceroid Lipofuscinosis. J Vet Intern Med. 2017; 31:149–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sondhi D, Hackett NR, Peterson DA, Stratton J, Baad M, Travis KM, Wilson JM, Crystal RG. Enhanced survival of the LINCL mouse following CLN2 gene transfer using the rh.10 rhesus macaque-derived adeno-associated virus vector. Mol Ther. 2007; 15:481–491. [DOI] [PubMed] [Google Scholar]
  • 108.Chang M, Cooper JD, Sleat DE, Cheng SH, Dodge JC, Passini MA, Lobel P, Davidson BL. Intraventricular enzyme replacement improves disease phenotypes in a mouse model of late infantile neuronal ceroid lipofuscinosis. Mol Ther. 2008; 16:649–656. [DOI] [PubMed] [Google Scholar]
  • 109.Xu S, Wang L, El-Banna M, Sohar I, Sleat DE, Lobel P. Large-volume intrathecal enzyme delivery increases survival of a mouse model of late infantile neuronal ceroid lipofuscinosis. Mol Ther. 2011; 19:1842–1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Geraets RD, Langin LM, Cain JT, Parker CM, Beraldi R, Kovacs AD, Weimer JM, Pearce DA. A tailored mouse model of CLN2 disease: A nonsense mutant for testing personalized therapies. PLoS One. 2017; 12:e0176526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Nemtsova Y, Wiseman JA, El-Banna M, Lobel P, Sleat DE. Inducible transgenic expression of tripeptidyl peptidase 1 in a mouse model of late-infantile neuronal ceroid lipofuscinosis. PLoS One. 2018; 13:e0192286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kovacs AD, Pearce DA. Finding the most appropriate mouse model of juvenile CLN3 (Batten) disease for therapeutic studies: the importance of genetic background and gender. Dis Model Mech. 2015; 8:351–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Sondhi D, Scott EC, Chen A, Hackett NR, Wong AM, Kubiak A, Nelvagal HR, Pearse Y, Cotman SL, Cooper JD, Crystal RG. Partial correction of the CNS lysosomal storage defect in a mouse model of juvenile neuronal ceroid lipofuscinosis by neonatal CNS administration of an adeno-associated virus serotype rh.10 vector expressing the human CLN3 gene. Hum Gene Ther. 2014; 25:223–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Bosch ME, Aldrich A, Fallet R, Odvody J, Burkovetskaya M, Schuberth K, Fitzgerald JA, Foust KD, Kielian T. Self-Complementary AAV9 Gene Delivery Partially Corrects Pathology Associated with Juvenile Neuronal Ceroid Lipofuscinosis (CLN3). J Neurosci. 2016; 36:9669–9682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Wiley LA, Burnight ER, Drack AV, Banach BB, Ochoa D, Cranston CM, Madumba RA, East JS, Mullins RF, Stone EM, Tucker BA. Using Patient-Specific Induced Pluripotent Stem Cells and Wild-Type Mice to Develop a Gene Augmentation-Based Strategy to Treat CLN3-Associated Retinal Degeneration. Hum Gene Ther. 2016; 27:835–846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Groh J, Berve K, Martini R. Fingolimod and Teriflunomide Attenuate Neurodegeneration in Mouse Models of Neuronal Ceroid Lipofuscinosis. Mol Ther. 2017; 25:1889–1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kerns S, Arnold M, Aldrich A, Fallet R, Kielian T, Padegimas L, Miller TJ. An improved, novel, systemically administered AAV gene therapy for treatment of CLN3 juvenile neuronal ceroid lipofuscinosis. Molecular Genetics and Metabolism. 2019; 126:S82–S83. [Google Scholar]
  • 118.Kerns S, Daum J, Bosch ME, Fallet R, Odvody J, Henson K, Spratt K, Kielian T. Intravenous administration of CLN3 gene therapy for juvenile neuronal ceroid lipofuscinosis. Mol Genet Metab. 2018; 123:S15–S153. [Google Scholar]
  • 119.Frugier T, Mitchell NL, Tammen I, Houweling PJ, Arthur DG, Kay GW, van Diggelen OP, Jolly RD, Palmer DN. A new large animal model of CLN5 neuronal ceroid lipofuscinosis in Borderdale sheep is caused by a nucleotide substitution at a consensus splice site (c.571+1G>A) leading to excision of exon 3. Neurobiol Dis. 2008; 29:306–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Jolly RD, Arthur DG, Kay GW, Palmer DN. Neuronal ceroid-lipofuscinosis in Borderdale sheep. N Z Vet J. 2002; 50:199–202. [DOI] [PubMed] [Google Scholar]
  • 121.Best HL, Neverman NJ, Wicky HE, Mitchell NL, Leitch B, Hughes SM. Characterisation of early changes in ovine CLN5 and CLN6 Batten disease neural cultures for the rapid screening of therapeutics. Neurobiol Dis. 2017; 100:62–74. [DOI] [PubMed] [Google Scholar]
  • 122.Mitchell NL, Russell KN, Wellby MP, Wicky HE, Schoderboeck L, Barrell GK, Melzer TR, Gray SJ, Hughes SM, Palmer DN. Longitudinal In Vivo Monitoring of the CNS Demonstrates the Efficacy of Gene Therapy in a Sheep Model of CLN5 Batten Disease. Mol Ther 2018; 26:2366–2378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Melville SA, Wilson CL, Chiang CS, Studdert VP, Lingaas F, Wilton AN. A mutation in canine CLN5 causes neuronal ceroid lipofuscinosis in Border collie dogs. Genomics. 2005; 86:287–294. [DOI] [PubMed] [Google Scholar]
  • 124.Gilliam D, Kolicheski A, Johnson GS, Mhlanga-Mutangadura T, Taylor JF, Schnabel RD, Katz ML. Golden Retriever dogs with neuronal ceroid lipofuscinosis have a two-base-pair deletion and frameshift in CLN5. Mol Genet Metab. 2015; 115:101–109. [DOI] [PubMed] [Google Scholar]
  • 125.Kleine Holthaus SM, Ribeiro J, Abelleira-Hervas L, Pearson RA, Duran Y, Georgiadis A, Sampson RD, Rizzi M, Hoke J, Maswood R, Azam S, Luhmann UFO, Smith AJ, Mole SE, Ali RR. Prevention of Photoreceptor Cell Loss in a Cln6(nclf) Mouse Model of Batten Disease Requires CLN6 Gene Transfer to Bipolar Cells. Mol Ther. 2018; 26:1343–1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Katz ML, Farias FH, Sanders DN, Zeng R, Khan S, Johnson GS, O’Brien DP. A missense mutation in canine CLN6 in an Australian shepherd with neuronal ceroid lipofuscinosis. J Biomed Biotechnol. 2011; 2011:198042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Tammen I, Cook RW, Nicholas FW, Raadsma HW. Neuronal ceroid lipofuscinosis in Australian Merino sheep: a new animal model. Eur J Paediatr Neurol. 2001; 5 Suppl A:37–41. [DOI] [PubMed] [Google Scholar]
