Abstract
Batten disease (also known as neuronal ceroid lipofuscinoses) constitutes a family of devastating lysosomal storage disorders that collectively represent the most common inherited paediatric neurodegenerative disorders worldwide. Batten disease can result from mutations in 1 of 13 genes. These mutations lead to a group of diseases with loosely overlapping symptoms and pathology. Phenotypically, patients with Batten disease have visual impairment and blindness, cognitive and motor decline, seizures and premature death. Pathologically, Batten disease is characterized by lysosomal accumulation of autofluorescent storage material, glial reactivity and neuronal loss. Substantial progress has been made towards the development of effective therapies and treatments for the multiple forms of Batten disease. In 2017, cerliponase alfa (Brineura), a tripeptidyl peptidase enzyme replacement therapy, became the first globally approved treatment for CLN2 Batten disease. Here, we provide an overview of the promising therapeutic avenues for Batten disease, highlighting current FDA-approved clinical trials and prospective future treatments.
Batten disease is a family of primarily autosomal recessive, progressive neuropaediatric disorders, also known as neuronal ceroid lipofuscinoses (NCLs), characterized by seizures and visual, cognitive and motor decline, ending in premature death. Batten disease is caused by mutations in 1 of 13 different genes1,2. The worldwide prevalence of Batten disease is ~1 in 100,000 live births3–5, and until the past few years, no effective treatments had been available to halt progression of these diseases. Therapy development for Batten disease has been limited because the function of a number of the disease-associated proteins is only partially understood. In 2017, the FDA approved an enzyme replacement therapy (ERT) called cerliponase alfa (Brineura; BioMarin Pharmaceutical), the first treatment to delay the progression of CLN2 Batten disease. In parallel with this momentous achievement, a number of research teams are using a multitude of therapeutic modalities to accelerate the development of novel treatments for other forms of Batten disease at an unprecedented pace. Here, we provide an overview of Batten disease, including the unique challenges faced by researchers studying this disease and the innovative strategies that they are pursuing to reshape the treatment landscape for these devastating diseases.
Classification of the NCLs
The first reported description of Batten disease was by Otto Christian Stengel in 1826. He described a case of progressive dementia and blindness in four siblings6. This initial report was followed by similar reports by Frederick Batten in 1903 (refs7,8). In 1969, the term NCL was coined on the basis of the ultrastructural pattern of accumulated lipofuscin or ceroid1 — a feature that helped to distinguish this group of diseases from similar neurological disorders. Before the discovery of mutated genes in NCLs, patients were classified by a combination of age of onset and ultrastructural patterns of these deposits9,10. The disease was first classified as infantile onset (with granular deposits11), late-infantile onset (with curvilinear profiles or rectilinear complex12), juvenile onset (with fingerprint profiles13) or adult onset (with granular deposits14). Additionally, an ultra-rare congenital NCL (with granular deposits) was identified15–23. Several cases described over the past decade do not follow these classic pathology-based classifications (for example, patients with TPP1 mutations who do not have onset until their early teens or with CLN6 mutations that result in disease onset in the late teen years). In addition, several genes have been identified in which mutations lead to the accumulation of autofluorescent storage material (ASM), and the associated conditions have been grouped as Batten disease. Thus, the field has slowly transitioned to a nomenclature that reflects genetic mutation rather than pathology.
The first genes associated with Batten disease were discovered in 1995 through exhaustive genetic linkage methods24,25. These findings opened the door for genetic characterization and classification of individual forms of the disease. Improvements in genetic sequencing technologies now enable Batten disease to be delineated according to mutations in 1 of 13 different genes26 (Table 1). In 2012, an updated classification system was proposed to provide thorough characterization of patients’ genetic background, taking into account biochemical and clinical phenotypes (reviewed previously27). In the proposed classification, the disorder classification has been simplified and codified numerically according to the affected gene (for instance, CLN1 Batten disease).
Table 1 |.
Batten disease subtypes and their clinical presentation
NCL subtype (affected gene) | Neuroradiological imaging findingsa | EEG abnormalities | Behavioural tendencies | Visual changes | Microscopic findings | Major clinical manifestations |
---|---|---|---|---|---|---|
CLN1 (PPT1)38,71,246–255 | Hyperintense, periventricular high-signal rims of white matter; decreased NAA and increased choline; and severely enlarged lateral ventricles | Loss of sleep spindles at ~2 years; attenuated reaction to passive eye opening and/or closing; background activity disturbances; and reduced amplitude | Irritability and hyperexcitability | Optic atrophy; unrecordable ERG at 4 years; and blindness | GRODs | Motor coordination loss; choreoathetosis; stereotypic movements; myoclonic jerks; decelerated head growth; and death by 10 years of age |
CLN2 (TPP1)38,71,243,253,256–259 | Infratentorial atrophy; hypointense thalamic nuclei; reduction in NAA; increased myo-inositol and Glu:Gln ratio in the white matter; and severely enlarged lateral ventricles | Occipital spike in response to slow flash; irregular slow activity; focal spikes; absence of sleep spindles; and large VEPs and SEPs | Behavioural disturbances, Including anxiety and agitation | Progressive vision loss leading to blindness; and diminished ERG | CLPs | Myoclonus; ataxia; motor decline; spasticity; dystonic features; choreoathetosis; hypotonia; seizures; and death in early adolescence |
CLN3 (CLN3)35,38,71,253,260–265 | Cerebellar atrophy; enlarged third ventricle and cerebral sulci; and hypointense thalamic nuclei | Progressive background disorganization; and spike-and-slow-wave complexes | Anxiety; aggression; delayed speech; and depression | Progressive loss of vision; and pigmentary retinopathy | FPPs and vacuolated lymphocytes | Seizures; rigidity; hypokinesia; impaired balance; myoclonus; and death in second or third decade |
CLN4 (DNAJC5)38,71,253,266,267 | Parieto-occipital cortical atrophy; cerebellar atrophy; hyperintense periventricular areas; and corpus callosum thinning | Slow background; polyphasic spikes; and slow-wave discharges | Inappropriate laughter | No visual impairment | GRODs; CLPs; and FPPs | Myoclonus; grand mal seizures; dementia; ataxia; and facial dyskinesia |
CLN5 (CLN5)38,54,55,71,264,268 | Cerebellar atrophy; diminished signal intensity in thalamic nuclei; and increased signal intensity in periventricular white matter and internal capsule | Large VEPs and SEPs; and occipital spikes in response to slow flash | ND | Progressive visual decline leading to blindness; and macular degeneration | GRODs; CLPs; and FPPs | Clumsiness; seizures; dementia; motor coordination loss; myoclonus; and death between 14 and 36 years of age |
CLN6 (CLN6)38,71,269 | Deep cortical layer-specific neuron loss; and cerebellar atrophy | Background slowing; and high-amplitude discharges in response to photic stimulation | ND | Visual failure leading to blindness | CLPs; RLC; and FPPs | Motor decline; seizures; dysarthria; and ataxia |
CLN7 (MFSD8)38,71 | Cerebellar atrophy; corpus callosum thinning; and hypointense thalamic nuclei | Occipital spikes; and background slowing | Personality changes | Visual failure leading to blindness | CLPs; RLC; and FPPs | Motor decline; seizures; and myoclonus |
CLN8 (CLN8)38,71,270–272 | Cerebellar atrophy; corpus callosum thinning; and hyperintensity of white matter | Slow background; components of high amplitude; epileptiform discharges; and abnormal VEPs and SEPs | Irritability; restlessness; and inactivity | Retinopathy; visual decline at around 4−6 years of age; and ERG absent | GRODs; CLPs; and FPPs | Myoclonus; tonic–clonic seizures; motor decline; and progressive dementia |
CLN10 (CTSD)15–22,38,71,273,274 | Diminished head growth in utero; myoclonic fetal seizures; enlarged lateral ventricles; hypointense cerebral and cerebellar white matter; and decreased NAA and increase in myo-inositol | Completely depleted EEG pattern | ND | ND | GRODs | Severe respiratory distress at birth; axial and limb hypotonia; extreme microcephaly; overriding sutures; and death within hours after birth |
CLN11 (GRN)38,71,275 | Cerebellar atrophy | Polyspike-wave discharges with posterior emphasis; and severe attenuation of rod and cone responses | ND | Progressive vision loss; and retinal dystrophy | FPPs | Myoclonic seizures; cerebellar ataxia; and cognitive decline |
CLN12 (ATP13A2)38,66,271–273 | Cortical and subcortical atrophy; and decreased glucose use in grey matter, especially the thalamus and posterior association cortex | ND | Mood disturbances and dysarthric speech | No visual changes | GRODs | Loss of coordination; myoclonus; seizures; spasticity; rigidity; akinesia; and muscular atrophy |
CLN13 (CTSF)38,71,276,277 | Cerebellar atrophy; frontal and parietal cortical atrophy; and periventricular hyperintensities | No epileptiform activity | Behaviour and personality Abnormalities and depression | ND | FPPs | Dementia; seizures; motor coordination decline; ataxia; tremor; dysarthria; and hyperreflexia |
CLN14 (KCTD7)38,66,276–278 | Cortical and cerebellar atrophy; and corpus callosum thinning | Slow dysrhythmia; multifocal high-amplitude epileptiform discharges; photosensitivity; and occipital spikes | ND | Visual loss; diminished pupillary light reflex; and optic atrophy | GRODs; CLPs; RLC; and FPPs | Motor decline; myoclonic seizures; ataxia; myoclonus; and dysarthria |
CLPs, curvilinear profiles; ERG, electroretinogram; FPPs, fingerprint profiles; GRODs, granular deposits; NAA, N-acetyl aspartate; NCL, neuronal ceroid lipofuscinose; ND, not determined; RLC, rectilinear complex; SEP, somatosensory evoked potential; VEP, visual evoked potential.
Clinical diagnosis and disease progression
The diagnosis of Batten disease is based on a combination of clinical signs and symptoms, ophthalmological evaluations, EEG and brain MRI and is subsequently confirmed with genetic and biochemical tests2,28,29. In developing countries, where access to neurophysiology, imaging and genetic testing is limited, cellular pathology and electroretinograms continue to be used as diagnostic tools. The clinical features historically used to identify Batten disease were the result of a compilation of case reports, and natural history reports have continued to inform disease characterization and clinical trial preparation8,30–34. In the clinical setting, age and onset of symptoms still play a key part in suspicion of Batten disease and eventually in pursuit of confirmatory genetic testing for the disease. Common clinical hallmarks of Batten disease include visual impairment, inability to achieve normal developmental milestones and/or developmental regression, behavioural problems, progressive cerebral atrophy, seizures, cognitive decline and dementia10,19,32,35–38. However, the order and frequency in which these symptoms present vary between the different subtypes and between the different variants for each genetic mutation. Although an extensive clinical review is out of the scope of this paper, we briefly discuss genotype–phenotype correlations of the most common neuropaediatric diseases. Clinical generalities for all subtypes of Batten disease are included in Table 1.
Common subtypes of Batten disease
CLN1.
Inheritance of CLN1 Batten disease is autosomal recessive. The disease is caused by mutations in palmitoyl protein thioesterase (encoded by PPT1) a lysosomal enzyme involved in the removal of palmitate residues from proteins. PPT1 is also associated with important cellular pathways, including synaptogenesis and synaptic maintenance, endosomal trafficking and lipid metabolism39–45. Classic onset of CLN1 disease is in the first year of life, with irritability, developmental arrest and rapid regression, deceleration of head circumference growth, hypotonia, myoclonic seizures and progressive vision loss with optic nerve atrophy. In some variants of CLN1 disease, the clinical presentation mimics CLN3 disease, with slower progression and less severe seizures than in typical CLN1 disease. This variant is probably associated with some residual activity of PPT1 that is sufficient to cause a protracted disease course but not enough to prevent disease progression. With regards to genotype–phenotype correlations, severe forms of the disease are associated with Arg151X homozygous mutations, whereas Thr75Pro substitution is associated with a later onset and more protracted clinical course than other CLN1 mutations46,47.
CLN2.
Inheritance of CLN2 Batten disease is autosomal recessive, and affected patients have mutations in the lysosomal enzyme tripeptidyl peptidase (encoded by TPP1). The proposed mechanism of action involves the removal of tripeptides from the amino terminus of small polypeptides. Classic clinical presentation includes acute onset of myoclonic seizures (usually refractory to medication), ataxia, developmental arrest and regression, central hypotonia with appendicular spasticity and rapidly progressing motor decline. Similar to CLN1 disease, residual TPP1 activity is associated with a protracted disease course, as patients who retain small amounts of active TPP1 demonstrate slow disease progression48. Although these protracted cases are rare globally, they occur in ~50% of affected individuals in the South American population48,49. Additionally, mutations in TPP1 have been described in a subgroup of patients with spinocerebellar ataxia 7, which further suggests that residual activity is associated with less severe or atypical forms of the disease50,51.
CLN3.
Inheritance of CLN3 Batten disease is autosomal recessive, and the disease is caused by mutations in battenin (encoded by CLN3), a ubiquitously expressed protein of unknown function that is associated with cellular homeostasis and neuronal survival. Patients usually present with central vision loss followed by behavioural and cognitive problems, motor decline, parkinsonism, speech apraxia with echolalia and seizures. Most of the patients harbour a 966 bp deletion in CLN3, and individuals with protracted disease have been reported, all of whom were compound heterozygous, with some lacking the most common mutation52. As with CLN1 and CLN2 disease, additional rare cases of CLN3 mutations have been reported that lead to late-onset (~20–40 years of age) nonsyndromic retinal degeneration53, adding to the clear genotype–phenotype correlations of these diseases.
CLN5.
Inheritance of CLN5 disease is autosomal recessive and is caused by mutations in ceroid lipofuscinosis neuronal protein 5 (encoded by CLN5), a transmembrane protein of unclear function. Most cases of CLN5 disease have been described in patients of Finnish descent, but mutations have been identified in patients with diverse ethnic backgrounds54–57. The most common clinical features include intellectual disability, ataxia and myoclonic epilepsy, and the classic form has an age of onset between 4 and 7 years. Similarly to other Batten disease forms, variants with late onset and/or mild symptoms have been reported58.
CLN6.