  • 128.Damme M, Brandenstein L, Fehr S, Jankowiak W, Bartsch U, Schweizer M, Hermans-Borgmeyer I, Storch S. Gene disruption of Mfsd8 in mice provides the first animal model for CLN7 disease. Neurobiol Dis. 2014; 65:12–24. [DOI] [PubMed] [Google Scholar]
  • 129.Jankowiak W, Brandenstein L, Dulz S, Hagel C, Storch S, Bartsch U. Retinal Degeneration in Mice Deficient in the Lysosomal Membrane Protein CLN7. Invest Ophthalmol Vis Sci. 2016; 57:4989–4998. [DOI] [PubMed] [Google Scholar]
  • 130.Brandenstein L, Schweizer M, Sedlacik J, Fiehler J, Storch S. Lysosomal dysfunction and impaired autophagy in a novel mouse model deficient for the lysosomal membrane protein Cln7. Hum Mol Genet. 2016; 25:777–791. [DOI] [PubMed] [Google Scholar]
  • 131.Faller KM, Bras J, Sharpe SJ, Anderson GW, Darwent L, Kun-Rodrigues C, Alroy J, Penderis J, Mole SE, Gutierrez-Quintana R, Guerreiro RJ. The Chihuahua dog: A new animal model for neuronal ceroid lipofuscinosis CLN7 disease? J Neurosci Res. 2016; 94:339–347. [DOI] [PubMed] [Google Scholar]
  • 132.McBride JL, Neuringer M, Ferguson B, Kohama SG, Tagge IJ, Zweig RC, Renner LM, McGill TJ, Stoddard J, Peterson S, Su W, Sherman LS, Domire JS, Ducore RM, Colgin LM, Lewis AD. Discovery of a CLN7 model of Batten disease in non-human primates. Neurobiol Dis. 2018; 119:65–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Katz ML, Khan S, Awano T, Shahid SA, Siakotos AN, Johnson GS. A mutation in the CLN8 gene in English Setter dogs with neuronal ceroid-lipofuscinosis. Biochem Biophys Res Commun. 2005; 327:541–547. [DOI] [PubMed] [Google Scholar]
  • 134.Hirz M, Drogemuller M, Schanzer A, Jagannathan V, Dietschi E, Goebel HH, Hecht W, Laubner S, Schmidt MJ, Steffen F, Hilbe M, Kohler K, Drogemuller C, Herden C. Neuronal ceroid lipofuscinosis (NCL) is caused by the entire deletion of CLN8 in the Alpenlandische Dachsbracke dog. Mol Genet Metab. 2017; 120:269–277. [DOI] [PubMed] [Google Scholar]
  • 135.Lingaas F, Guttersrud OA, Arnet E, Espenes A. Neuronal ceroid lipofuscinosis in Salukis is caused by a single base pair insertion in CLN8. Anim Genet. 2018; 49:52–58. [DOI] [PubMed] [Google Scholar]
  • 136.Saftig P, Hetman M, Schmahl W, Weber K, Heine L, Mossmann H, Koster A, Hess B, Evers M, von Figura K, et al. Mice deficient for the lysosomal proteinase cathepsin D exhibit progressive atrophy of the intestinal mucosa and profound destruction of lymphoid cells. EMBO J. 1995; 14:3599–3608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Tyynela J, Sohar I, Sleat DE, Gin RM, Donnelly RJ, Baumann M, Haltia M, Lobel P. A mutation in the ovine cathepsin D gene causes a congenital lysosomal storage disease with profound neurodegeneration. EMBO J. 2000; 19:2786–2792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Awano T, Katz ML, O’Brien DP, Taylor JF, Evans J, Khan S, Sohar I, Lobel P, Johnson GS. A mutation in the cathepsin D gene (CTSD) in American Bulldogs with neuronal ceroid lipofuscinosis. Mol Genet Metab. 2006; 87:341–348. [DOI] [PubMed] [Google Scholar]
  • 139.Kayasuga Y, Chiba S, Suzuki M, Kikusui T, Matsuwaki T, Yamanouchi K, Kotaki H, Horai R, Iwakura Y, Nishihara M. Alteration of behavioural phenotype in mice by targeted disruption of the progranulin gene. Behav Brain Res. 2007; 185:110–118. [DOI] [PubMed] [Google Scholar]
  • 140.Yin F, Banerjee R, Thomas B, Zhou P, Qian L, Jia T, Ma X, Ma Y, Iadecola C, Beal MF, Nathan C, Ding A. Exaggerated inflammation, impaired host defense, and neuropathology in progranulin-deficient mice. J Exp Med. 2010; 207:117–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Yin F, Dumont M, Banerjee R, Ma Y, Li H, Lin MT, Beal MF, Nathan C, Thomas B, Ding A. Behavioral deficits and progressive neuropathology in progranulin-deficient mice: a mouse model of frontotemporal dementia. FASEB J. 2010; 24:4639–4647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Schultheis PJ, Fleming SM, Clippinger AK, Lewis J, Tsunemi T, Giasson B, Dickson DW, Mazzulli JR, Bardgett ME, Haik KL, Ekhator O, Chava AK, Howard J, Gannon M, Hoffman E, Chen Y, Prasad V, Linn SC, Tamargo RJ, Westbroek W, Sidransky E, Krainc D, Shull GE. Atp13a2-deficient mice exhibit neuronal ceroid lipofuscinosis, limited alpha-synuclein accumulation and age-dependent sensorimotor deficits. Hum Mol Genet. 2013; 22:2067–2082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Hersrud SL, Geraets RD, Weber KL, Chan CH, Pearce DA. Plasma biomarkers for neuronal ceroid lipofuscinosis. FEBS J. 2016; 283:459–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Sleat DE, Tannous A, Sohar I, Wiseman JA, Zheng H, Qian M, Zhao C, Xin W, Barone R, Sims KB, Moore DF, Lobel P. Proteomic Analysis of Brain and Cerebrospinal Fluid from the Three Major Forms of Neuronal Ceroid Lipofuscinosis Reveals Potential Biomarkers. J Proteome Res. 2017; 16:3787–3804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Stamberger H, Nikanorova M, Willemsen MH, Accorsi P, Angriman M, Baier H, Benkel-Herrenbrueck I, Benoit V, Budetta M, Caliebe A, Cantalupo G, Capovilla G, Casara G, Courage C, Deprez M, Destree A, Dilena R, Erasmus CE, Fannemel M, Fjaer R, Giordano L, Helbig KL, Heyne HO, Klepper J, Kluger GJ, Lederer D, Lodi M, Maier O, Merkenschlager A, Michelberger N, Minetti C, Muhle H, Phalin J, Ramsey K, Romeo A, Schallner J, Schanze I, Shinawi M, Sleegers K, Sterbova K, Syrbe S, Traverso M, Tzschach A, Uldall P, Van Coster R, Verhelst H, Viri M, Winter S, Wolff M, Zenker M, Zoccante L, De Jonghe P, Helbig I, Striano P, Lemke JR, Moller RS, Weckhuysen S. STXBP1 encephalopathy: A neurodevelopmental disorder including epilepsy. Neurology. 2016; 86:954–962. [DOI] [PubMed] [Google Scholar]
  • 146.Sindelar M, Dyke JP, Deeb RS, Sondhi D, Kaminsky SM, Kosofsky BE, Ballon DJ, Crystal RG, Gross SS. Untargeted Metabolite Profiling of Cerebrospinal Fluid Uncovers Biomarkers for Severity of Late Infantile Neuronal Ceroid Lipofuscinosis (CLN2, Batten Disease). Sci Rep. 2018; 8:15229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Lebrun AH, Moll-Khosrawi P, Pohl S, Makrypidi G, Storch S, Kilian D, Streichert T, Otto B, Mole SE, Ullrich K, Cotman S, Kohlschutter A, Braulke T, Schulz A. Analysis of potential biomarkers and modifier genes affecting the clinical course of CLN3 disease. Mol Med. 2011; 17:1253–1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Timm D, Cain JT, Geraets RD, White KA, Koh SY, Kielian T, Pearce DA, Hastings ML, Weimer JM. Searching for novel biomarkers using a mouse model of CLN3-Batten disease. PLoS One. 2018; 13:e0201470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Abbott NJ. Blood-brain barrier structure and function and the challenges for CNS drug delivery. J Inherit Metab Dis. 2013; 36:437–449. [DOI] [PubMed] [Google Scholar]
  • 150.Naso MF, Tomkowicz B, Perry WL 3rd, Strohl WR. Adeno-Associated Virus (AAV) as a Vector for Gene Therapy. BioDrugs. 2017; 31:317–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Ginn SL, Amaya AK, Alexander IE, Edelstein M, Abedi MR. Gene therapy clinical trials worldwide to 2017: An update. J Gene Med. 2018; 20:e3015. [DOI] [PubMed] [Google Scholar]
  • 152.Colella P, Ronzitti G, Mingozzi F. Emerging Issues in AAV-Mediated In Vivo Gene Therapy. Mol Ther Methods Clin Dev. 2018; 8:87–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Desnick RJ, Schuchman EH. Enzyme replacement therapy for lysosomal diseases: lessons from 20 years of experience and remaining challenges. Annu Rev Genomics Hum Genet. 2012; 13:307–335. [DOI] [PubMed] [Google Scholar]
  • 154.Giugliani R, Vairo F, Kubaski F, Poswar F, Riegel M, Baldo G, Saute JA. Neurological manifestations of lysosomal disorders and emerging therapies targeting the CNS. Lancet Child Adolesc Health. 2018; 2:56–68. [DOI] [PubMed] [Google Scholar]
  • 155.Solomon M, Muro S. Lysosomal enzyme replacement therapies: Historical development, clinical outcomes, and future perspectives. Adv Drug Deliv Rev. 2017; 118:109–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Pardridge WM. CSF, blood-brain barrier, and brain drug delivery. Expert Opin Drug Deliv. 2016; 13:963–975. [DOI] [PubMed] [Google Scholar]
  • 157.Kohan R, Cismondi IA, Oller-Ramirez AM, Guelbert N, Anzolini TV, Alonso G, Mole SE, de Kremer DR, de Halac NI. Therapeutic approaches to the challenge of neuronal ceroid lipofuscinoses. Curr Pharm Biotechnol. 2011; 12:867–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Kollmann K, Uusi-Rauva K, Scifo E, Tyynela J, Jalanko A, Braulke T. Cell biology and function of neuronal ceroid lipofuscinosis-related proteins. Biochim Biophys Acta. 2013; 1832:1866–1881. [DOI] [PubMed] [Google Scholar]
  • 159.US Food and Drug Administration. FDA approves first treatment for a form of Batten disease https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-form-batten-disease [last accessed 6/18/19]
  • 160.Miller WP, Rothman SM, Nascene D, Kivisto T, DeFor TE, Ziegler RS, Eisengart J, Leiser K, Raymond G, Lund TC, Tolar J, Orchard PJ. Outcomes after allogeneic hematopoietic cell transplantation for childhood cerebral adrenoleukodystrophy: the largest single-institution cohort report. Blood. 2011; 118:1971–1978. [DOI] [PubMed] [Google Scholar]
  • 161.Kruer MC, Pearce DA, Orchard PJ, Steiner RD. Prospects for stem cell therapy in neuronal ceroid lipofuscinosis. Regen Med. 2013; 8:527–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Biffi A, Montini E, Lorioli L, Cesani M, Fumagalli F, Plati T, Baldoli C, Martino S, Calabria A, Canale S, Benedicenti F, Vallanti G, Biasco L, Leo S, Kabbara N, Zanetti G, Rizzo WB, Mehta NA, Cicalese MP, Casiraghi M, Boelens JJ, Del Carro U, Dow DJ, Schmidt M, Assanelli A, Neduva V, Di Serio C, Stupka E, Gardner J, von Kalle C, Bordignon C, Ciceri F, Rovelli A, Roncarolo MG, Aiuti A, Sessa M, Naldini L. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science. 2013; 341:1233158. [DOI] [PubMed] [Google Scholar]
  • 163.Milward K, Hester J, Wood KJ. Isolation of Human Regulatory T Lymphocytes by Fluorescence-Activated Cell Sorting. Methods Mol Biol. 2019; 1899:43–54. [DOI] [PubMed] [Google Scholar]
  • 164.Bayever E, Ladisch S, Philippart M, Brill N, Nuwer M, Sparkes RS, Feig SA. Bone-marrow transplantation for metachromatic leucodystrophy. Lancet. 1985; 2:471–473. [DOI] [PubMed] [Google Scholar]
  • 165.Krivit W, Aubourg P, Shapiro E, Peters C. Bone marrow transplantation for globoid cell leukodystrophy, adrenoleukodystrophy, metachromatic leukodystrophy, and Hurler syndrome. Curr Opin Hematol. 1999; 6:377–382. [DOI] [PubMed] [Google Scholar]
  • 166.Asheuer M, Pflumio F, Benhamida S, Dubart-Kupperschmitt A, Fouquet F, Imai Y, Aubourg P, Cartier N. Human CD34+ cells differentiate into microglia and express recombinant therapeutic protein. Proc Natl Acad Sci U S A. 2004; 101:3557–3562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Biffi A, Capotondo A, Fasano S, del Carro U, Marchesini S, Azuma H, Malaguti MC, Amadio S, Brambilla R, Grompe M, Bordignon C, Quattrini A, Naldini L. Gene therapy of metachromatic leukodystrophy reverses neurological damage and deficits in mice. J Clin Invest. 2006; 116:3070–3082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Lindvall O, Kokaia Z. Stem cells for the treatment of neurological disorders. Nature. 2006; 441:1094–1096. [DOI] [PubMed] [Google Scholar]
  • 169.Aboody K, Capela A, Niazi N, Stern JH, Temple S. Translating stem cell studies to the clinic for CNS repair: current state of the art and the need for a Rosetta stone. Neuron. 2011; 70:597–613. [DOI] [PubMed] [Google Scholar]
  • 170.Jin HK, Carter JE, Huntley GW, Schuchman EH. Intracerebral transplantation of mesenchymal stem cells into acid sphingomyelinase-deficient mice delays the onset of neurological abnormalities and extends their life span. J Clin Invest. 2002; 109:1183–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Chaudhuri A, Bhattacharya N. Human Neural Stem Cell Transplants in Neurological Disorders: Current Trends and Future Options In: Bhattacharya N, Stubblefield P, editors. Human Fetal Tissue Transplantation. London: Springer London; 2013. p. 265–268. [Google Scholar]
  • 172.Sevin C, Aubourg P, Cartier N. Enzyme, cell and gene-based therapies for metachromatic leukodystrophy. J Inherit Metab Dis. 2007; 30:175–183. [DOI] [PubMed] [Google Scholar]