The CLN6 disease subtype was initially described in a large cohort of patients from Costa Rica, where this form is one of the most common causes of Batten disease in the country. Nevertheless, patients with CLN6 disease have been identified from across a broad range of ethnic backgrounds52,59. CLN6 disease is caused by mutations in ceroid lipofuscinosis neuronal protein 6 (encoded by CLN6), a transmembrane protein of unknown function, and is inherited in an autosomal recessive manner. Symptoms include early-onset seizures, motor decline and ataxia, halt in developmental milestones and, subsequently, developmental regression and progressive cognitive decline and speech problems. Classic forms of CLN6 disease have an onset of symptoms between 3 and 5 years of age, and some variants are associated with a protracted disease course with slow progression. Additionally, a rare form of adult-onset CLN6 disease, known as Kufs disease, exists in both autosomal recessive and dominant forms and manifests at around age 30 years. This variant might have therapeutic implications, as it might be amenable to pre-symptomatic treatments that would be otherwise impossible in paediatric-onset forms of the disease without prenatal genetic diagnosis14,60.
Diagnosis
Many neurodegenerative diseases of childhood share similar symptoms, so delayed diagnosis of Batten disease is not uncommon61. Definitive diagnosis can be challenging in infants and toddlers, in whom thorough neurological and ophthalmological evaluations require a skilled and knowledgeable professional. Batten disease presents after a period of apparently normal development, despite the lack of a functional protein that is important for brain function, and so one must conclude that a small therapeutic window exists where successful interventions can halt and/or prevent the progression of the disease. Consequently, early diagnosis is crucial for optimal therapeutic results, and prospective trials must include patients who are only mildly affected by disease. In this respect, the clinician must be familiar with the clinical features of the disease and suspect the diagnosis in order to pursue confirmatory testing as quickly as possible.
Microscopic, biochemical and genetic assays
Conventional diagnosis of Batten disease phenotypes had been based on microscopic analysis of storage deposits. However, genetic testing and enzyme activity assays are now standard of care. These approaches are readily available in most developed countries and can be conducted reliably prenatally with amniotic fluid or fetal cells23,24,38,62–65. In countries where genetic and biochemical testing is not readily available, skin biopsy samples can be collected, and accumulation of lipopigments, a pathological hallmark of these disorders, can be evaluated38,66–70. Electron microscopic analysis of the ultrastructural patterns of cellular deposits helps to delineate patients into possible subtypes of Batten disease. Lipopigment morphotypes tend to correlate strongly with genotype37, but genotype does not always align with the clinical presentation of the disease. Thus, storage deposits can be used as confirmation of Batten disease subtype rather than as a strictly diagnostic tool, with different morphotypes indicative of different forms of the disease38,71 (summarized in Table 1). Additionally, patients with CLN3 Batten disease have vacuolated lymphocytes; this feature can be assessed by regular blood smear and can aid differential diagnosis of patients72,73.
Biochemical enzyme activity assays of three lysosomal enzymes that are mutated in various forms of the disease (PPT1, TPP1 and cathepsin D (encoded by CTSD)) are conclusive methods for diagnosis of CLN1, CLN2 and CLN10 Batten disease, respectively23,74–77. For PPT1 assays, patient samples — including fibroblasts, leukocytes, amniocytes, dried blood spots and chorionic villi — are mixed with a synthetic substrate that is hydrolysed by active PPT1 enzyme to release a detectable fluorophore76,77. Absence of the fluorophore confirms CLN1 disease. Similarly, TPP1 and CTSD activity assays use fluorogenic Ala–Ala–Phe-coupled and haemoglobin-coupled synthetic substrates, respectively, in which the active enzyme of interest cleaves the substrate for fluorophore activation78–80. Each of these assays provides a rapid and reliable tool to aid clinicians in delineation between prospective disorders during initial diagnosis.
Technological advances in genomic sequencing make whole-exome, whole-genome or direct Sanger sequencing the most definitive analytical tools to precisely identify patient mutations29,81,82. These methods are particularly helpful in patients with clinical variants and/or patients with atypical or novel mutations. The vast differences in age of onset, clinical presentations, severity and disease course of Batten disease mean that genetic testing remains one of the most informative tools that clinicians and scientists can use to determine specific genetic mutations that might result in protein dysfunction. Currently, clinicians are primarily using commercially available gene panels or whole-exome sequencing to diagnose patients with Batten disease. A mutation database now exists that catalogues all the known variant mutations of Batten disease genes, consisting of ~446 disease-causing mutations81. Continual collaboration between clinicians and investigators is essential to reach prompt diagnoses for patients affected with these devastating diseases.
Neuropathology
Several pathological hallmarks exist for Batten disease, and these changes are well characterized in human tissues and animal models (Tables 1,2). Massive neuronal loss and accumulation of intracellular ASM are predominant features in all patients with Batten disease. Cortical, subcortical, cerebellar, brainstem and spinal cord neurons are affected by pathology to varying degrees depending on the disease variant. Cortical layer-specific loss of neurons has been described in layers II and V in CLN2, CLN3 and CLN5 disease83–85, layers II and III in CLN4 disease85, the occipital lobe and layer V in CLN6 disease85 and complete disorganization of neurons in CLN10 disease15,20. Hippocampal neuron degeneration and microglial activation occur selectively in the CA2–CA4 regions, whereas the CA1 region is seemingly spared85–87. Cortical and hippocampal neuron loss is accompanied by loss of Purkinje and granular cells in the cerebellum and dentate nucleus, extreme glial activation, severe astrocytosis and demyelination of white matter1,15,17,20,21,85,88. Visual problems generally comprise one or more of the following: retinal atrophy, photoreceptor death, gliosis, bull’s eye maculopathy, disc atrophy, peripheral pigmentary changes and attenuation of retinal vasculature12,85,89–92.
Table 2 |.
Summary of mouse models of Batten disease used for therapeutic development
Mouse model | Mutation | Selective CNS pathologya | Visual phenotype | Behavioural phenotype | Therapeutic testing | Refs |
---|---|---|---|---|---|---|
CLN1 | Ppt1−/− | GABAergic neuron loss in cortex, thalamic nuclei and cerebellum; and early neuronal loss in cervical, thoracic ventral horn and lumbar dorsal horn | Vision loss | Seizures and loss of motor coordination; and death at ~6 months | ERT, AAV2, AAV2/5, AAV2/9, SCT, SMT or BMT | 121,122,146,147,154,155,179,232,235,278–287 |
Cln1R151X | As for Ppt1−/− | As for Ppt1−/− | As for Ppt1−/− | SMT | 288,289 | |
CLN2 | Cln2−/− | Disruption of myelin in white matter of corticospinal tracts; mild forebrain atrophy; and Purkinje cell degeneration | ND | Tremors, seizures and loss of motor coordination; and abnormal hunched gait | ERT, AAV1, AAV2, AAV5, AAV8, AAVrh.10 or SMT | 125,149,151,195,290–292 |
Cln2R208X | As for Cln2−/− | ND | As for Cln2−/− | SMT | 293 | |
CLN3 | Cln3Δex7–8 | Purkinje cell loss; and GABAergic interneuron loss | Retinal atrophy and altered pupillary light reflex | Motor coordination deficits | AAV2, AAV9 or SMT | 157,294–300 |
Cln3Δex1–6 | Neuronal loss in striatum and cerebellum | Dorsal lateral geniculate nuclei degeneration; optic nerve degeneration; and retinal atrophy | Motor coordination deficits | SMT | 158,211,231,299,301–305 | |
CLN6 | Cln6nclf | Decreased cortical and cerebellar glutamine, glutamate and GABA | Retinal atrophy | Hindlimb paresis at ~8 months; memory and learning deficits; and death at 12–15 months | AAV2, AAV9 orSCT | 156,171,306–308 |
CLN8 | Cln8mnd | Neuronal death in hippocampus and spinal cord; and GABAergic neuron loss | Retinal atrophy (photoreceptor cells) | Seizures; limb paresis at 6 months; hyperactivity; aggression; memory loss; and death at ~12 months | SMT | 309–318 |
CLN10 | Ctsd−/− | ASM, astrocytosis and microglial activation in somatosensory cortex and/or thalamic nuclei; axonal degeneration; and neuronal death | ND | Congenital disease | AAV1/2 | 80,188,319–322 |
AAV, adeno-associated virus; AAV1/2, AAV1 cassette with an AAV2 capsid; AAV2/5, AAV2 cassette with an AAV5 capsid; AAV2/9, AAV2 cassette with an AAV9 capsid; ASM, autofluorescent storage material; BMT, bone marrow transplantation; ERT, enzyme replacement therapy; ND, not determined; SCT, stem cell therapy; SMT, small-molecule treatment.
All models present with ASM accumulation, astrocytosis, microglial activation and neuronal loss in the cortex and thalamic nuclei; regional model-specific changes are highlighted.
Advances in therapy development
Although little is known about the cellular function of many of the CLN proteins (reviewed previously93), robust animal models that effectively recapitulate features of human disease (Table 2) have provided an improved understanding of the pathology and disease hallmarks of Batten disease that has accelerated therapeutic development. Many different tailored mouse models have been engineered to match individual, uncommon mutations from patients to enable specific therapies to be tested. Large, multi-laboratory collaborations among the Batten disease research community have also substantially increased the rate at which novel treatments that slow or halt the rapid progression of Batten disease are being developed and tested. Additionally, researchers and clinicians have teamed up to develop a number of much needed tools to monitor patients as the clinical phase of drug discovery is initiated (box 1). Together with cerliponase alfa, these advances have enabled numerous promising therapies to rapidly enter preclinical and clinical testing phases for multiple Batten disease subtypes. We highlight a number of these studies here and consider their limitations and therapeutic challenges.
Box 1 |. Developing comprehensive tools for studying Batten disease.
For >30 years, a number of patient advocacy groups have worked tirelessly with federal, academic and private partners to change the drug development system, which seemingly worked against drug discovery for rare diseases and condemned them to remain essentially orphaned. In 1983, the US Congress passed the Orphan Drug Act, which uses tax credits, grants and extended market exclusivity to incentivize development of treatments for rare diseases. Many countries worldwide followed with similar initiatives. In 2002, the US Rare Disease Act was enacted and strengthened therapy development efforts by establishing the Office of Rare Diseases and allocating increased research funding. Even with these giant strides forward, most rare diseases still have ineffective treatments and cures. In 2016, the US 21st Century Cures Act was enacted to “expedite the discovery, development and delivery of new treatment and cures and maintain America’s global status as the leader in biomedical innovation.” These combined efforts have enabled the development and marketing of drugs for rare diseases and have increased the number of treatments brought to market from fewer than 10 between 1973 and 1983 to well over 600 to date241.
However, for many diseases, including Batten disease, the fight is not over. By definition, patients with rare diseases are few. Methodologies for finding patients and tracking natural history as well as standardized disease outcome tools are needed.
Standardized patient rating scale
The University of Rochester Batten Center developed a Unified Batten Disease Rating Scale (UBDRS). The UBDRS initially focused on evaluating and tracking the disease progression of patients with CLN3 Batten disease but subsequently has been developed for use in establishing patient ratings for various forms of Batten disease8,31,242,243.
Comprehensive natural history studies
Over the past decade, efforts have been made to develop detailed natural histories of individual forms of Batten disease, an essential resource required for successful and efficacious clinical studies8,32,34. In the past few years, the DEM-CHILD International NCL Registry was established through a European and US-based consortium to continuously develop and refine patient assessment tools, monitor the prevalence of each form of Batten disease and develop detailed natural history studies that link genetic mutation information with clinical data for all forms of Batten disease244.
Centralized patient registries
A number of patient advocacy groups, family foundations and academic research teams, including the Batten Disease Support and Research Association, have driven efforts to collate comprehensive registries of patients with Batten disease worldwide. These group registries focus on one form of the disease, for example, the Weill Cornell CNS scale137 and the Hamburg CLN2 scale30. In the past few years, the Coordination of Rare Diseases at Sanford (CoRDS) programme has included development of a centralized international patient registry for all forms of Batten disease with the goal of connecting as many patients with Batten disease with clinicians and researchers as possible to help advance treatments and cures for these rare diseases245.
Development and continuous refinement of these tools will ensure that, when therapies do become available, the necessary resources for successful clinical programmes are in place. Moreover, rather than working separately, many of these efforts are coordinated to ensure information from one programme works in concert with that from other efforts.
Use of animal models in Batten disease
Animal models of Batten disease enable the genetic, molecular, biochemical and metabolic mechanisms that lead to the rapid neurodegeneration observed in patients to be studied on a substantially abbreviated timescale. Animal models now exist for nearly every form of Batten disease (a comprehensive list of these models is reviewed elsewhere94). In the past few years, these models have been key contributors to preclinical testing of the safety and efficacy of prospective therapies and, in some cases, have helped researchers to tailor therapies to individual mutations (Table 2). In this respect, mouse models have been an integral part of Batten disease research. However, neurodevelopmental, neuroanatomical and mechanistic differences between mice and humans present challenges for translation and are apparent in the phenotypic presentation of Batten disease mouse models (reviewed previously95,96). Specifically, disease presentation varies depending on strain in several Batten disease mouse models, retinal degeneration does not occur or is limited in several models, and most models show delayed or reduced mortality compared with their human counterparts97 (reviewed previously98). Perhaps as a consequence of these differences, several clinical trials have shown that therapies that showed promise in these mouse models lacked efficacy in patients (although they have generally been safe and well tolerated)99–101.
Large animal models resemble humans more closely than mouse models do in terms of anatomy, physiology, size, lifespan, biochemistry and genetics, and bridge the substantial gap between the current Batten disease mouse models and the clinic (reviewed previously102). A variety of models have been developed or discovered in sheep, dogs and pigs, and a naturally occurring nonhuman primate model of CLN7 Batten disease was also reported in the past year103–108. Although each of these models has clear limitations with regards to translation to the clinic — including size, cost and unavoidable anatomical and physiological differences — each of these species has the potential to improve the accuracy of not only models of Batten disease but also models of therapeutic delivery mechanisms, pharmacodynamics and toxicology109–112. Swine models of Batten disease might prove particularly insightful, given the similarities in pig and human brain development, structure and size113,114. Indeed, swine models of human diseases such as ataxia telangiectasia115, cystic fibrosis116,117 and neurofibromatosis type 1 (ref.118) have been very useful in recapitulating disease hallmarks in instances where mouse models have failed.
Enzyme replacement therapy
ERT is a treatment strategy for enzyme deficiencies that introduces purified recombinant enzymes via intravenous, intracerebroventricular or intrathecal injection. The injected enzyme is then delivered to the correct cellular compartment via receptor-mediated uptake. Four of the Batten disease subtypes result from deficiencies in soluble lysosomal enzymes: CLN1 (PPT1), CLN2 (TPP1), CLN10 (CTSD) and CLN13 (cathepsin F (CTSF))81,119. Preclinical studies on the efficacy of recombinant human PPT1 ERT indicated that both intravenous and intrathecal delivery systems were well tolerated in Ppt1−/− mice and reduced many of the pathological hallmarks of the disease, including ASM, astrocytosis and glial activation120–123. In vitro experiments have also tested potential ERT of a recombinant human TPP1 (rhTPP1) proenzyme in human TPP1-deficient fibroblasts. The proenzyme is not enzymatically active until acidification autocatalytically converts it to the mature form124. This process requires efficient trafficking and targeting of the enzyme to the lysosomal compartment of recipient cells. Trafficking of lysosomal hydrolases, including TPP1, requires mannose-6-phosphate post-translational modification for proper endocytosis and targeting of the proteins to the lysosome124,125 (fig. 1). rhTPP1 retains mannose-6-phosphate post-translational modifications, which results in receptor-mediated endocytosis of the enzyme by the mannose-6-phosphate receptor, trafficking to the lysosomal compartment, restoration of TPP1 activity and reduction in ASM accumulation in fibroblasts124. Successful treatment of patients with Batten disease would require efficient targeting of ERTs to the CNS, which necessitates that the protein bypasses the blood–brain barrier (BBB). Consequently, intravenous administration of rhTPP1 is unlikely to be efficacious. Permeabilization of the BBB is possible126–128, but this process can further exacerbate neuronal damage129,130 and therefore is not ideal for treatment in all patients. In initial safety and efficacy tests of rhTPP1 delivery in mouse and dog models of CLN2 disease, recombinant enzyme was delivered through catheters implanted in the lateral ventricle (intracerebroventricular) or subarachnoid space (intrathecal) that remained intact during the duration of the studies125,131. This delivery method resulted in widespread diffusion of rhTPP1 in both hemispheres of the brain and significant reduction in the region-specific neuronal loss, ASM, reactive astrocytosis and tremors associated with CLN2 Batten disease progression125,131. Additionally, rhTPP1 treatment preserved cognitive function and significantly extended the lifespan of CLN2 model animals in a dose-dependent manner.
Fig. 1. Gene therapy and enzyme replacement therapy strategies in Batten disease.
a | Gene therapy as a treatment for Batten disease. Viral vectors are being explored to introduce a corrected copy of the genes mutated in Batten disease. Tropism, biodistribution and carrying capacity must be considered in selecting the type or serotype of virus to ensure the most efficient delivery. Modified adeno-associated viruses (AAVs) target expression of the CLN genes to affected cells. Once transduced into the cell, the target transgene is episomally expressed (1) (solid arrows), although limited evidence supports low-level random integration (2) (dashed arrows). The transgene is then translated, and the mature protein is trafficked to its resident location, with careful monitoring after viral transduction to ensure targeting to the correct sites. b | Enzyme replacement therapy (ERT) as a treatment for Batten disease. Multiple forms of Batten disease result from mutations in soluble lysosomal enzymes and thus might be amenable to cross-correction by ERT to partially restore levels of these enzymes in the CNS. This strategy, successfully demonstrated by cerliponase alfa, is currently available for treatment of CLN2 Batten disease. Cerliponase alfa, a recombinant proenzyme containing a mannose-6-phosphate post-translational modification, is delivered intraventricularly to the patient’s brain. This proenzyme is targeted to the mannose-6-phosphate receptor on the plasma membrane and endocytosed into the cell (1), where it fuses with the late endosome (2) before being delivered to the lysosome, where the acidic environment autocatalytically converts the enzyme to the mature, active form (3). CLN3, battenin; CLN5, ceroid lipofuscinosis neuronal protein 5; CLN6, ceroid lipofuscinosis neuronal protein 6; CLN8, ceroid lipofuscinosis neuronal protein 8; CTSD, cathepsin D; CTSF, cathepsin F; ER, endoplasmic reticulum; ERGIC, endoplasmic reticulum-Golgi intermediate compartment; PPT1, palmitoyl protein thioesterase; TPP1, tripeptidyl peptidase.
On the basis of these preclinical results, BioMarin Pharmaceutical manufactured an rhTPP1 enzyme, cerliponase alfa (also known as BMN 190 or Brineura), and evaluated its safety and efficacy for treatment of patients with CLN2 Batten disease in a phase I/II study132, the results of which were published in 2018. In the initial trial, 24 patients between the ages of 3 and 16 years with CLN2 Batten disease were enrolled133. Three multicentre, multi-national trials are currently underway134–136. Patient inclusion criteria for the initial 48-week open-label dose-escalation study were as follows: ≥3 years of age at enrolment; diagnosis of CLN2 Batten disease by TPP1 enzyme activity in leukocytes or by molecular analysis identifying two known pathogenic mutations; and a two-domain score of 3–6 on the motor–gait and language domains of the Hamburg Scale133. Exclusion criteria included previous receipt of stem cell therapy, gene therapy or ERT for CLN2 disease; diagnosis of additional neurological disease; contraindications for neurosurgery or MRI; an episode of generalized motor status epilepticus or severe infection within 4 weeks of the first infusion; presence of ventricular abnormality or shunt; and known hypersensitivity to any components of cerliponase alfa.
Baseline clinical scores (including both the Weill Cornell CNS scale137 and the Hamburg CLN2 scale30 (NCL-2 rating scales), which quantify seizures, loss of language, motor and visual function), vital signs and EEG, electrocardiography and MRI findings were recorded before surgical implantation of the intracerebroventricular reservoir and cannula in the lateral ventricle of the right hemisphere. Implantation was confirmed by MRI. Blood and cerebrospinal fluid (CSF) were also sampled to monitor potential biomarkers, immunogenicity and pharmacokinetics during the study. All patients received cerliponase alfa at the time of surgery or within 14 days after surgery and every other week thereafter. Infusions were administered at 2.5 ml/h over 4 h, followed by electrolyte infusion. Patients 1–9 were assigned to three cohorts of three patients, who each received an initial dose of 30, 100 or 300 mg. For the dose-escalation period, each initial dose was administered for 4 weeks before increasing to the subsequent dosing schedule. After the escalation period, all participants received a stable dose of 300 mg cerliponase alfa every other week for at least 48 weeks. Of the 24 patients enrolled in the initial trial, 23 are enrolled in the ongoing long-term extension study135 for continued treatment and monitoring for 240 weeks.
Efficacy of the ERT in patients was monitored with the NCL-2 rating scales for motor–language scores and total scores in four domains — motor skills, language, vision and seizures133 — and compared with natural history data from 42 patients from two CLN2 Batten disease registries (Weill Cornell CNS scale137 and Hamburg CLN2 scale30). Rates of decline from baseline in the motor–language score were calculated over the 48-week and 96-week periods133. The scoring for motor and language function consisted of a score of 0–3 for each domain, for a total score of 6. A score of 3 represented normal gait and intelligible language with no decline noted. A 1-point decline in the motor score indicated the ability to walk with obvious instability and possible falls. A 2-point decline corresponded to the requirement for assistance to walk or having the ability to only crawl, and a score of 0 represented loss of the ability to walk or crawl. A 1-point decline in the language domain score indicated recognizable abnormalities in speech, a 2-point decline represented language that was difficult to understand with little ability to formulate intelligible words, and a score of 0 corresponded to loss of vocalizations and no intelligible words. Secondary efficacy parameters were based on brain atrophy (grey matter volumes), as measured with high-resolution T1-weighted MRI, and changes after treatment were descriptively summarized. Primary efficacy changes in treated patients were compared with matched patients in the historical control group that had the closest values in baseline scores, age and genotype. Safety analysis consisted of reporting of any adverse events, summarized by system organ class, preferred medical term, relationship to treatment and severity.
ERT resulted in substantial delay of motor, language and visual decline, as evidenced by stabilization of NCL-2 rating scores and substantial reduction in cortical volume loss. Historical control patients experienced a mean (± s.d.) adjusted rate of decline in motor–language scores of 2.06 ± 0.15 points per 48-week period, whereas the treated patients never reached a 2-point drop in motor–language scores and averaged declines of 0.38 ± 0.10 points over the same period133. Direct comparison of 1:1 matched pairs among the treated and historical groups revealed a mean motor–language score decrease of 0.20 ± 0.67 and 0.50 ± 0.71 points for the treated group compared with 1.90 ± 1.23 and 2.80 ± 1.10 points for historical controls at 48 weeks and 96 weeks of treatment, respectively133. Additionally, two treated patients who had perfect motor–language scores at the start of the study did not experience a single-point decline at the conclusion of 96 weeks. Total four-domain score declines for treated patients were 0.30 ± 1.70 and 0.40 ± 2.08 compared with 2.80 ± 2.04 and 4.30 ± 2.26 for historical controls after 48 weeks and 96 weeks of treatment, respectively133. The importance of this finding is emphasized if one considers that small declines on the NCL-2 rating scale (a simple 0–3 score system) can represent the difference between walking or being wheelchair bound. Grey matter volumes of treated patients decreased by 6.7% on average; however, volumetric changes were not recorded for historical controls. Nevertheless, this value is much lower than that reported from separate studies, which indicate an average loss of cortical volume of ~15–20% annually for patients with untreated CLN2 Batten disease138.
Gene therapy
Adeno-associated virus (AAV)-mediated gene therapy is a promising option for the treatment of neurodegenerative diseases and lysosomal storage disorders and is effective in several models of Batten disease. AAV-mediated gene therapy has been shown to be safe and effective in several clinical trials for lysosomal storage disorders, including Pompe disease and mucopolysaccharidoses139–142 (reviewed previously143). Previous studies have also shown that reintroduction of a lysosomal enzyme via gene therapy can rescue enzyme activity systemically and in the CNS. In 2018, Abeona Therapeutics reported that, in patients with mucopolysaccharidosis type IIIA (a lysosomal storage disorder associated with mutations in the lysosomal enzyme SGSH), a single intravenous dose of self-complementary AAV9 (scAAV9) containing human SGSH (hSGSH) was well tolerated, crossed the BBB, increased SGSH enzyme activity, reduced accumulation of heparin sulfate in CSF and urine and improved cognition143. To date, nearly 40 clinical trials have been listed on the NIH online clinical trial registry that are using various serotypes of AAV to treat a variety of neurodegenerative diseases and lysosomal storage disorders, including Parkinson disease (PD), Leber hereditary optic neuropathy, Pompe disease, mucopolysaccharidosis types I, II, IIIa, IIIb and VI, CLN6 Batten disease, CLN2 Batten disease, spinal muscular atrophy, Alzheimer disease (AD) and Charcot–Marie–Tooth neuropathy type 1a.
AAV carries single-stranded DNA and naturally infects humans. Twelve AAV serotypes and >108 serovars exist, each differing in its antigenicity and tropism (reviewed previously144,145). Successful infection, transduction and biodistribution of the AAV for therapy depend on a number of factors, including the route of administration and the specific tropism of the different serotypes. Efficiency of gene cassette expression can be increased by using scAAV vectors, which eliminates the need for double-stranded DNA synthesis at the cost of a reduction in the capacity of the gene expression cassette. When targeting AAVs to the CNS, it is important to consider not only the dose, route, tropism and targeting of cells outside the CNS but also the potential for immune response to the viral capsid or transgene product. AAV exposure in humans is a common occurrence and results in production of neutralizing antibodies that can negatively affect AAV transduction and gene transfer. Thus, patients need to be monitored for the presence of antibodies against AAV or the transgene, and in cases where they are present, alternative approaches need to be explored.
Several preclinical studies have focused on the use of various AAV serotypes in treatment of various forms of Batten disease. Multiple intracranial injections of AAV2 encoding human PPT1 (hPPT1) have been used to successfully treat a mouse model of CLN1 Batten disease (Ppt1−/−). AAV2–hPPT1 increased PPT1 enzyme activity and rescued many of the classic Batten disease pathological features, but only in areas near the injection site, probably owing to limited viral spread throughout the CNS146. In addition, a greater number of viral injections was associated with a greater rescue of the Batten disease pathology, including a correction of motor and learning behaviours. However, injection number had no effect on median survival147.
Studies in a canine model of CLN2 Batten disease showed that intraventricular delivery of AAV2 encoding canine TPP1 (caTPP1) into the circulating CSF led to widespread transduction of AAV2–caTPP1 to the ependymal lining of the third and fourth ventricles. TPP1, secreted from the ependymal cells, was subsequently detected in the cortex and cerebellum. This increase in the levels of TPP1 delayed the onset of Batten disease symptoms, reduced glial activation, rescued behavioural phenotypes and increased longevity. Coadministration of mycophenolate mofetil, an inhibitor of B and T lymphocyte proliferation, starting 5 days before AAV2–caTPP1 administration to reduce the number of neutralizing antibodies led to even greater increases in longevity. Although secreted caTPP1 was taken up by neurons, the virus was only able to transduce the ependymal cells rather than spreading throughout the CNS148.
Although AAV2 has displayed strong local levels of transduction, distribution of AAV2 from the local site of administration has been hindered by its strong heparin sulfate proteoglycan binding149. To rectify this issue, the AAV2 gene expression cassette was combined with capsids from other AAV serotypes. One study used a recombinant AAV1 capsid expressing hTPP1 in an AAV2 gene expression cassette. AAV1–hTTP1 was injected into the striatum, hippocampus deep cerebellar nucleus, motor cortex, thalamus and medulla of 4-week-old pre-symptomatic or 11-week-old post-symptomatic Cln2 mutant mice. Treatment at both time points restored TPP1 activity to wild-type levels, and mice treated presymptomatically had better pathological outcomes, including reduction in ASM, decreased axon degeneration, improved motor function and increased median lifespan. These data indicate that gene therapy has the potential to delay and prevent Batten disease pathology but not reverse it150. Delivery of hTTP1 using AAVrh.10, an AAV serotype derived from rhesus macaques, yielded a higher level of TPP1 activity than AAV2 and had a broader distribution in a mouse model151,152. AAVrh.10 carrying human CLN3 also rescued several phenotypes associated with CLN3 Batten disease in mice, including astrocyte activation but not microglial activation153.