  • 173.Bredius RG, Laan LA, Lankester AC, Poorthuis BJ, van Tol MJ, Egeler RM, Arts WF. Early marrow transplantation in a pre-symptomatic neonate with late infantile metachromatic leukodystrophy does not halt disease progression. Bone Marrow Transplant. 2007; 39:309–310. [DOI] [PubMed] [Google Scholar]
  • 174.Willingham MC, Pastan IH, Sahagian GG, Jourdian GW, Neufeld EF. Morphologic study of the internalization of a lysosomal enzyme by the mannose 6-phosphate receptor in cultured Chinese hamster ovary cells. Proc Natl Acad Sci U S A. 1981; 78:6967–6971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Ghosh P, Dahms NM, Kornfeld S. Mannose 6-phosphate receptors: new twists in the tale. Nat Rev Mol Cell Biol. 2003; 4:202–212. [DOI] [PubMed] [Google Scholar]
  • 176.Beck M Therapy for lysosomal storage disorders. IUBMB Life. 2010; 62:33–40. [DOI] [PubMed] [Google Scholar]
  • 177.Deverman BE, Ravina BM, Bankiewicz KS, Paul SM, Sah DWY. Gene therapy for neurological disorders: progress and prospects. Nat Rev Drug Discov. 2018; 17:641–659. [DOI] [PubMed] [Google Scholar]
  • 178.Morgan RA, Gray D, Lomova A, Kohn DB. Hematopoietic Stem Cell Gene Therapy: Progress and Lessons Learned. Cell Stem Cell. 2017; 21:574–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Gowing G, Svendsen S, Svendsen CN. Ex vivo gene therapy for the treatment of neurological disorders. Prog Brain Res. 2017; 230:99–132. [DOI] [PubMed] [Google Scholar]
  • 180.Hudry E, Vandenberghe LH. Therapeutic AAV Gene Transfer to the Nervous System: A Clinical Reality. Neuron. 2019; 102:263. [DOI] [PubMed] [Google Scholar]
  • 181.Auricchio A, Kobinger G, Anand V, Hildinger M, O’Connor E, Maguire AM, Wilson JM, Bennett J. Exchange of surface proteins impacts on viral vector cellular specificity and transduction characteristics: the retina as a model. Hum Mol Genet. 2001; 10:3075–3081. [DOI] [PubMed] [Google Scholar]
  • 182.Aschauer DF, Kreuz S, Rumpel S. Analysis of transduction efficiency, tropism and axonal transport of AAV serotypes 1, 2, 5, 6, 8 and 9 in the mouse brain. PLoS One. 2013; 8:e76310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Pillay S, Carette JE. Host determinants of adeno-associated viral vector entry. Curr Opin Virol. 2017; 24:124–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Janson C, McPhee S, Bilaniuk L, Haselgrove J, Testaiuti M, Freese A, Wang DJ, Shera D, Hurh P, Rupin J, Saslow E, Goldfarb O, Goldberg M, Larijani G, Sharrar W, Liouterman L, Camp A, Kolodny E, Samulski J, Leone P. Clinical protocol. Gene therapy of Canavan disease: AAV-2 vector for neurosurgical delivery of aspartoacylase gene (ASPA) to the human brain. Hum Gene Ther. 2002; 13:1391–1412. [DOI] [PubMed] [Google Scholar]
  • 185.Herzog CD, Bishop KM, Brown L, Wilson A, Kordower JH, Bartus RT. Gene transfer provides a practical means for safe, long-term, targeted delivery of biologically active neurotrophic factor proteins for neurodegenerative diseases. Drug Deliv Transl Res. 2011; 1:361–382. [DOI] [PubMed] [Google Scholar]
  • 186.Leone P, Shera D, McPhee SW, Francis JS, Kolodny EH, Bilaniuk LT, Wang DJ, Assadi M, Goldfarb O, Goldman HW, Freese A, Young D, During MJ, Samulski RJ, Janson CG. Long-term follow-up after gene therapy for canavan disease. Sci Transl Med. 2012; 4:165ra163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Hocquemiller M, Giersch L, Audrain M, Parker S, Cartier N. Adeno-Associated Virus-Based Gene Therapy for CNS Diseases. Hum Gene Ther. 2016; 27:478–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Lau AA, Hemsley KM. Adeno-associated viral gene therapy for mucopolysaccharidoses exhibiting neurodegeneration. J Mol Med (Berl). 2017; 95:1043–1052. [DOI] [PubMed] [Google Scholar]
  • 189.Kaplitt MG, Feigin A, Tang C, Fitzsimons HL, Mattis P, Lawlor PA, Bland RJ, Young D, Strybing K, Eidelberg D, During MJ. Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson’s disease: an open label, phase I trial. Lancet. 2007; 369:2097–2105. [DOI] [PubMed] [Google Scholar]
  • 190.Worgall S, Sondhi D, Hackett NR, Kosofsky B, Kekatpure MV, Neyzi N, Dyke JP, Ballon D, Heier L, Greenwald BM, Christos P, Mazumdar M, Souweidane MM, Kaplitt MG, Crystal RG. Treatment of late infantile neuronal ceroid lipofuscinosis by CNS administration of a serotype 2 adeno-associated virus expressing CLN2 cDNA. Hum Gene Ther. 2008; 19:463–474. [DOI] [PubMed] [Google Scholar]
  • 191.Janson CG, Romanova LG, Leone P, Nan Z, Belur L, McIvor RS, Low WC. Comparison of Endovascular and Intraventricular Gene Therapy With Adeno-Associated Virus-alpha-L-Iduronidase for Hurler Disease. Neurosurgery. 2014; 74:99–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Rafii MS, Baumann TL, Bakay RA, Ostrove JM, Siffert J, Fleisher AS, Herzog CD, Barba D, Pay M, Salmon DP, Chu Y, Kordower JH, Bishop K, Keator D, Potkin S, Bartus RT. A phase1 study of stereotactic gene delivery of AAV2-NGF for Alzheimer’s disease. Alzheimers Dement. 2014; 10:571–581. [DOI] [PubMed] [Google Scholar]
  • 193.Aubourg P Gene therapy for leukodystrophy: progress, challenges and opportunities. Expert Opinion on Orphan Drugs. 2016; 4:359–367. [Google Scholar]
  • 194.Choudhury SR, Hudry E, Maguire CA, Sena-Esteves M, Breakefield XO, Grandi P. Viral vectors for therapy of neurologic diseases. Neuropharmacology. 2017; 120:63–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Joshi CR, Labhasetwar V, Ghorpade A. Destination Brain: the Past, Present, and Future of Therapeutic Gene Delivery. J Neuroimmune Pharmacol. 2017; 12:51–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Tardieu M, Zerah M, Gougeon ML, Ausseil J, de Bournonville S, Husson B, Zafeiriou D, Parenti G, Bourget P, Poirier B, Furlan V, Artaud C, Baugnon T, Roujeau T, Crystal RG, Meyer C, Deiva K, Heard JM. Intracerebral gene therapy in children with mucopolysaccharidosis type IIIB syndrome: an uncontrolled phase 1/2 clinical trial. Lancet Neurol. 2017; 16:712–720. [DOI] [PubMed] [Google Scholar]
  • 197.Zaiss AK, Muruve DA. Immunity to adeno-associated virus vectors in animals and humans: a continued challenge. Gene Ther. 2008; 15:808–816. [DOI] [PubMed] [Google Scholar]