An AAV2 cassette with an AAV5 capsid (AAV2/5) expressing hPPT1 has been used to rescue phenotypes in Ppt1−/− mice. A bilateral intracranial injection into the anterior cortex at postnatal day 1 restored PPT1 enzyme activity to wild-type levels, increased lifespan by 10 weeks, reduced activation of microglia and astrocytes and ameliorated motor deficits for 7 months154. In a subsequent study, AAV2/9–hPPT1 was used in the same mouse model and administered via intrathecal injection to the lumbar spinal cord, an intracranial injection to the anterior cortex, hippocampus and cerebellum or a combination of both at postnatal day 1 or 2. Both intrathecal and intracranial injections yielded many improvements in Batten disease pathology, including reduction of ASM, improved motor function, reduced microglial and astrocyte activation and increased median survival. As might be expected, the spinal intrathecal injection had greater local effects in the spinal cord, whereas the intracranial injection had greater effects in the cortex; however, the greatest overall benefits were observed in the animals that received combined intrathecal and intracranial injections, which increased median survival by 6–8 months compared with either injection alone155.
The population of cells targeted for transduction might also influence the efficacy of viral treatments. One study compared the efficacy of two AAV serotypes to correct Batten disease-related pathologies in the eye. An AAV2/8 virus carrying human CLN6 delivered intravitreally to Cln6nclf mice (an established model of CLN6 disease) was unable to correct photoreceptor loss. However, bipolar cell-specific expression of a modified AAV2/2 serotype 7m8 successfully prevented photoreceptor loss156.
In other studies, an AAV9 capsid has been combined with the AAV2 inverted terminal repeat (ITR) gene cassette to generate a scAAV9–hCLN3 vector driven by one of two promoters, a Mecp2 promoter driving low expression or a chicken β-actin (CB) promoter driving high expression. The scAAV9–hCLN3 vector was delivered intravenously into 1-month-old Cln3Δex7/8 mice (knock-in mice with exons 7 and 8 removed)157. The high-expression CB promoter virus resulted in a threefold to eightfold increase in Cln3 expression compared with the Mecp2 promoter; however, the increase in levels of CLN3 did not correlate with increased benefit. The low-expression promoter actually corrected more disease pathologies, including motor coordination, reduced astrocytosis, microglial activation and lysosomal pathology, whereas the CB promoter increased glial activation in the thalamus and failed to rescue motor deficit. However, whether these differing outcomes were due to change in gene expression or the types of cells expressing the scAAV9–hCLN3 was unclear158.
Preclinical studies show that a single intracerebroventricular injection of scAAV9.cb.hCLN6 in the Cln6nclf mouse model and intrathecal injection in non-human primates result in widespread CNS expression of the CLN6 transcript, including in the eye and optic nerve, and result in substantial reduction of ASM and reactive gliosis159,160. On the basis of these data, a phase I/II clinical trial of scAAV9.cb.hCLN6 for treatment of CLN6 Batten disease was initiated161, and is currently underway. The study involves use of an AAV9 capsid carrying the AAV2 gene cassette with hCLN6 injected intrathecally into the subarachnoid space of the lumbar spine of patients. Using a similar paradigm with a modified lower expression promoter, the same group conducted preclinical studies of scAAV9.hCLN3 in the Cln3Δex7/8 mouse model and nonhuman primates and reported similar widespread CNS expression and reduction of pathological hallmarks162. These observations formed the basis of a recently initiated phase I/IIa clinical trial163.
Additionally, one clinical trial164 assessed the efficacy of AAV2 for treatment of CLN2 Batten disease. Twelve intracranial injections of AAV2 encoding hTPP1 were administered to patients in the moderate or severe stages of CLN2 Batten disease, according to the Steinfeld et al. neurological assessment scale30. Results indicated that the treatment was well tolerated and that no serious adverse events were attributable to the administration of AAV, and the rate of decline was significantly slowed when compared with natural history studies165 according to the neurological assessment scale. Two other trials are ongoing166,167 that use a similar paradigm to the first, with the exception of the use of an AAVrh.10 serotype with the aim to achieve a broader biodistribution of viral transduction151,153.
Stem cell therapies
Several groups have sought to use stem cell-based therapies to address the visual deficits associated with progression of Batten disease168. One such study involved use of a clonal neural stem cell (NSC) line that was transduced with a lentivirus that expressed ciliary neurotrophic factor (CNTF), a cytokine that has been shown to rescue retinal degeneration in various animal models169,170. These CTNF-expressing NSCs were injected intravitreally into one eye, and control green fluorescent protein (GFP)-expressing NSCs were injected into the other. The NSCs subsequently attached and formed a cell layer on the lens where they stopped proliferation and preferentially differentiated into CNTF-secreting astrocytes, enabling the consistent secretion of the neuroprotective cytokine. Six weeks after the injection, the retinae injected with CTNF-expressing cells showed increased retinal thickness and increased photoreceptor numbers compared with GFP controls171. In another study, bone marrow-derived mesenchymal stem cells (MSCs) were transduced with AAV2–hTPP1 and injected into the eyes in a canine model of CLN2 Batten disease. The injected MSCs remained in the vitreous and could be observed for 9 weeks after injection. Eyes from PPT1-null dogs that were not treated with the hTPP1-expressing MSCs began to display retinal lesions at 7 months that progressively worsened, whereas hTPP1-treated dogs developed no eye lesions and had preserved retinal function as observed by retinal histology and electroretinography172.
In a phase I dose-escalation trial173, human CNS-derived stem cells (HuCNS-SCs) that secreted endogenous TPP1 and PTT1 were engrafted into patients with CLN1 or CLN2 Batten disease. Patients were injected with a total of 5 × 108 cells (low dose) or 1 × 109 cells (high dose) at six subcortical sites and in both lateral ventricles. The treatment was well tolerated, with no adverse events that were associated with the course of Batten disease. Post-mortem PCR analysis of samples from two of the patients detected engrafted stem cells 357 days and 918 days after injection, indicating integration and longevity of HuCNS-SCs after engraftment101.
Small-molecule therapies
Over the past few decades, several pharmaceutical and biological agents (referred to collectively here as small-molecule therapies) have been tested in various models of Batten disease. On the basis of the pathogenesis of Batten disease, preclinical approaches have predominately focused on small molecules that improve lysosomal or autophagic health, serve as immune modulators or neuroprotective agents, or increase transcript or protein abundance.
Pharmacological chaperones and readthrough technologies.
Lysosomal enzymes undergo a careful quality check process. Much of the quality control process is completed in the endoplasmic reticulum, where these enzymes are produced before trafficking to the lysosome, although some evidence supports a Golgi-mediated quality check that involves trafficking of the protein back to the endoplasmic reticulum for degradation174. Enzymes that are unstable or improperly folded might not be trafficked to the lysosome, might accumulate inappropriately in other organelles or might be prematurely degraded175. As many patients with Batten disease carry missense mutations that can result in enzymes that are present but only partially functional, pharmacological chaperones are a promising therapeutic agent for these individuals176. Pharmacological chaperones are small molecules that bind to and stabilize target proteins in the endoplasmic reticulum, enabling them to be properly trafficked to the lysosome and circumvent premature degradation177,178. Although a number of chaperones have been tested preclinically in a number of lysosomal storage disorders, only a few of these agents have moved forward to clinical trials177. In Batten disease, one study of the efficacy of chaperones in lymphoblast lines from patients with CLN1 disease showed a twofold increase in PPT1 activity179. Currently, no other pharmacological chaperones have been tested in Batten disease cell lines or animal models.
Nonsense mutations that yield premature stop codons and truncated protein products or nonsense-mediated decay have been reported in patients with Batten disease180. Compounds that increase transcript and/or protein abundance, such as readthrough compounds (compounds that override premature termination codons (PTCs) resulting from nonsense mutations) and antisense oligonucleotides, have been tested with varying success. Ataluren (also known as PTC124) is an orally administered small molecule that prevents premature translation termination at PTCs by interacting with the ribosome and promoting insertion of near-cognate tRNAs at the nonsense site181. Ataluren treatment increased transcript levels in several cell lines from patients with CLN1 Batten disease and in several tissues in mouse models of CLN1 disease but was not able to cross the BBB at appropriate doses. Similarly, gentamicin, an aminoglycoside antibiotic that binds to the aminoacyl-tRNA site of the 30S subunit, successfully increased PPT1 and TPP1 transcript levels and enzyme activity in cell lines from patients with CLN1 or CLN2 Batten disease182,183 but has not been tested in mouse models of Batten disease to date. Antisense oligonucleotides, small modified nucleic acids that bind to target RNA sequences to elicit a specific change in translation, have been used successfully in various genetic diseases184 (including spinal muscular atrophy) but have yet to be applied to Batten disease.
Autophagy modulators and substrate reduction therapies.
Deficits in autophagy have been implicated in several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), AD and PD185–187. In several mouse models of Batten disease, membrane-bound LC3-II (a characteristic marker of autophagic membranes) was shown to be a constituent of storage material pathology, which is suggestive of immature autophagosome formation and improper autophagosome–lysosome fusion188–190. Additionally, CLN3 has been found to interact with the autophagic enzymes ATG3 and ATG7, which further implies a role for autophagy in Batten disease pathology191. Autophagy modulators have been explored as small-molecule therapeutics for Batten disease. Specifically, small-molecule PPARα agonists, such as fenofibrate, bezafibrate and gemfibrozil, were found to have beneficial effects in lymphoblast lines from patients with CLN3 Batten disease, including mitigation of autophagy deficits192–194. The FDA-approved agent gemfibrozil was later shown to have benefits in a mouse model of CLN2 Batten disease, in which it decreased cellular accumulates, improved motor coordination and increased longevity195. Mechanistically, PPARα is known to enhance levels of transcription factor EB (TFEB), which subsequently binds to promoters of genes involved in lysosome biogenesis, increasing their expression196,197. TFEB translocation to the nucleus can be targeted therapeutically by inhibition of protein kinase B (AKT). Transport of TFEB to the nucleus is regulated by AKT-driven phosphorylation of TFEB, and inhibition of that phosphorylation results in increased nuclear TFEB and activation of the coordinated lysosomal expression and regulation (CLEAR) signalling network. TFEB activation similarly resulted in enhanced clearance of aggregates, improved behaviour and increased longevity in multiple mouse models of Batten disease, which demonstrates the therapeutic potential of targeting this pathway198. The therapeutic potential of targeting autophagy and/or TFEB through other pathways such as the PI3K–mTOR pathway or AMP-activated protein kinase remains to be examined in Batten disease199.
Substrate reduction therapies (SRTs) inhibit the enzymes required for the production of the accumulates that typically aggregate in lysosomal storage disorders. In the context of Batten disease, cysteamine bitartrate (Cystagon; Orphan Europe), an SRT currently used for cystinosis, successfully reduced cysteine thioester accumulates in patients with CLN1 Batten disease, although it did not delay disease progression in the small number of patients observed200,201. Further preclinical testing of SRTs is needed to determine the potential of these treatments for patients with Batten disease.
Immune modulators and neuroprotective compounds.
Neuroinflammation is a prominent feature of Batten disease and has been shown to exacerbate neurodegeneration202,203. Additionally, patients with CLN3 Batten disease produce autoantibodies against several proteins, including glutamic acid decarboxylase 2 (GAD2, also known as GAD65) and α-fetoprotein, which suggests an autoimmune component to these diseases119,124,204,205. Immunotherapies have been explored in a number of other neurodegenerative diseases, including PD, ALS and AD, with preclinical success206–209. Surprisingly, even the complete depletion of all microglia in the brain was well tolerated in AD mouse models and reduced plaque pathology and improved cognition (such as that accomplished by PLX5622, an inhibitor of colony-stimulating factor 1 receptor signalling that is required for microglial survival)210. Immunomodulation has been broadly studied in Batten disease. Treatment of Cln3 mutant mice with mycophenolate mofetil improved motor coordination, reduced levels of serum autoantibodies and reduced neuroinflammation211. On the basis of these results, a phase II safety trial of mycophenolate mofetil was initiated in 2011 (ref.99,212). Although the compound was well tolerated, it was generally ineffective in preventing clinical outcomes in patients with CLN3 disease. However, as the compound was given for only a short time, further study might be warranted to determine the potential of mycophenolate mofetil in a clinical setting99. In CLN1 and CLN3 disease mouse models, the FDA-approved small molecules fingolimod (which impairs lymphocyte emigration into the brain via sphingosine-1-phosphate receptor modulation) and teriflunomide (which reduces the proliferation of activated immune cells via pyrimidine nucleotide synthesis inhibition) reduced neuron loss, brain atrophy and retinal thinning213–216. Studies of the efficacy of the steroid prednisolone showed that it reduced levels of GAD65 autoantibodies and improved motor symptoms in older patients with CLN3 Batten disease (aged 17 and 18 years) but did not improve motor symptoms or autoantibody levels in younger patients (aged 6–13 years), though the treatment did have a positive effect on these patients’ IQs217. Additionally, prednisolone produced adverse psychiatric effects and recurrent infections in many patients. However, the efficacy of other steroids is currently being explored in other neurodegenerative diseases, including allopregnanolone in Niemann–Pick disease type C and vamorolone in Duchenne muscular dystrophy218–220. Several agents that modulate neuroinflammation indirectly through the regulation of cAMP levels have been screened in preclinical models of Batten disease, including a series of phosphodiesterase 4 inhibitors (rolipram, roflumilast or PF-06266047), all of which had a positive effect on Batten disease-related pathologies and behaviours221. Taken together, the findings suggest that immunotherapies have clinical relevance to patients with Batten disease but must be weighed carefully against adverse effects of immune modulation222.
Lastly, the effects of neuroprotective compounds such as cannabinoids, σ1 receptor agonists, excitotoxicity and oxidative stress reducers, cytoskeletal stabilizers, c-Abl tyrosine kinase inhibitors and anti-apoptotic compounds have been studied broadly in neurodegenerative disorders223–229. Excitotoxicity has been of particular interest in Batten disease, as increased AMPA receptor activity has been reported in the cerebellum of Cln3 mutant mice, and antagonist treatment substantially improved motor coordination in this model230,231. Other neuroprotective compounds studied in the context of Batten disease include flupirtine, an anti-apoptotic compound with multiple modes of action that was beneficial in patient cell lines, and N-(tert-butyl)hydroxylamine, an antioxidant that improved motor coordination and survival in Ppt1 mutant mice232,233. To date, clinical trials have not been initiated for any uniquely neuroprotective compounds specifically for any form of Batten disease, but use of these compounds in other neurodegenerative disease could pave the way for future studies.
Combinatorial treatments.
Over the past several decades, it has become increasingly common to target multiple pathologies of the same disease to maximize benefit. In diseases such as Batten disease, in which the genes of interest are expressed across several cell and tissue types, one treatment is unlikely to fend off all clinical presentations. Use of multiple therapies has been shown to improve treatment efficacy in animal models, such as use of teriflunomide with fingolimod in two CLN mouse models, ibuprofen with lamotrigine in Cln3 mutant mice216,234,235 and AAV2/5–5-PPT1 and bone marrow transplant in Ppt1−/− mice. Single molecules that target multiple pathologies or use of various routes of administration for the same therapy might also be beneficial. With gene therapy, the field has seen success with combined forebrain and cerebellar delivery of AAV2–PPT1 in Ppt1−/− mice147, combined intracranial and intrathecal delivery of AAV9 in Ppt1−/− mice and AAV1 and AAV2 delivery across several distinct brain regions150,155. Taken together, use of several treatment strategies might offer additional benefits to patients with neurodegenerative disease, but the benefits of this approach must be weighed carefully against the additional adverse effects that combined treatments might bring236.
Therapeutic challenges and considerations.
Several challenges must be overcome to design a suitable clinical trial in Batten disease, in which patient populations can be small and consequently underpowered, the therapeutic window for treatment can be small or elapsed in some patients and natural history studies can be lacking or incomplete, with the latter issue yielding a lack of reliable, non-invasive outcome measurements of therapeutic efficacy (reviewed previously237). Despite the focus of the Batten disease community in the past few years on biomarker discovery in a variety of subtypes, a consistent, robust, non-invasive biomarker has yet to be discovered238,239. Additionally, substantial practical challenges exist for patients, clinicians and researchers in that much of the expertise in Batten disease is concentrated in a few facilities, which limits access to clinical information and care240.
Conclusion
The successful development of treatments for rare disease, including Batten disease, requires the development and validation of an arsenal of tools with which to tackle these debilitating conditions. Over the past two decades, scientists and clinicians within the Batten disease community have worked to ensure that tools are in place to enable progression towards effective treatments at an unprecedented pace. Armed with a comprehensive battery of well-characterized models of many forms of Batten disease, including large and small animal models and a vast bank of patient cell lines (including libraries of patient-induced pluripotent stem cell lines), as well as well-established pathological and behavioural strategies for monitoring disease progression in these models, the preclinical Batten disease research community sits at a turning point. Modern tools for drug discovery, including high-content and high-throughput screening and advances in medical chemistry, enable us to move agents through (or eliminate ineffective agents from) the drug discovery pipeline much more efficiently than ever before. Growing partnerships with pharmaceutical companies have provided translational scientists with access to libraries of novel and repurposed agents for screening in Batten disease. Globally, Batten disease clinical research teams are working together to ensure that comprehensive natural history studies, patient registries and diagnostic rating scales are in place well before the launch of clinical trials rather than belatedly trying to develop these resources once a therapy is ready to enter a clinical programme. The advances in ERT, gene therapy and pharmaceuticals in Batten disease, in combination with the genomic medicine revolution that biomedical research is entering, will set an unprecedented pace for the speed of development of much needed therapies for Batten disease. Moreover, access to early patient diagnosis and the ever increasing number of clinical trials opening for patients have drawn new scientists with unique skill sets to the field, and the Batten disease research community is becoming a model of how effective, efficient rare disease research can be accomplished by working together.
Key points.
The FDA approval of the enzyme replacement therapy cerliponase alfa (Brineura) for the treatment of CLN2 Batten disease is an important milestone in Batten disease therapy.
Promising results from preclinical research indicate that gene therapy — particularly approaches that use adeno-associated virus — represents a promising treatment option for patients in the near future.
Many of the preclinical strategies being explored for the treatment of one form of Batten disease could have applications across multiple subtypes of Batten disease and even other lysosomal storage disorders.
Investigators now recognize that a single treatment might not be sufficient to halt disease progression and are exploring combinatorial approaches to tackle multiple aspects of Batten disease progression.
A battery of new preclinical and clinical tools have been developed that facilitate effective therapy development in Batten disease, enabling an unprecedented acceleration in drug discovery for these fatal disorders.
Acknowledgements
T.B.J., J.T.C., K.A.W. and J.M.W. are supported in part by funding to J.M.W. from the Charlotte and Gwenyth Gray Foundation, the Haley’s Heroes Foundation, the Beat Batten Foundation and the Sebastian Velona Foundation and from NIH R01NS082283.
Footnotes
Competing interests
The authors declare no competing interests.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Reviewer information
Nature Reviews Neurology thanks J. Mink and other anonymous reviewer(s) for their contribution to the peer review of this work.
References
- 1.Zeman W & Dyken P Neuronal ceroid-lipofuscinosis (Batten’s disease): relationship to amaurotic family idiocy? Pediatrics 44, 570–583 (1969). [PubMed] [Google Scholar]
- 2.Aldrich A & Kielian T Central nervous system fibrosis is associated with fibrocyte-like infiltrates. Am. J. Pathol 179, 2952–2962 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rider JA & Rider DL Batten disease: past, present, and future. Am. J. Med. Genet. Suppl 5, 21–26 (1988). [DOI] [PubMed] [Google Scholar]
- 4.Santavuori P Neuronal ceroid-lipofuscinoses in childhood. Brain Dev 10, 80–83 (1988). [DOI] [PubMed] [Google Scholar]
- 5.Jalanko A & Braulke T Neuronal ceroid lipofuscinoses. Biochim. Biophys. Acta 1793, 697–709 (2009). [DOI] [PubMed] [Google Scholar]
- 6.Stengel OC Beretning om et maerkeligt Sygdomstilfaelde hos fire Sødskende I Nærheden af Röraas. Eyr 1, 347–352 (1826). [Google Scholar]
- 7.Batten FE Cerebral degeneration with symmetrical changes in the maculae in two members of a family. Trans. Ophthalmol. Soc. UK 23, 386–390 (1903). [Google Scholar]
- 8.Adams HR & Mink JW, University of Rochester Batten Center Study Group. Neurobehavioral features and natural history of juvenile neuronal ceroid lipofuscinosis (Batten disease). J. Child Neurol 28, 1128–1136 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Haltia M The neuronal ceroid-lipofuscinoses: from past to present. Biochim. Biophys. Acta 1762, 850–856 (2006). [DOI] [PubMed] [Google Scholar]
- 10.Goebel HH, Zeman W & Pilz H Ultrastructural investigations of peripheral nerves in neuronal ceroid-lipofuscinoses (NCL). J. Neurol 213, 295–303 (1976). [DOI] [PubMed] [Google Scholar]
- 11.Haltia M, Rapola J & Santavuori P Infantile type of so-called neuronal ceroid-lipofuscinosis. Histological and electron microscopic studies. Acta Neuropathol 26, 157–170 (1973). [DOI] [PubMed] [Google Scholar]
- 12.Anderson GW, Goebel HH & Simonati A Human pathology in NCL. Biochim. Biophys. Acta 1832, 1807–1826 (2013). [DOI] [PubMed] [Google Scholar]
- 13.Goebel HH Fingerprint inclusions in non-vacuolated lymphocytes in juvenile neuronal ceroid-lipofuscinosis. Clin. Neuropathol 4, 210–213 (1985). [PubMed] [Google Scholar]
- 14.Nijssen PC et al. Autosomal dominant adult neuronal ceroid lipofuscinosis: a novel form of NCL with granular osmiophilic deposits without palmitoyl protein thioesterase 1 deficiency. Brain Pathol 13, 574–581 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Siintola E et al. Cathepsin D deficiency underlies congenital human neuronal ceroid-lipofuscinosis. Brain 129, 1438–1445 (2006). [DOI] [PubMed] [Google Scholar]
- 16.Sandbank U Congenital amaurotic idiocy. Pathol. Eur 3, 226–229 (1968). [PubMed] [Google Scholar]
- 17.Humphreys S, Lake BD & Scholtz CL Congenital amaurotic idiocy — a pathological, histochemical, biochemical and ultrastructural study. Neuropathol. Appl. Neurobiol 11, 475–484 (1985). [DOI] [PubMed] [Google Scholar]
- 18.Barohn RJ, Dowd DC & Kagan-Hallet KS Congenital ceroid-lipofuscinosis. Pediatr. Neurol 8, 54–59 (1992). [DOI] [PubMed] [Google Scholar]
- 19.Meyer S et al. Congenital CLN disease in two siblings. Wien. Med. Wochenschr 165, 210–213 (2015). [DOI] [PubMed] [Google Scholar]
- 20.Norman RM & Wood N A congenital form of amaurotic family idiocy. J. Neurol. Psychiatry 4, 175–190 (1941). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Garborg I, Torvik A, Hals J, Tangsrud SE & Lindemann R Congenital neuronal ceroid lipofuscinosis. A case report. Acta Pathol. Microbiol. Immunol. Scand. A 95, 119–125 (1987). [DOI] [PubMed] [Google Scholar]
- 22.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 29, 48–54 (1954). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Fritchie K et al. Novel mutation and the first prenatal screening of cathepsin D deficiency (CLN10). Acta Neuropathol 117, 201–208 (2009). [DOI] [PubMed] [Google Scholar]
- 24.Goebel HH et al. Prenatal diagnosis of infantile neuronal ceroid-lipofuscinosis: a combined electron microscopic and molecular genetic approach. Brain Dev 17, 83–88 (1995). [DOI] [PubMed] [Google Scholar]
- 25.Lerner TJ et al. Isolation of a novel gene underlying Batten disease, CLN3. Cell 82, 949–957 (1995). [DOI] [PubMed] [Google Scholar]
- 26.Weimer JM, Kriscenski-Perry E, Elshatory Y & Pearce DA The neuronal ceroid lipofuscinoses: mutations in different proteins result in similar disease. Neuromolecular Med 1, 111–124 (2002). [DOI] [PubMed] [Google Scholar]
- 27.Williams RE & Mole SE New nomenclature and classification scheme for the neuronal ceroid lipofuscinoses. Neurology 79, 183–191 (2012). [DOI] [PubMed] [Google Scholar]
- 28.Kohan R et al. An integrated strategy for the diagnosis of neuronal ceroid lipofuscinosis types 1 (CLN1) and 2 (CLN2) in eleven Latin American patients. Clin. Genet 76, 372–382 (2009). [DOI] [PubMed] [Google Scholar]
- 29.Kohan R et al. The neuronal ceroid lipofuscinoses program: a translational research experience in Argentina. Biochim. Biophys. Acta 1852, 2301–2311 (2015). [DOI] [PubMed] [Google Scholar]
- 30.Steinfeld R et al. Late infantile neuronal ceroid lipofuscinosis: quantitative description of the clinical course in patients with CLN2 mutations. Am. J. Med. Genet 112, 347–354 (2002). [DOI] [PubMed] [Google Scholar]
- 31.de Blieck EA et al. Methodology of clinical research in rare diseases: development of a research program in juvenile neuronal ceroid lipofuscinosis (JNCL) via creation of a patient registry and collaboration with patient advocates. Contemp. Clin. Trials 35, 48–54 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mink JW, Augustine EF, Adams HR, Marshall FJ & Kwon JM Classification and natural history of the neuronal ceroid lipofuscinoses. J. Child Neurol 28, 1101–1105 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Nickel M et al. Disease characteristics and progression in patients with late-infantile neuronal ceroid lipofuscinosis type 2 (CLN2) disease: an observational cohort study. Lancet Child Adolesc. Health 2, 582–590 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Simonati A et al. Phenotype and natural history of variant late infantile ceroid-lipofuscinosis 5. Dev. Med. Child Neurol 59, 815–821 (2017). [DOI] [PubMed] [Google Scholar]
- 35.Dolisca SB, Mehta M, Pearce DA, Mink JW & Maria BL Batten disease: clinical aspects, molecular mechanisms, translational science, and future directions. J. Child Neurol 28, 1074–1100 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Goebel HH & Wisniewski KE Current state of clinical and morphological features in human NCL. Brain Pathol 14, 61–69 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Williams RE et al. Diagnosis of the neuronal ceroid lipofuscinoses: an update. Biochim. Biophys. Acta 1762, 865–872 (2006). [DOI] [PubMed] [Google Scholar]
- 38.Radke J, Stenzel W & Goebel HH Human NCL neuropathology. Biochim. Biophys. Acta 1852, 2262–2266 (2015). [DOI] [PubMed] [Google Scholar]
- 39.Vesa J et al. Mutations in the palmitoyl protein thioesterase gene causing infantile neuronal ceroid lipofuscinosis. Nature 376, 584–587 (1995). [DOI] [PubMed] [Google Scholar]
- 40.Greaves J & Chamberlain LH Palmitoylation-dependent protein sorting. J. Cell Biol 176, 249–254 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kakela R, Somerharju P & Tyynela J Analysis of phospholipid molecular species in brains from patients with infantile and juvenile neuronal-ceroid lipofuscinosis using liquid chromatography-electrospray ionization mass spectrometry. J. Neurochem 84, 1051–1065 (2003). [DOI] [PubMed] [Google Scholar]
- 42.Lyly A et al. Deficiency of the INCL protein Ppt1 results in changes in ectopic F1-ATP synthase and altered cholesterol metabolism. Hum. Mol. Genet 17, 1406–1417 (2008). [DOI] [PubMed] [Google Scholar]
- 43.Ahtiainen L et al. Palmitoyl protein thioesterase 1 (PPT1) deficiency causes endocytic defects connected to abnormal saposin processing. Exp. Cell Res 312, 1540–1553 (2006). [DOI] [PubMed] [Google Scholar]