  • 198.Thwaite R, Pages G, Chillon M, Bosch A. AAVrh.10 immunogenicity in mice and humans. Relevance of antibody cross-reactivity in human gene therapy. Gene Ther. 2015; 22:196–201. [DOI] [PubMed] [Google Scholar]
  • 199.Fitzpatrick Z, Leborgne C, Barbon E, Masat E, Ronzitti G, van Wittenberghe L, Vignaud A, Collaud F, Charles S, Simon Sola M, Jouen F, Boyer O, Mingozzi F. Influence of Pre-existing Anti-capsid Neutralizing and Binding Antibodies on AAV Vector Transduction. Mol Ther Methods Clin Dev. 2018; 9:119–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Hughes SM, Hope KM, Xu JB, Mitchell NL, Palmer DN. Inhibition of storage pathology in prenatal CLN5-deficient sheep neural cultures by lentiviral gene therapy. Neurobiol Dis. 2014; 62:543–550. [DOI] [PubMed] [Google Scholar]
  • 201.Palmer DN, Neverman NJ, Chen JZ, Chang CT, Houweling PJ, Barry LA, Tammen I, Hughes SM, Mitchell NL. Recent studies of ovine neuronal ceroid lipofuscinoses from BARN, the Batten Animal Research Network. Biochim Biophys Acta. 2015; 1852:2279–2286. [DOI] [PubMed] [Google Scholar]
  • 202.Kinarivala N, Trippier PC. Progress in the Development of Small Molecule Therapeutics for the Treatment of Neuronal Ceroid Lipofuscinoses (NCLs). J Med Chem. 2016; 59:4415–4427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Zhang Z, Butler JD, Levin SW, Wisniewski KE, Brooks SS, Mukherjee AB. Lysosomal ceroid depletion by drugs: therapeutic implications for a hereditary neurodegenerative disease of childhood. Nat Med. 2001; 7:478–484. [DOI] [PubMed] [Google Scholar]
  • 204.Hofmann SL, Lee LA, Lu JY, Verkruyse LA. Palmitoyl-protein thioesterase and the molecular pathogenesis of infantile neuronal ceroid lipofuscinosis. Neuropediatrics. 1997; 28:27–30. [DOI] [PubMed] [Google Scholar]
  • 205.Rogawski MA. Revisiting AMPA receptors as an antiepileptic drug target. Epilepsy Curr. 2011; 11:56–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.French JA, Krauss GL, Biton V, Squillacote D, Yang H, Laurenza A, Kumar D, Rogawski MA. Adjunctive perampanel for refractory partial-onset seizures: randomized phase III study 304. Neurology. 2012; 79:589–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Kovacs AD, Pearce DA. Attenuation of AMPA receptor activity improves motor skills in a mouse model of juvenile Batten disease. Exp Neurol. 2008; 209:288–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Kovacs AD, Saje A, Wong A, Szenasi G, Kiricsi P, Szabo E, Cooper JD, Pearce DA. Temporary inhibition of AMPA receptors induces a prolonged improvement of motor performance in a mouse model of juvenile Batten disease. Neuropharmacology. 2011; 60:405–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Kovacs AD, Weimer JM, Pearce DA. Selectively increased sensitivity of cerebellar granule cells to AMPA receptor-mediated excitotoxicity in a mouse model of Batten disease. Neurobiol Dis. 2006; 22:575–585. [DOI] [PubMed] [Google Scholar]
  • 210.Dhar S, Bitting RL, Rylova SN, Jansen PJ, Lockhart E, Koeberl DD, Amalfitano A, Boustany RM. Flupirtine blocks apoptosis in batten patient lymphoblasts and in human postmitotic CLN3- and CLN2-deficient neurons. Ann Neurol. 2002; 51:448–466. [DOI] [PubMed] [Google Scholar]
  • 211.Makoukji J, Saadeh F, Mansour KA, El-Sitt S, Al Ali J, Kinarivala N, Trippier PC, Boustany RM. Flupirtine derivatives as potential treatment for the neuronal ceroid lipofuscinoses. Ann Clin Transl Neurol. 2018; 5:1089–1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Park JW, Woo KJ, Lee JT, Lim JH, Lee TJ, Kim SH, Choi YH, Kwon TK. Resveratrol induces pro-apoptotic endoplasmic reticulum stress in human colon cancer cells. Oncol Rep. 2007; 18:1269–1273. [PubMed] [Google Scholar]
  • 213.Ghosh A, Rangasamy SB, Modi KK, Pahan K. Gemfibrozil, food and drug administration-approved lipid-lowering drug, increases longevity in mouse model of late infantile neuronal ceroid lipofuscinosis. J Neurochem. 2017; 141:423–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Hu J, Lu JY, Wong AM, Hynan LS, Birnbaum SG, Yilmaz DS, Streit BM, Lenartowicz EM, Thompson TC, Cooper JD, Hofmann SL. Intravenous high-dose enzyme replacement therapy with recombinant palmitoyl-protein thioesterase reduces visceral lysosomal storage and modestly prolongs survival in a preclinical mouse model of infantile neuronal ceroid lipofuscinosis. Mol Genet Metab. 2012; 107:213–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.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. [DOI] [PubMed] [Google Scholar]
  • 216.Tamaki SJ, Jacobs Y, Dohse M, Capela A, Cooper JD, Reitsma M, He D, Tushinski R, Belichenko PV, Salehi A, Mobley W, Gage FH, Huhn S, Tsukamoto AS, Weissman IL, Uchida N. Neuroprotection of host cells by human central nervous system stem cells in a mouse model of infantile neuronal ceroid lipofuscinosis. Cell Stem Cell. 2009; 5:310–319. [DOI] [PubMed] [Google Scholar]
  • 217.Selden NR, Al-Uzri A, Huhn SL, Koch TK, Sikora DM, Nguyen-Driver MD, Guillaume DJ, Koh JL, Gultekin SH, Anderson JC, Vogel H, Sutcliffe TL, Jacobs Y, Steiner RD. Central nervous system stem cell transplantation for children with neuronal ceroid lipofuscinosis. J Neurosurg Pediatr. 2013; 11:643–652. [DOI] [PubMed] [Google Scholar]
  • 218.Griffey M, Bible E, Vogler C, Levy B, Gupta P, Cooper J, Sands MS. Adeno-associated virus 2-mediated gene therapy decreases autofluorescent storage material and increases brain mass in a murine model of infantile neuronal ceroid lipofuscinosis. Neurobiol Dis. 2004; 16:360–369. [DOI] [PubMed] [Google Scholar]
  • 219.Griffey MA, Wozniak D, Wong M, Bible E, Johnson K, Rothman SM, Wentz AE, Cooper JD, Sands MS. CNS-directed AAV2-mediated gene therapy ameliorates functional deficits in a murine model of infantile neuronal ceroid lipofuscinosis. Mol Ther. 2006; 13:538–547. [DOI] [PubMed] [Google Scholar]
  • 220.Shyng C, Nelvagal HR, Dearborn JT, Tyynela J, Schmidt RE, Sands MS, Cooper JD. Synergistic effects of treating the spinal cord and brain in CLN1 disease. Proc Natl Acad Sci U S A. 2017; 114:E5920–E5929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Rozenberg A, Lykken E, Spratt K, Miller TJ, Gray S. Combination dosing of CLN1 gene therapy extends lifespan in a mouse model of infantile neuronal ceroid lipofuscinosis 2018. S124 p. [Google Scholar]