- 44.Kielar C et al. Molecular correlates of axonal and synaptic pathology in mouse models of Batten disease. Hum. Mol. Genet 18, 4066–4080 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kim SJ et al. Palmitoyl protein thioesterase-1 deficiency impairs synaptic vesicle recycling at nerve terminals, contributing to neuropathology in humans and mice. J. Clin. Invest 118, 3075–3086 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Das AK et al. Molecular genetics of palmitoyl-protein thioesterase deficiency in the US. J. Clin. Invest 102, 361–370 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Das AK, Lu JY & Hofmann SL Biochemical analysis of mutations in palmitoyl-protein thioesterase causing infantile and late-onset forms of neuronal ceroid lipofuscinosis. Hum. Mol. Genet 10, 1431–1439 (2001). [DOI] [PubMed] [Google Scholar]
- 48.Kohan R et al. Neuronal ceroid lipofuscinosis type CLN2: a new rationale for the construction of phenotypic subgroups based on a survey of 25 cases in South America. Gene 516, 114–121 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kay GW, Verbeek MM, Furlong JM, Willemsen MAAP & Palmer DN Neuropeptide changes and neuroactive amino acids in CSF from humans and sheep with neuronal ceroid lipofuscinoses (NCLs, Batten disease). Neurochem. Int 55, 783–788 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Breedveld GJ et al. A new locus for a childhood onset, slowly progressive autosomal recessive spinocerebellar ataxia maps to chromosome 11p15. J. Med. Genet 41, 858–866 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sun Y et al. Autosomal recessive spinocerebellar ataxia 7 (SCAR7) is caused by variants in TPP1, the gene involved in classic late-infantile neuronal ceroid lipofuscinosis 2 disease (CLN2 disease). Hum. Mutat 34, 706–713 (2013). [DOI] [PubMed] [Google Scholar]
- 52.Mole SE, Williams RE & Goebel HH Correlations between genotype, ultrastructural morphology and clinical phenotype in the neuronal ceroid lipofuscinoses. Neurogenetics 6, 107–126 (2005). [DOI] [PubMed] [Google Scholar]
- 53.Ku CA et al. Detailed clinical phenotype and molecular genetic findings in CLN3-associated isolated retinal degeneration. JAMA Ophthalmol 135, 749–760 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Santavuori P, Rapola J, Sainio K & Raitta C A variant of Jansky-Bielschowsky disease. Neuropediatrics 13, 135–141 (1982). [DOI] [PubMed] [Google Scholar]
- 55.Santavuori P et al. The spectrum of Jansky-Bielschowsky disease. Neuropediatrics 22, 92–96 (1991). [DOI] [PubMed] [Google Scholar]
- 56.Xin W et al. CLN5 mutations are frequent in juvenile and late-onset non-Finnish patients with NCL. Neurology 74, 565–571 (2010). [DOI] [PubMed] [Google Scholar]
- 57.Pineda-Trujillo N et al. A CLN5 mutation causing an atypical neuronal ceroid lipofuscinosis of juvenile onset. Neurology 64, 740–742 (2005). [DOI] [PubMed] [Google Scholar]
- 58.Mancini C et al. Adult-onset autosomal recessive ataxia associated with neuronal ceroid lipofuscinosis type 5 gene (CLN5) mutations. J. Neurol 262, 173–178 (2015). [DOI] [PubMed] [Google Scholar]
- 59.Haines JL et al. Chromosomal localization of two genes underlying late-infantile neuronal ceroid lipofuscinosis. Neurogenetics 1, 217–222 (1998). [DOI] [PubMed] [Google Scholar]
- 60.Berkovic SF, Carpenter S, Andermann F, Andermann E & Wolfe LS Kufs’ disease: a critical reappraisal. Brain 111, 27–62 (1988). [DOI] [PubMed] [Google Scholar]
- 61.Fietz M et al. Diagnosis of neuronal ceroid lipofuscinosis type 2 (CLN2 disease): expert recommendations for early detection and laboratory diagnosis. Mol. Genet. Metab 119, 160–167 (2016). [DOI] [PubMed] [Google Scholar]
- 62.Lake BD, Young EP & Winchester BG Prenatal diagnosis of lysosomal storage diseases. Brain Pathol 8, 133–149 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Munroe PB et al. Prenatal diagnosis of Batten’s disease. Lancet 347, 1014–1015 (1996). [DOI] [PubMed] [Google Scholar]
- 64.Chow CW, Borg J, Billson VR & Lake BD Fetal tissue involvement in the late infantile type of neuronal ceroid lipofuscinosis. Prenat. Diagn 13, 833–841 (1993). [DOI] [PubMed] [Google Scholar]
- 65.Rapola J, Salonen R, Ammala P & Santavuori P Prenatal diagnosis of infantile neuronal ceroid-lipofuscinosis, INCL: morphological aspects. J. Inherit. Metab. Dis 16, 349–352 (1993). [DOI] [PubMed] [Google Scholar]
- 66.Martin JJ & de Groote C Involvement of the skin in late infantile and juvenile amaurotic idiocies (neuronal ceroid-lipofuscinoses). Pathol. Eur 9, 263–272 (1974). [PubMed] [Google Scholar]
- 67.Ceuterick C & Martin JJ Diagnostic role of skin or conjunctival biopsies in neurological disorders. An update. J. Neurol. Sci 65, 179–191 (1984). [DOI] [PubMed] [Google Scholar]
- 68.Brett EM & Lake BD Reassessment of rectal approach to neuropathology in childhood: review of 307 biopsies over 11 years. Arch. Dis. Child 50, 753–762 (1975). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Rapola J, Santavuori P & Savilahti E Suction biopsy of rectal mucosa in the diagnosis of infantile and juvenile types of neuronal ceroid lipofuscinoses. Hum. Pathol 15, 352–360 (1984). [DOI] [PubMed] [Google Scholar]
- 70.Ceuterick-de Groote C & Martin JJ Extracerebral biopsy in lysosomal and peroxisomal disorders. Ultrastructural findings. Brain Pathol 8, 121–132 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Nita DA, Mole SE & Minassian BA Neuronal ceroid lipofuscinoses. Epilept. Disord 18, 73–88 (2016). [DOI] [PubMed] [Google Scholar]
- 72.Anderson G, Smith VV, Malone M & Sebire NJ Blood film examination for vacuolated lymphocytes in the diagnosis of metabolic disorders; retrospective experience of more than 2,500 cases from a single centre. J. Clin. Pathol 58, 1305–1310 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Goebel HH The neuronal ceroid-lipofuscinoses. J. Child Neurol 10, 424–437 (1995). [DOI] [PubMed] [Google Scholar]
- 74.Mole SE & Williams RE Neuronal ceroid-lipofuscinoses. GeneReviews https://www.ncbi.nlm.nih.gov/books/NBK1428 (updated 1 Aug 2013).
- 75.Junaid MA, Sklower Brooks S, Wisniewski KE & Pullarkat RK A novel assay for lysosomal pepstatin-insensitive proteinase and its application for the diagnosis of late-infantile neuronal ceroid lipofuscinosis. Clin. Chim. Acta 281, 169–176 (1999). [DOI] [PubMed] [Google Scholar]
- 76.van Diggelen OP et al. A rapid fluorogenic palmitoyl-protein thioesterase assay: pre- and postnatal diagnosis of INCL. Mol. Genet. Metab 66, 240–244 (1999). [DOI] [PubMed] [Google Scholar]
- 77.Voznyi YV et al. A new simple enzyme assay for pre- and postnatal diagnosis of infantile neuronal ceroid lipofuscinosis (INCL) and its variants. J. Med. Genet 36, 471–474 (1999). [PMC free article] [PubMed] [Google Scholar]
- 78.Vines DJ & Warburton MJ Classical late infantile neuronal ceroid lipofuscinosis fibroblasts are deficient in lysosomal tripeptidyl peptidase I. FEBS Lett 443, 131–135 (1999). [DOI] [PubMed] [Google Scholar]
- 79.Sohar I, Lin L & Lobel P Enzyme-based diagnosis of classical late infantile neuronal ceroid lipofuscinosis: comparison of tripeptidyl peptidase I and pepstatin-insensitive protease assays. Clin. Chem 46, 1005–1008 (2000). [PubMed] [Google Scholar]
- 80.Partanen S et al. A replacement of the active-site aspartic acid residue 293 in mouse cathepsin D affects its intracellular stability, processing and transport in HEK-293 cells. Biochem. J 369, 55–62 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Mole SE & Cotman SL Genetics of the neuronal ceroid lipofuscinoses (Batten disease). Biochim. Biophys. Acta 1852, 2237–2241 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Patiño LC et al. Exome sequencing is an efficient tool for variant late-infantile neuronal ceroid lipofuscinosis molecular diagnosis. PLOS ONE 9, e109576 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Braak H & Goebel HH Loss of pigment-laden stellate cells: a severe alteration of the isocortex in juvenile neuronal ceroid-lipofuscinosis. Acta Neuropathol 42, 53–57 (1978). [DOI] [PubMed] [Google Scholar]
- 84.Braak H & Goebel HH Pigmentoarchitectonic pathology of the isocortex in juvenile neuronal ceroid-lipofuscinosis: axonal enlargements in layer IIIab and cell loss in layer V. Acta Neuropathol 46, 79–83 (1979). [DOI] [PubMed] [Google Scholar]
- 85.Haltia M The neuronal ceroid-lipofuscinoses. J. Neuropathol. Exp. Neurol 62, 1–13 (2003). [DOI] [PubMed] [Google Scholar]
- 86.Haltia M, Herva R, Suopanki J, Baumann M & Tyynela J Hippocampal lesions in the neuronal ceroid lipofuscinoses. Eur. J. Paediatr. Neurol 5 (Suppl. A), 209–211 (2001). [DOI] [PubMed] [Google Scholar]
- 87.Tyynela J, Cooper JD, Khan MN, Shemilts SJ & Haltia M Hippocampal pathology in the human neuronal ceroid-lipofuscinoses: distinct patterns of storage deposition, neurodegeneration and glial activation. Brain Pathol 14, 349–357 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Tyynela J, Suopanki J, Santavuori P, Baumann M & Haltia M Variant late infantile neuronal ceroid-lipofuscinosis: pathology and biochemistry. J. Neuropathol. Exp. Neurol 56, 369–375 (1997). [DOI] [PubMed] [Google Scholar]
- 89.Collins J, Holder GE, Herbert H & Adams GG Batten disease: features to facilitate early diagnosis. Br. J. Ophthalmol 90, 1119–1124 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Spalton DJ, Taylor DS & Sanders MD Juvenile Batten’s disease: an ophthalmological assessment of 26 patients. Br. J. Ophthalmol 64, 726–732 (1980). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Birch DG Retinal degeneration in retinitis pigmentosa and neuronal ceroid lipofuscinosis: an overview. Mol. Genet. Metab 66, 356–366 (1999). [DOI] [PubMed] [Google Scholar]
- 92.Elleder M & Tyynela J Incidence of neuronal perikaryal spheroids in neuronal ceroid lipofuscinoses (Batten disease). Clin. Neuropathol 17, 184–189 (1998). [PubMed] [Google Scholar]
- 93.Carcel-Trullols J, Kovacs AD & Pearce DA Cell biology of the NCL proteins: what they do and don’t do. Biochim. Biophys. Acta 1852, 2242–2255 (2015). [DOI] [PubMed] [Google Scholar]
- 94.Bond M, Holthaus SM, Tammen I, Tear G & Russell C Use of model organisms for the study of neuronal ceroid lipofuscinosis. Biochim. Biophys. Acta 1832, 1842–1865 (2013). [DOI] [PubMed] [Google Scholar]
- 95.Courtine G et al. Can experiments in nonhuman primates expedite the translation of treatments for spinal cord injury in humans? Nat. Med 13, 561–566 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.van der Worp HB et al. Can animal models of disease reliably inform human studies? PLOS Med 7, e1000245 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.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 8, 351–361 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Shacka JJ Mouse models of neuronal ceroid lipofuscinoses: useful pre-clinical tools to delineate disease pathophysiology and validate therapeutics. Brain Res. Bull 88, 43–57 (2012). [DOI] [PubMed] [Google Scholar]
- 99.Augustine EF et al. Short-term administration of mycophenolate is well-tolerated in CLN3 disease (juvenile neuronal ceroid lipofuscinosis). JIMD Rep 10.1007/8904_2018_113 (2018). [DOI] [PMC free article] [PubMed]
- 100.Levin SW et al. Oral cysteamine bitartrate and N-acetylcysteine for patients with infantile neuronal ceroid lipofuscinosis: a pilot study. Lancet Neurol 13, 777–787 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Selden NR et al. Central nervous system stem cell transplantation for children with neuronal ceroid lipofuscinosis. J. Neurosurg. Pediatr 11, 643–652 (2013). [DOI] [PubMed] [Google Scholar]
- 102.Weber K & Pearce DA Large animal models for Batten disease: a review. J. Child Neurol 28, 1123–1127 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Beraldi R, Mdaki K, Kovacs AD & Pearce DA Generation of a Juvenile Batten disease porcine model [abstract O15]. Presented at the 15th International Conference of Neuronal Ceroid Lipofuscinosis (Batten disease) in Boston, MA, USA (2016). [Google Scholar]
- 104.McBride JL et al. Discovery of a CLN7 model of Batten disease in non-human primates. Neurobiol. Dis 119, 65–78 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Katz ML et al. A mutation in the CLN8 gene in English Setter dogs with neuronal ceroid-lipofuscinosis. Biochem. Biophys. Res. Commun 327, 541–547 (2005). [DOI] [PubMed] [Google Scholar]
- 106.Melville SA et al. A mutation in canine CLN5 causes neuronal ceroid lipofuscinosis in Border collie dogs. Genomics 86, 287–294 (2005). [DOI] [PubMed] [Google Scholar]
- 107.Tammen I et al. A missense mutation (c.184C>T) in ovine CLN6 causes neuronal ceroid lipofuscinosis in Merino sheep whereas affected South Hampshire sheep have reduced levels of CLN6 mRNA. Biochim. Biophys. Acta 1762, 898–905 (2006). [DOI] [PubMed] [Google Scholar]
- 108.Tyynela J et al. Congenital ovine neuronal ceroid lipofuscinosis — a cathepsin D deficiency with increased levels of the inactive enzyme. Eur. J. Paediatr. Neurol 5 (Suppl. A), 43–45 (2001). [DOI] [PubMed] [Google Scholar]
- 109.Wei X et al. Initial experience with a juvenile sheep model for evaluation of the pediatric intracorporeal ventricular assist devices [corrected]. ASAIO J 59, 75–80 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Sorby-Adams AJ, Vink R & Turner RJ Large animal models of stroke and traumatic brain injury as translational tools. Am. J. Physiol. Regul. Integr. Comp. Physiol 315, R165–R190 (2018). [DOI] [PubMed] [Google Scholar]
- 111.Swindle MM, Makin A, Herron AJ, Clubb FJ Jr & Frazier KS Swine as models in biomedical research and toxicology testing. Vet. Pathol 49, 344–356 (2012). [DOI] [PubMed] [Google Scholar]
- 112.Phillips KA et al. Why primate models matter. Am. J. Primatol 76, 801–827 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Jelsing J et al. The postnatal development of neocortical neurons and glial cells in the Gottingen minipig and the domestic pig brain. J. Exp. Biol 209, 1454–1462 (2006). [DOI] [PubMed] [Google Scholar]
- 114.Pond WG et al. Perinatal ontogeny of brain growth in the domestic pig. Proc. Soc. Exp. Biol. Med 223, 102–108 (2000). [DOI] [PubMed] [Google Scholar]
- 115.Beraldi R et al. A novel porcine model of ataxia telangiectasia reproduces neurological features and motor deficits of human disease. Hum. Mol. Genet 24, 6473–6484 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Welsh MJ, Rogers CS, Stoltz DA, Meyerholz DK & Prather RS Development of a porcine model of cystic fibrosis. Trans. Am. Clin. Climatol. Assoc 120, 149–162 (2009). [PMC free article] [PubMed] [Google Scholar]
- 117.Stoltz DA et al. Cystic fibrosis pigs develop lung disease and exhibit defective bacterial eradication at birth. Sci. Transl Med 2, 29ra31 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.White KA et al. A porcine model of neurofibromatosis type 1 (NF1) that mimics the human disease. JCI Insight 3, 120402 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Chattopadhyay S et al. An autoantibody inhibitory to glutamic acid decarboxylase in the neurodegenerative disorder Batten disease. Hum. Mol. Genet 11, 1421–1431 (2002). [DOI] [PubMed] [Google Scholar]
- 120.Andrews WJ, Magee AG, Gardiner PV, Fleming I & Morris TC Paroxysmal nocturnal haemoglobinuria and diabetes mellitus. Ulster Med. J 59, 84–86 (1990). [PMC free article] [PubMed] [Google Scholar]
- 121.Hu J et al. 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 107, 213–221 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Lu JY, Hu J & Hofmann SL Human recombinant palmitoyl-protein thioesterase-1 (PPT1) for preclinical evaluation of enzyme replacement therapy for infantile neuronal ceroid lipofuscinosis. Mol. Genet. Metab 99, 374–378 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Lu JY et al. Intrathecal enzyme replacement therapy improves motor function and survival in a preclinical mouse model of infantile neuronal ceroid lipofuscinosis. Mol. Genet. Metab 116, 98–105 (2015). [DOI] [PubMed] [Google Scholar]
- 124.Chattopadhyay S, Kriscenski-Perry E, Wenger DA & Pearce DA An autoantibody to GAD65 in sera of patients with juvenile neuronal ceroid lipofuscinoses. Neurology 59, 1816–1817 (2002). [DOI] [PubMed] [Google Scholar]
- 125.Chang M et al. Intraventricular enzyme replacement improves disease phenotypes in a mouse model of late infantile neuronal ceroid lipofuscinosis. Mol. Ther 16, 649–656 (2008). [DOI] [PubMed] [Google Scholar]
- 126.Young PP, Fantz CR & Sands MS VEGF disrupts the neonatal blood-brain barrier and increases life span after non-ablative BMT in a murine model of congenital neurodegeneration caused by a lysosomal enzyme deficiency. Exp. Neurol 188, 104–114 (2004). [DOI] [PubMed] [Google Scholar]
- 127.Neuwelt EA et al. Delivery of hexosaminidase A to the cerebrum after osmotic modification of the blood—brain barrier. Proc. Natl Acad. Sci. USA 78, 5838–5841 (1981). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Saraiva C et al. Nanoparticle-mediated brain drug delivery: Overcoming blood-brain barrier to treat neurodegenerative diseases. J. Control. Release 235, 34–47 (2016). [DOI] [PubMed] [Google Scholar]
- 129.da Fonseca AC et al. The impact of microglial activation on blood-brain barrier in brain diseases. Front. Cell. Neurosci 8, 362 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Rite I, Machado A, Cano J & Venero JL Blood-brain barrier disruption induces in vivo degeneration of nigral dopaminergic neurons. J. Neurochem 101, 1567–1582 (2007). [DOI] [PubMed] [Google Scholar]
- 131.Vuillemenot BR et al. Nonclinical evaluation of CNS-administered TPP1 enzyme replacement in canine CLN2 neuronal ceroid lipofuscinosis. Mol. Genet. Metab 114, 281–293 (2015). [DOI] [PubMed] [Google Scholar]
- 132.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT01907087 (2018). [DOI] [PubMed]
- 133.Schulz A et al. Study of intraventricular cerliponase alfa for CLN2 disease. N. Engl. J. Med 378, 1898–1907 (2018). [DOI] [PubMed] [Google Scholar]
- 134.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT02485899 (2018). [DOI] [PubMed]
- 135.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT02678689 (2018). [DOI] [PubMed]
- 136.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT02963350 (2017). [DOI] [PubMed]
- 137.Worgall S et al. Neurological deterioration in late infantile neuronal ceroid lipofuscinosis. Neurology 69, 521 (2007).17679671 [Google Scholar]
- 138.Lobel U et al. Volumetric description of brain atrophy in neuronal ceroid lipofuscinosis 2: supratentorial gray matter shows uniform disease progression. AJNR Am. J. Neuroradiol 37, 1938–1943 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT00976352 (2018). [DOI] [PubMed]
- 140.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT01474343 (2014). [DOI] [PubMed]
- 141.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT02053064 (2017). [DOI] [PubMed]
- 142.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT02716246 (2018). [DOI] [PubMed]
- 143.Sawamoto K, Chen HH, Almeciga-Diaz CJ, Mason RW & Tomatsu S Gene therapy for mucopolysaccharidoses. Mol. Genet. Metab 123, 59–68 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Colella P, Ronzitti G & Mingozzi F Emerging issues in AAV-mediated in vivo gene therapy. Mol. Ther. Methods Clin. Dev 8, 87–104 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Murlidharan G, Samulski RJ & Asokan A Biology of adeno-associated viral vectors in the central nervous system. Front. Mol. Neurosci 7, 76 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Griffey M et al. 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 16, 360–369 (2004). [DOI] [PubMed] [Google Scholar]
- 147.Griffey MA et al. CNS-directed AAV2-mediated gene therapy ameliorates functional deficits in a murine model of infantile neuronal ceroid lipofuscinosis. Mol. Ther 13, 538–547 (2006). [DOI] [PubMed] [Google Scholar]
- 148.Katz ML et al. AAV gene transfer delays disease onset in a TPP1-deficient canine model of the late infantile form of Batten disease. Sci. Transl Med 7, 313ra180 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Opie SR et al. Identification of amino acid residues in the capsid proteins of adeno-associated virus type 2 that contribute to heparan sulfate proteoglycan binding. J. Virol 77, 6995–7006 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Cabrera-Salazar MA et al. Timing of therapeutic intervention determines functional and survival outcomes in a mouse model of late infantile batten disease. Mol. Ther 15, 1782–1788 (2007). [DOI] [PubMed] [Google Scholar]
- 151.Sondhi D et al. Enhanced survival of the LINCL mouse following CLN2 gene transfer using the rh.10 rhesus macaque-derived adeno-associated virus vector. Mol. Ther 15, 481–491 (2007). [DOI] [PubMed] [Google Scholar]
- 152.Mietzsch M, Broecker F, Reinhardt A, Seeberger PH & Heilbronn R Differential adeno-associated virus serotype-specific interaction patterns with synthetic heparins and other glycans. J. Virol 88, 2991–3003 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Sondhi D et al. 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 25, 223–239 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Macauley SL et al. An anti-neuroinflammatory that targets dysregulated glia enhances the efficacy of CNS-directed gene therapy in murine infantile neuronal ceroid lipofuscinosis. J. Neurosci 34, 13077–13082 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Shyng C et al. Synergistic effects of treating the spinal cord and brain in CLN1 disease. Proc. Natl Acad. Sci. USA 114, E5920–E5929 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Kleine Holthaus SM et al. Prevention of photoreceptor cell loss in a Cln6nclf mouse model of Batten disease requires CLN6 gene transfer to bipolar cells. Mol. Ther 26, 1343–1353 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Cotman SL et al. Cln3(Deltaex7/8) knock-in mice with the common JNCL mutation exhibit progressive neurologic disease that begins before birth. Hum. Mol. Genet 11, 2709–2721 (2002). [DOI] [PubMed] [Google Scholar]
- 158.Bosch ME et al. Self-complementary AAV9 gene delivery partially corrects pathology associated with juvenile neuronal ceroid lipofuscinosis (CLN3). J. Neurosci 36, 9669–9682 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Cain JT et al. Testing safety and efficacy of AAV9-CLN6 gene therapy in a mouse model of CLN6-Batten disease [P11]. Presented at the 15th International Conference of Neuronal Ceroid Lipofuscinosis (Batten disease) in Boston, MA, USA (2016). [Google Scholar]
- 160.Likhite S et al. Gene therapy for the CLN6 Batten disease: in vivo validation and safety study into a non-human primate model [P48]. Presented at the 15th International Conference of Neuronal Ceroid Lipofuscinosis (Batten disease) in Boston, MA, USA (2016). [Google Scholar]
- 161.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT02725580 (2018). [DOI] [PubMed]
- 162.Johnson TB et al. Intrathecal scAAV9-CLN3 administration: potential gene therapy for CLN3-Batten disease [abstract]. Presented at the 16th International Conference on Neuronal Ceroid Lipofuscinosis (2018). [Google Scholar]
- 163.US National Library of Medicine. ClinicalTrials.govhttps://ClinicalTrials.gov/ct2/show/NCT03770572 (2018). [DOI] [PubMed]
- 164.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT00151216 (2018). [DOI] [PubMed]
- 165.Worgall S et al. Treatment of late infantile neuronal ceroid lipofuscinosis by CNS administration of a serotype 2 adeno-associated virus expressing CLN2 cDNA. Hum. Gene Ther 19, 463–474 (2008). [DOI] [PubMed] [Google Scholar]
- 166.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT01161576 (2018). [DOI] [PubMed]
- 167.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT01414985 (2018). [DOI] [PubMed]
- 168.Wiley LA et al. 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 27, 835–846 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Wenzel A, Grimm C, Samardzija M & Reme CE Molecular mechanisms of light-induced photoreceptor apoptosis and neuroprotection for retinal degeneration. Prog. Retin. Eye Res 24, 275–306 (2005). [DOI] [PubMed] [Google Scholar]
- 170.Wen R, Tao W, Li Y & Sieving PA CNTF and retina. Prog. Retin. Eye Res 31, 136–151 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Jankowiak W et al. Sustained neural stem cell-based intraocular delivery of CNTF attenuates photoreceptor loss in the nclf mouse model of neuronal ceroid lipofuscinosis. PLOS ONE 10, e0127204 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Tracy CJ et al. Intravitreal implantation of TPP1-transduced stem cells delays retinal degeneration in canine CLN2 neuronal ceroid lipofuscinosis. Exp. Eye Res 152, 77–87 (2016). [DOI] [PubMed] [Google Scholar]
- 173.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT00337636 (2015). [DOI] [PubMed]
- 174.Arvan P, Zhao X, Ramos-Castaneda J & Chang A Secretory pathway quality control operating in Golgi, plasmalemmal, and endosomal systems. Traffic 3, 771–780 (2002). [DOI] [PubMed] [Google Scholar]
- 175.Ellgaard L & Helenius A ER quality control: towards an understanding at the molecular level. Curr. Opin. Cell Biol 13, 431–437 (2001). [DOI] [PubMed] [Google Scholar]
- 176.Parenti G Treating lysosomal storage diseases with pharmacological chaperones: from concept to clinics. EMBO Mol. Med 1, 268–279 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Valenzano KJ et al. Identification and characterization of pharmacological chaperones to correct enzyme deficiencies in lysosomal storage disorders. Assay Drug Dev. Technol 9, 213–235 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Fan JQ A counterintuitive approach to treat enzyme deficiencies: use of enzyme inhibitors for restoring mutant enzyme activity. Biol. Chem 389, 1–11 (2008). [DOI] [PubMed] [Google Scholar]
- 179.Dawson G, Schroeder C & Dawson PE Palmitoyl:protein thioesterase (PPT1) inhibitors can act as pharmacological chaperones in infantile Batten disease. Biochem. Biophys. Res. Commun 395, 66–69 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.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 33, 42–63 (2012). [DOI] [PubMed] [Google Scholar]
- 181.Roy B et al. Ataluren stimulates ribosomal selection of near-cognate tRNAs to promote nonsense suppression. Proc. Natl Acad. Sci. USA 113, 12508–12513 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Sleat DE, Sohar I, Gin RM & Lobel P Aminoglycoside-mediated suppression of nonsense mutations in late infantile neuronal ceroid lipofuscinosis. Eur. J. Paediatr. Neurol 5 (Suppl. A), 57–62 (2001). [DOI] [PubMed] [Google Scholar]
- 183.Miller JN, Chan CH & Pearce DA The role of nonsense-mediated decay in neuronal ceroid lipofuscinosis. Hum. Mol. Genet 22, 2723–2734 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Schoch KM & Miller TM Antisense oligonucleotides: translation from mouse models to human neurodegenerative diseases. Neuron 94, 1056–1070 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Ramesh N & Pandey UB Autophagy Dysregulation in ALS: when protein aggregates get out of hand. Front. Mol. Neurosci 10, 263 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Uddin MS et al. Autophagy and Alzheimer’s disease: from molecular mechanisms to therapeutic implications. Front. Aging Neurosci 10, 04 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Wang B, Abraham N, Gao G & Yang Q Dysregulation of autophagy and mitochondrial function in Parkinson’s disease. Transl Neurodegener 5, 19 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Koike M et al. Participation of autophagy in storage of lysosomes in neurons from mouse models of neuronal ceroid-lipofuscinoses (Batten disease). Am. J. Pathol 167, 1713–1728 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Cao Y et al. Autophagy is disrupted in a knock-in mouse model of juvenile neuronal ceroid lipofuscinosis. J. Biol. Chem 281, 20483–20493 (2006). [DOI] [PubMed] [Google Scholar]