  • 222.Griffey M, Macauley SL, Ogilvie JM, Sands MS. AAV2-mediated ocular gene therapy for infantile neuronal ceroid lipofuscinosis. Mol Ther. 2005; 12:413–421. [DOI] [PubMed] [Google Scholar]
  • 223.Macauley SL, Roberts MS, Wong AM, McSloy F, Reddy AS, Cooper JD, Sands MS. Synergistic effects of central nervous system-directed gene therapy and bone marrow transplantation in the murine model of infantile neuronal ceroid lipofuscinosis. Ann Neurol. 2012; 71:797–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Roberts MS, Macauley SL, Wong AM, Yilmas D, Hohm S, Cooper JD, Sands MS. Combination small molecule PPT1 mimetic and CNS-directed gene therapy as a treatment for infantile neuronal ceroid lipofuscinosis. J Inherit Metab Dis. 2012; 35:847–857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Klawe C, Maschke M. Flupirtine: pharmacology and clinical applications of a nonopioid analgesic and potentially neuroprotective compound. Expert Opinion on Pharmacotherapy. 2009; 10:1495–1500. [DOI] [PubMed] [Google Scholar]
  • 226.Gavin M, Wen GY, Messing J, Adelman S, Logush A, Jenkins EC, Brown WT, Velinov M. Substrate Reduction Therapy in Four Patients with Milder CLN1 Mutations and Juvenile-Onset Batten Disease Using Cysteamine Bitartrate. JIMD Rep. 2013; 11:87–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Levin SW, Baker EH, Zein WM, Zhang Z, Quezado ZM, Miao N, Gropman A, Griffin KJ, Bianconi S, Chandra G, Khan OI, Caruso RC, Liu A, Mukherjee AB. Oral cysteamine bitartrate and N-acetylcysteine for patients with infantile neuronal ceroid lipofuscinosis: a pilot study. Lancet Neurol. 2014; 13:777–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.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. Mol Ther Methods Clin Dev. 2017; 4:204–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Katz ML, Coates JR, Sibigtroth CM, Taylor JD, Carpentier M, Young WM, Wininger FA, Kennedy D, Vuillemenot BR, O’Neill CA. Enzyme replacement therapy attenuates disease progression in a canine model of late-infantile neuronal ceroid lipofuscinosis (CLN2 disease). J Neurosci Res. 2014; 92:1591–1598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Sinclair J, Whiting R, Robinson G, Bibi K, Nguyen A, Cherukuri A, Henshaw J, De Hart G, Chandra S, O’Neill C, Katz M. Intravitreal enzyme replacement therapy attenuates retinal disease progression in a canine model of neuronal ceroid lipofuscinosis type 2 (CLN2). Molecular Genetics and Metabolism. 2018; 123:S132. [Google Scholar]
  • 231.Schulz A, Ajayi T, Specchio N, de Los Reyes E, Gissen P, Ballon D, Dyke JP, Cahan H, Slasor P, Jacoby D, Kohlschutter A, Group CLNS. Study of Intraventricular Cerliponase Alfa for CLN2 Disease. N Engl J Med. 2018; 378:1898–1907. [DOI] [PubMed] [Google Scholar]
  • 232.Cherukuri A, Cahan H, de Hart G, Van Tuyl A, Slasor P, Bray L, Henshaw J, Ajayi T, Jacoby D, O’Neill CA, Schweighardt B. Immunogenicity to cerliponase alfa intracerebroventricular enzyme replacement therapy for CLN2 disease: Results from a Phase 1/2 study. Clin Immunol. 2018; 197:68–76. [DOI] [PubMed] [Google Scholar]
  • 233.Tracy CJ, Whiting RE, Pearce JW, Williamson BG, Vansteenkiste DP, Gillespie LE, Castaner LJ, Bryan JN, Coates JR, Jensen CA, Katz ML. Intravitreal implantation of TPP1-transduced stem cells delays retinal degeneration in canine CLN2 neuronal ceroid lipofuscinosis. Exp Eye Res. 2016; 152:77–87. [DOI] [PubMed] [Google Scholar]
  • 234.Haskell RE, Hughes SM, Chiorini JA, Alisky JM, Davidson BL. Viral-mediated delivery of the late-infantile neuronal ceroid lipofuscinosis gene, TPP-I to the mouse central nervous system. Gene Ther. 2003; 10:34–42. [DOI] [PubMed] [Google Scholar]
  • 235.Passini MA, Dodge JC, Bu J, Yang W, Zhao Q, Sondhi D, Hackett NR, Kaminsky SM, Mao Q, Shihabuddin LS, Cheng SH, Sleat DE, Stewart GR, Davidson BL, Lobel P, Crystal RG. Intracranial delivery of CLN2 reduces brain pathology in a mouse model of classical late infantile neuronal ceroid lipofuscinosis. J Neurosci. 2006; 26:1334–1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Sondhi D, Johnson L, Purpura K, Monette S, Souweidane MM, Kaplitt MG, Kosofsky B, Yohay K, Ballon D, Dyke J, Kaminksy SM, Hackett NR, Crystal RG. Long-term expression and safety of administration of AAVrh.10hCLN2 to the brain of rats and nonhuman primates for the treatment of late infantile neuronal ceroid lipofuscinosis. Hum Gene Ther Methods. 2012; 23:324–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Sondhi D, Peterson DA, Edelstein AM, del Fierro K, Hackett NR, Crystal RG. Survival advantage of neonatal CNS gene transfer for late infantile neuronal ceroid lipofuscinosis. Exp Neurol. 2008; 213:18–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Katz ML, Tecedor L, Chen Y, Williamson BG, Lysenko E, Wininger FA, Young WM, Johnson GC, Whiting RE, 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. 2015; 7:313ra180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Souweidane MM, Fraser JF, Arkin LM, Sondhi D, Hackett NR, Kaminsky SM, Heier L, Kosofsky BE, Worgall S, Crystal RG, Kaplitt MG. Gene therapy for late infantile neuronal ceroid lipofuscinosis: neurosurgical considerations. J Neurosurg Pediatr. 2010; 6:115–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Kim K, Kleinman HK, Lee HJ, Pahan K. Safety and potential efficacy of gemfibrozil as a supportive treatment for children with late infantile neuronal ceroid lipofuscinosis and other lipid storage disorders. Orphanet J Rare Dis. 2017; 12:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Burnight ER, Giacalone JC, Cooke JA, Thompson JR, Bohrer LR, Chirco KR, Drack AV, Fingert JH, Worthington KS, Wiley LA, Mullins RF, Stone EM, Tucker BA. CRISPR-Cas9 genome engineering: Treating inherited retinal degeneration. Prog Retin Eye Res. 2018; 65:28–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Aldrich A, Bosch ME, Fallet R, Odvody J, Burkovetskaya M, Rama Rao KV, Cooper JD, Drack AV, Kielian T. Efficacy of phosphodiesterase-4 inhibitors in juvenile Batten disease (CLN3). Ann Neurol. 2016; 80:909–923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Cialone J, Augustine EF, Newhouse N, Adams H, Vierhile A, Marshall FJ, de Blieck EA, Kwon J, Rothberg PG, Mink JW. Parent-reported benefits of flupirtine in juvenile neuronal ceroid lipofuscinosis (Batten disease; CLN3) are not supported by quantitative data. J Inherit Metab Dis. 2011; 34:1075–1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Palmieri M, Pal R, Nelvagal HR, Lotfi P, Stinnett GR, Seymour ML, Chaudhury A, Bajaj L, Bondar VV, Bremner L, Saleem U, Tse DY, Sanagasetti D, Wu SM, Neilson JR, Pereira FA, Pautler RG, Rodney GG, Cooper JD, Sardiello M. mTORC1-independent TFEB activation via Akt inhibition promotes cellular clearance in neurodegenerative storage diseases. Nat Commun. 2017; 8:14338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Aberg L, Talling M, Harkonen T, Lonnqvist T, Knip M, Alen R, Rantala H, Tyynela J. Intermittent prednisolone and autoantibodies to GAD65 in juvenile neuronal ceroid lipofuscinosis. Neurology. 2008; 70:1218–1220. [DOI] [PubMed] [Google Scholar]
  • 246.Mitchison HM, Bernard DJ, Greene ND, Cooper JD, Junaid MA, Pullarkat RK, de Vos N, Breuning MH, Owens JW, Mobley WC, Gardiner RM, Lake BD, Taschner PE, Nussbaum RL. Targeted disruption of the Cln3 gene provides a mouse model for Batten disease. The Batten Mouse Model Consortium [corrected]. Neurobiol Dis. 1999; 6:321–334. [DOI] [PubMed] [Google Scholar]
  • 247.Chattopadhyay S, Ito M, Cooper JD, Brooks AI, Curran TM, Powers JM, Pearce DA. An autoantibody inhibitory to glutamic acid decarboxylase in the neurodegenerative disorder Batten disease. Hum Mol Genet. 2002; 11:1421–1431. [DOI] [PubMed] [Google Scholar]
  • 248.Seehafer SS, Ramirez-Montealegre D, Wong AM, Chan CH, Castaneda J, Horak M, Ahmadi SM, Lim MJ, Cooper JD, Pearce DA. Immunosuppression alters disease severity in juvenile Batten disease mice. J Neuroimmunol. 2011; 230:169–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Chang JW, Choi H, Cotman SL, Jung YK. Lithium rescues the impaired autophagy process in CbCln3(Deltaex7/8/Deltaex7/8) cerebellar cells and reduces neuronal vulnerability to cell death via IMPase inhibition. J Neurochem. 2011; 116:659–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Sarkar S, Floto RA, Berger Z, Imarisio S, Cordenier A, Pasco M, Cook LJ, Rubinsztein DC. Lithium induces autophagy by inhibiting inositol monophosphatase. J Cell Biol. 2005; 170:1101–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Augustine EF, Beck CA, Adams HR, Defendorf S, Vierhile A, Timm D, Weimer JM, Mink JW, Marshall FJ. Short-Term Administration of Mycophenolate Is Well-Tolerated in CLN3 Disease (Juvenile Neuronal Ceroid Lipofuscinosis). JIMD Rep. 2019; 43:117–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Drack AV, Mullins RF, Pfeifer WL, Augustine EF, Stasheff SF, Hong SD. Immunosuppressive Treatment for Retinal Degeneration in Juvenile Neuronal Ceroid Lipofuscinosis (Juvenile Batten Disease). Ophthalmic Genet. 2015; 36:359–364. [DOI] [PubMed] [Google Scholar]
  • 253.Mirza M, Volz C, Karlstetter M, Langiu M, Somogyi A, Ruonala MO, Tamm ER, Jagle H, Langmann T. Progressive retinal degeneration and glial activation in the CLN6 (nclf) mouse model of neuronal ceroid lipofuscinosis: a beneficial effect of DHA and curcumin supplementation. PLoS One. 2013; 8:e75963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Cain JT, Likhite S, Whitel K, Meyers K, Timm D, Rinaldi F, Hughes SM, Lee SY, Kaspar BK, Weimer JM, editors. Testing safety and efficacy of AAV9-CLN6 gene therapy in a mouse model of CLN6-Batten disease The 15th International Conference on Neuronal Ceriod Lipofuscinosis (Batten Disease); 2016 October 5–8, 2016; Boston. [Google Scholar]
  • 255.Likhite S, Timm D, White KA, Cain JT, Dennys-Rivers C, Rinaldi F, Motti D, Corcoran S, Weimer JM, Kaspar BK, Meyer K. From Bench to Bedside: Gene Therapy for Batten (CLN6) Disease. Mol Ther. 2018; 26:307–308. [Google Scholar]
  • 256.Weimer J, Cain J, Johnson T, White K, Likhite S, Meyer K. Promise of AAV9 gene therapy in the treatment of Batten disease: Systematic approach in therapy design reduces pathological and behavioral deficits and prolongs survival in mouse models of CLN3-, CLN6-, and CLN8-Batten disease 2019. S151 p. [Google Scholar]
  • 257.Piguet F, Alves S, Cartier N. Clinical Gene Therapy for Neurodegenerative Diseases: Past, Present, and Future. Hum Gene Ther. 2017; 28:988–1003. [DOI] [PubMed] [Google Scholar]
  • 258.Byrne BJ. Safety First: Perspective on Patient-Centered Development of AAV Gene Therapy Products. Mol Ther. 2018; 26:669–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Amicus Therapeutics. Amicus establishes gene therapy pipeline for lysosomal storage disorders (LSDs) Conference call and webcast. http://ir.amicusrx.com/static-files/c62086c7-1aae-4e0e-83d2-1a2e4ebc65cc2018.[last accessed 6/18/19]
  • 260.Amicus Therapeutics. Amicus Data Overview at 15th Annual WorldSymposium. http://ir.amicusrx.com/static-files/e5022131-ff0b-4b03-960c-de1f512cce8b2019. [last accessed 6/18/19]
  • 261.Mitchell N. Longitudinal studies and the development of gene therapy for ovine neuronal ceroid lipofuscinoses. 2016. [Thesis, University of Otago, Christchurch, New Zealand] [Google Scholar]
  • 262.Kay GW, Palmer DN. Chronic oral administration of minocycline to sheep with ovine CLN6 neuronal ceroid lipofuscinosis maintains pharmacological concentrations in the brain but does not suppress neuroinflammation or disease progression. J Neuroinflammation. 2013; 10:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Yu TW, Hu C, Kim J, Larson A, Lee A, Black L, El Achkar CM, Soucy A, Vaze J, Armant M, Belur NR, Kuniholm A, Douville J, Augustine EF, Pendergast M, Goldkind S, Tyndall K, Goodlett B, Waisbren S, Riley B, Cornelissen L, Pereira L, Reed C, Snyder R, Patterson A, Poduri A, Mazzulli JR, Biffi A, Bodamer O, Berde C, editors. PgmNr 3570: Patient-customized oligonucleotide therapy for an ultra-rare genetic disease. American Society of Human Genetics; San Diego, October 16–20, 2018. [Google Scholar]
  • 264.Deeg HJ, Shulman HM, Albrechtsen D, Graham TC, Storb R, Koppang N. Batten’s disease: failure of allogeneic bone marrow transplantation to arrest disease progression in a canine model. Clin Genet. 1990; 37:264–270. [DOI] [PubMed] [Google Scholar]
  • 265.Elger B, Schneider M, Winter E, Carvelli L, Bonomi M, Fracasso C, Guiso G, Colovic M, Caccia S, Mennini T. Optimized synthesis of AMPA receptor antagonist ZK 187638 and neurobehavioral activity in a mouse model of neuronal ceroid lipofuscinosis. ChemMedChem. 2006; 1:1142–1148. [DOI] [PubMed] [Google Scholar]
  • 266.Cooper JD, Messer A, Feng AK, Chua-Couzens J, Mobley WC. Apparent loss and hypertrophy of interneurons in a mouse model of neuronal ceroid lipofuscinosis: evidence for partial response to insulin-like growth factor-1 treatment. J Neurosci. 1999; 19:2556–2567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Zeman RJ, Peng H, Etlinger JD. Clenbuterol retards loss of motor function in motor neuron degeneration mice. Exp Neurol. 2004; 187:460–467. [DOI] [PubMed] [Google Scholar]
  • 268.Shevtsova Z, Garrido M, Weishaupt J, Saftig P, Bahr M, Luhder F, Kugler S. CNS-expressed cathepsin D prevents lymphopenia in a murine model of congenital neuronal ceroid lipofuscinosis. Am J Pathol. 2010; 177:271–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Pike LS, Tannous BA, Deliolanis NC, Hsich G, Morse D, Tung CH, Sena-Esteves M, Breakefield XO. Imaging gene delivery in a mouse model of congenital neuronal ceroid lipofuscinosis. Gene Ther. 2011; 18:1173–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Arrant AE, Onyilo VC, Unger DE, Roberson ED. Progranulin Gene Therapy Improves Lysosomal Dysfunction and Microglial Pathology Associated with Frontotemporal Dementia and Neuronal Ceroid Lipofuscinosis. J Neurosci. 2018; 38:2341–2358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Amado DA, Rieders JM, Diatta F, Hernandez-Con P, Singer A, Mak JT, Zhang J, Lancaster E, Davidson BL, Chen-Plotkin AS. AAV-Mediated Progranulin Delivery to a Mouse Model of Progranulin Deficiency Causes T Cell-Mediated Toxicity. Mol Ther. 2019; 27:465–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Abeona Therapeutics. Pipeline https://www.abeonatherapeutics.com/research-development#pipeline2019 [last accessed 6/18/19]
  • 273.Amicus Therapeutics. Programs and Pipeline https://www.amicusrx.com/programs-pipeline/2018 [last accessed 6/18/19]
  • 274.Biomarin. Products/pipeline https://www.biomarin.com/products/brineura2019 [last accessed 6/18/19]
  • 275.SBIRSource. Development of N-tert-(Butyl)hydroxylamine (NtBuHA) as a therapeutic agent for treating Infantile Neuronal Ceroid Lipofuscinosis (INCL). In: DHHS, editor. https://sbirsource.com/sbir/awards/167461-development-of-n-tert-butyl-hydroxylamine-ntbuha-as-a-therapeutic-agent-for-treating-infantile-neuronal-ceroid-lipofuscinosis-incl2019. [last accessed 6/18/19]
  • 276.Polaryx Therapeutics. The Pipeline https://www.polaryx.com/pipeline.html2017 [last accessed 6/18/19]
  • 277.Regenxbio. RGX-181 is our product candidate for the treatment of late-infantile neuronal ceroid lipofuscinosis Type 2 (CLN2 disease), a form of Batten disease. https://www.regenxbio.com/rgx-181/2018 [last accessed 6/18/19]
  • 278.Spark Therapeutics. Our Scientific Platform and Programs http://sparktx.com/scientific-platform-programs/2018 [last accessed 6/18/19]
  • 279.Rosenberg JB, Kaplitt MG, De BP, Chen A, Flagiello T, Salami C, Pey E, Zhao L, Ricart Arbona RJ, Monette S, Dyke JP, Ballon DJ, Kaminsky SM, Sondhi D, Petsko GA, Paul SM, Crystal RG. AAVrh.10-Mediated APOE2 Central Nervous System Gene Therapy for APOE4-Associated Alzheimer’s Disease. Hum Gene Ther Clin Dev. 2018; 29:24–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Smalley E First AAV gene therapy poised for landmark approval. Nat Biotechnol. 2017; 35:998–999. [DOI] [PubMed] [Google Scholar]
  • 281.US Food and Drug Administration. FDA approves innovative gene therapy to treat pediatric patients with spinal muscular atrophy, a rare disease and leading genetic cause of infant mortality In: FDA, editor. https://www.fda.gov/news-events/press-announcements/fda-approves-innovative-gene-therapy-treat-pediatric-patients-spinal-muscular-atrophy-rare-disease2019. [last accessed 6/18/19]
  • 282.Darrow JJ. Luxturna: FDA documents reveal the value of a costly gene therapy. Drug Discov Today. 2019; 24:949–954. [DOI] [PubMed] [Google Scholar]
  • 283.Lukacs Z, Nickel M, Murko S, Nieves Cobos P, Schulz A, Santer R, Kohlschutter A. Validity of a rapid and simple fluorometric tripeptidyl peptidase 1 (TPP1) assay using dried blood specimens to diagnose CLN2 disease. Clin Chim Acta. 2019; 492:69–71. [DOI] [PubMed] [Google Scholar]
  • 284.Mathavarajah S, McLaren MD, Huber RJ. Cln3 function is linked to osmoregulation in a Dictyostelium model of Batten disease. Biochim Biophys Acta Mol Basis Dis. 2018; 1864:3559–3573. [DOI] [PubMed] [Google Scholar]
  • 285.Somogyi A, Petcherski A, Beckert B, Huebecker M, Priestman DA, Banning A, Cotman SL, Platt FM, Ruonala MO, Tikkanen R. Altered Expression of Ganglioside Metabolizing Enzymes Results in GM3 Ganglioside Accumulation in Cerebellar Cells of a Mouse Model of Juvenile Neuronal Ceroid Lipofuscinosis. Int J Mol Sci. 2018; 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.NCBI. CLN6 CLN6 transmembrane ER protein [ Homo sapiens (human) ] In: NLM, editor. https://www.ncbi.nlm.nih.gov/gene/549822019. [last accessed 6/18/19]
  • 287.Danyukova T, Ariunbat K, Thelen M, Brocke-Ahmadinejad N, Mole SE, Storch S. Loss of CLN7 results in depletion of soluble lysosomal proteins and impaired mTOR reactivation. Hum Mol Genet. 2018; 27:1711–1722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Batten Disease Family Association. CLN10 disease, congenital, neonatal and late infantile http://www.bdfa-uk.org.uk/cln10-disease-congenital-neonatal-and-late-infantile/2014 [last accessed 6/18/19]

RESOURCES