- 190.Cotman SL & Staropoli JF The juvenile Batten disease protein, CLN3, and its role in regulating anterograde and retrograde post-Golgi trafficking. Clin. Lipidol 7, 79–91 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Behrends C, Sowa ME, Gygi SP & Harper JW Network organization of the human autophagy system. Nature 466, 68–76 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Hong M et al. Fibrates inhibit the apoptosis of Batten disease lymphoblast cells via autophagy recovery and regulation of mitochondrial membrane potential. In Vitro Cell. Dev. Biol. Anim 52, 349–355 (2016). [DOI] [PubMed] [Google Scholar]
- 193.Combs CK, Bates P, Karlo JC & Landreth GE Regulation of beta-amyloid stimulated proinflammatory responses by peroxisome proliferator-activated receptor alpha. Neurochem. Int 39, 449–457 (2001). [DOI] [PubMed] [Google Scholar]
- 194.Deplanque D et al. Peroxisome proliferator-activated receptor-alpha activation as a mechanism of preventive neuroprotection induced by chronic fenofibrate treatment. J. Neurosci 23, 6264–6271 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.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 141, 423–435 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Ghosh A et al. Activation of peroxisome proliferator-activated receptor alpha induces lysosomal biogenesis in brain cells: implications for lysosomal storage disorders. J. Biol. Chem 290, 10309–10324 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Sardiello M et al. A gene network regulating lysosomal biogenesis and function. Science 325, 473–477 (2009). [DOI] [PubMed] [Google Scholar]
- 198.Palmieri M et al. mTORC1-independent TFEB activation via Akt inhibition promotes cellular clearance in neurodegenerative storage diseases. Nat. Commun 8, 14338 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Heras-Sandoval D, Perez-Rojas JM, Hernandez-Damian J & Pedraza-Chaverri J The role of PI3K/AKT/mTOR pathway in the modulation of autophagy and the clearance of protein aggregates in neurodegeneration. Cell Signal 26, 2694–2701 (2014). [DOI] [PubMed] [Google Scholar]
- 200.Gavin M et al. Substrate reduction therapy in four patients with milder CLN1 mutations and juvenile-onset Batten disease using cysteamine bitartrate. JIMD Rep 11, 87–92 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT00028262 (2016). [DOI] [PubMed]
- 202.Groh J et al. Immune cells perturb axons and impair neuronal survival in a mouse model of infantile neuronal ceroid lipofuscinosis. Brain 136, 1083–1101 (2013). [DOI] [PubMed] [Google Scholar]
- 203.Groh J et al. Sialoadhesin promotes neuroinflammation-related disease progression in two mouse models of CLN disease. Glia 64, 792–809 (2016). [DOI] [PubMed] [Google Scholar]
- 204.Ramirez-Montealegre D et al. Autoimmunity to glutamic acid decarboxylase in the neurodegenerative disorder Batten disease. Neurology 64, 743–745 (2005). [DOI] [PubMed] [Google Scholar]
- 205.Castaneda JA & Pearce DA Identification of alpha-fetoprotein as an autoantigen in juvenile Batten disease. Neurobiol. Dis 29, 92–102 (2008). [DOI] [PubMed] [Google Scholar]
- 206.De Virgilio A et al. Parkinson’s disease: autoimmunity and neuroinflammation. Autoimmun. Rev 15, 1005–1011 (2016). [DOI] [PubMed] [Google Scholar]
- 207.Khalid SI, Ampie L, Kelly R, Ladha SS & Dardis C Immune modulation in the treatment of amyotrophic lateral sclerosis: a review of clinical trials. Front. Neurol 8, 486 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.McGeer PL, Rogers J, McGeer & Inflammation, E. G. Antiinflammatory agents, and Alzheimer’s disease: the last 22 years. J. Alzheimers Dis 54, 853–857 (2016). [DOI] [PubMed] [Google Scholar]
- 209.Spangenberg EE & Green KN Inflammation in Alzheimer’s disease: lessons learned from microglia-depletion models. Brain Behav. Immun 61, 1–11 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Dagher NN et al. Colony-stimulating factor 1 receptor inhibition prevents microglial plaque association and improves cognition in 3xTg-AD mice. J. Neuroinflamm 12, 139 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Seehafer SS et al. Immunosuppression alters disease severity in juvenile Batten disease mice. J. Neuroimmunol 230, 169–172 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.US National Library of Medicine. ClinicalTrials.govhttps://clinicaltrials.gov/show/NCT01399047 (2017). [DOI] [PubMed]
- 213.Brinkmann V et al. Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat. Rev. Drug Discov 9, 883–897 (2010). [DOI] [PubMed] [Google Scholar]
- 214.Chun J & Brinkmann V A mechanistically novel, first oral therapy for multiple sclerosis: the development of fingolimod (FTY720, Gilenya). Discov. Med 12, 213–228 (2011). [PMC free article] [PubMed] [Google Scholar]
- 215.Melzer N & Meuth SG Disease-modifying therapy in multiple sclerosis and chronic inflammatory demyelinating polyradiculoneuropathy: common and divergent current and future strategies. Clin. Exp. Immunol 175, 359–372 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Groh J, Berve K & Martini R Fingolimod and teriflunomide attenuate neurodegeneration in mouse models of neuronal ceroid lipofuscinosis. Mol. Ther 25, 1889–1899 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Aberg L et al. Intermittent prednisolone and autoantibodies to GAD65 in juvenile neuronal ceroid lipofuscinosis. Neurology 70, 1218–1220 (2008). [DOI] [PubMed] [Google Scholar]
- 218.Ahmad I et al. Allopregnanolone treatment, both as a single injection or repetitively, delays demyelination and enhances survival of Niemann-Pick C mice. J. Neurosci. Res 82, 811–821 (2005). [DOI] [PubMed] [Google Scholar]
- 219.Mellon SH, Gong W & Schonemann MD Endogenous and synthetic neurosteroids in treatment of Niemann-Pick Type C disease. Brain Res. Rev 57, 410–420 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Reeves EKM, Hoffman EP, Nagaraju K, Damsker JM & McCall JM VBP15: preclinical characterization of a novel anti-inflammatory delta 9,11 steroid. Bioorg. Med. Chem 21, 2241–2249 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Aldrich A et al. Efficacy of phosphodiesterase-4 inhibitors in juvenile Batten disease (CLN3). Ann. Neurol 80, 909–923 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Amor S et al. Inflammation in neurodegenerative diseases—an update. Immunology 142, 151–166 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Osuna-Zazuetal MA, Ponce-Gomez JA & Perez-Neri I Neuroprotective mechanisms of cannabinoids in brain ischemia and neurodegenerative disorders [Spanish]. Invest. Clin 56, 188–200 (2015). [PubMed] [Google Scholar]
- 224.Nguyen L et al. Role of sigma-1 receptors in neurodegenerative diseases. J. Pharmacol. Sci 127, 17–29 (2015). [DOI] [PubMed] [Google Scholar]
- 225.Dong XX, Wang Y & Qin ZH Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol. Sin 30, 379–387 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Benitez-King G, Ramirez-Rodriguez G, Ortiz L & Meza I The neuronal cytoskeleton as a potential therapeutical target in neurodegenerative diseases and schizophrenia. Curr. Drug Targets CNS Neurol. Disord 3, 515–533 (2004). [DOI] [PubMed] [Google Scholar]
- 227.Kim GH, Kim JE, Rhie SJ & Yoon S The role of oxidative stress in neurodegenerative diseases. Exp. Neurobiol 24, 325–340 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Karuppagounder SS et al. The c-Abl inhibitor, nilotinib, protects dopaminergic neurons in a preclinical animal model of Parkinson’s disease. Sci. Rep 4, 4874 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Zhang S, Tang MB, Luo HY, Shi CH & Xu YM Necroptosis in neurodegenerative diseases: a potential therapeutic target. Cell Death Dis 8, e2905 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Kovacs AD et al. Temporary inhibition of AMPA receptors induces a prolonged improvement of motor performance in a mouse model of juvenile Batten disease. Neuropharmacology 60, 405–409 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Kovacs AD & Pearce DA Attenuation of AMPA receptor activity improves motor skills in a mouse model of juvenile Batten disease. Exp. Neurol 209, 288–291 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Sarkar C et al. Neuroprotection and lifespan extension in Ppt1(−/−) mice by NtBuHA: therapeutic implications for INCL. Nat. Neurosci 16, 1608–1617 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Dhar S et al. Flupirtine blocks apoptosis in batten patient lymphoblasts and in human postmitotic CLN3-and CLN2-deficient neurons. Ann. Neurol 51, 448–466 (2002). [DOI] [PubMed] [Google Scholar]
- 234.Cooper J et al. Testing combinatorial therapies for juvenile Batten disease. Mol. Genet. Metab 123, S33 (2018). [Google Scholar]
- 235.Macauley SL et al. Synergistic effects of central nervous system-directed gene therapy and bone marrow transplantation in the murine model of infantile neuronal ceroid lipofuscinosis. Ann. Neurol 71, 797–804 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Agrawal N et al. Identification of combinatorial drug regimens for treatment of Huntington’s disease using Drosophila. Proc. Natl Acad. Sci. USA 102, 3777–3781 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Augustine EF, Adams HR & Mink JW Clinical trials in rare disease: challenges and opportunities. J. Child Neurol 28, 1142–1150 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Timm D et al. Searching for novel biomarkers using a mouse model of CLN3-Batten disease. PLOS ONE 13, e0201470 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Hersrud SL, Geraets RD, Weber KL, Chan CH & Pearce DA Plasma biomarkers for neuronal ceroid lipofuscinosis. FEBS J 283, 459–471 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Stoller JK The challenge of rare diseases. Chest 153, 1309–1314 (2018). [DOI] [PubMed] [Google Scholar]
- 241.US Department of Health & Human Services. Developing products for rare diseases & conditions FDA.gov https://www.fda.gov/ForIndustry/ucm2005525.htm (updated 10 May 2018).
- 242.Cialone J et al. Quantitative telemedicine ratings in Batten disease: implications for rare disease research. Neurology 77, 1808–1811 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Marshall FJ et al. A clinical rating scale for Batten disease: reliable and relevant for clinical trials. Neurology 65, 275–279 (2005). [DOI] [PubMed] [Google Scholar]
- 244.Schulz A et al. The DEM-CHILD NCL patient database: a tool for the evaluation of therapies in neuronal ceroid lipofuscinoses (NCL). Eur. J. Paediatr. Neurol 19, S16 (2015). [Google Scholar]
- 245.Sanford Research. Welcome to the Coordination of Rare Diseases at Sanford (CoRDS)! Sanford Research http://www.sanfordresearch.org/SpecialPrograms/cords/ (2018).
- 246.Vanhanen SL et al. Neuroradiological findings (MRS, MRI, SPECT) in infantile neuronal ceroid-lipofuscinosis (infantile CLN1) at different stages of the disease. Neuropediatrics 35, 27–35 (2004). [DOI] [PubMed] [Google Scholar]
- 247.Vanhanen SL, Raininko R, Autti T & Santavuori P MRI evaluation of the brain in infantile neuronal ceroid-lipofuscinosis. Part 2: MRI findings in 21 patients. J. Child Neurol 10, 444–450 (1995). [DOI] [PubMed] [Google Scholar]
- 248.Vanhanen SL, Raininko R & Santavuori P Early differential diagnosis of infantile neuronal ceroid lipofuscinosis, Rett syndrome, and Krabbe disease by CT and MR. AJNR Am. J. Neuroradiol 15, 1443–1453 (1994). [PMC free article] [PubMed] [Google Scholar]
- 249.Vanhanen SL, Raininko R, Santavuori P, Autti T & Haltia M MRI evaluation of the brain in infantile neuronal ceroid-lipofuscinosis. Part 1: postmortem MRI with histopathologic correlation. J. Child Neurol 10, 438–443 (1995). [DOI] [PubMed] [Google Scholar]
- 250.Vanhanen SL, Sainio K, Lappi M & Santavuori P EEG and evoked potentials in infantile neuronal ceroid-lipofuscinosis. Dev. Med. Child Neurol 39, 456–463 (1997). [DOI] [PubMed] [Google Scholar]
- 251.Veneselli E, Biancheri R, Buoni S & Fois A Clinical and EEG findings in 18 cases of late infantile neuronal ceroid lipofuscinosis. Brain Dev 23, 306–311 (2001). [DOI] [PubMed] [Google Scholar]
- 252.Westmoreland BF, Groover RV & Sharbrough FW Electrographic findings in three types of cerebromacular degeneration. Mayo Clin. Proc 54, 12–21 (1979). [PubMed] [Google Scholar]
- 253.Kohlschutter A, Gardiner RM & Goebel HH Human forms of neuronal ceroid-lipofuscinosis (Batten disease): consensus on diagnostic criteria, Hamburg 1992. J. Inherit. Metab. Dis 16, 241–244 (1993). [DOI] [PubMed] [Google Scholar]
- 254.Baker EH, Levin SW, Zhang Z & Mukherjee AB MRI brain volume measurements in infantile neuronal ceroid lipofuscinosis. AJNR Am. J. Neuroradiol 38, 376–382 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Santavuori P, Raininko R, Vanhanen SL, Launes J & Sainio K MRI of the brain, EEG sleep spindles and SPECT in the early diagnosis of infantile neuronal ceroid lipofuscinosis. Dev. Med. Child Neurol 34, 61–65 (1992). [DOI] [PubMed] [Google Scholar]
- 256.Seitz D et al. MR imaging and localized proton MR spectroscopy in late infantile neuronal ceroid lipofuscinosis. AJNR Am. J. Neuroradiol 19, 1373–1377 (1998). [PMC free article] [PubMed] [Google Scholar]
- 257.Dyke JP et al. Assessment of disease severity in late infantile neuronal ceroid lipofuscinosis using multiparametric MR imaging. AJNR Am. J. Neuroradiol 34, 884–889 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Dyke JP et al. Assessing disease severity in late infantile neuronal ceroid lipofuscinosis using quantitative MR diffusion-weighted imaging. AJNR Am. J. Neuroradiol 28, 1232–1236 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Williams RE et al. Management strategies for CLN2 disease. Pediatr. Neurol 69, 102–112 (2017). [DOI] [PubMed] [Google Scholar]
- 260.Autti T, Raininko R, Launes J, Nuutila A & Santavuori P Jansky-Bielschowsky variant disease: CT, MRI, and SPECT findings. Pediatr. Neurol 8, 121–126 (1992). [DOI] [PubMed] [Google Scholar]
- 261.Autti T, Raininko R, Vanhanen SL & Santavuori P Magnetic resonance techniques in neuronal ceroid lipofuscinoses and some other lysosomal diseases affecting the brain. Curr. Opin. Neurol 10, 519–524 (1997). [DOI] [PubMed] [Google Scholar]
- 262.Jarvela I et al. Clinical and magnetic resonance imaging findings in Batten disease: analysis of the major mutation (1.02-kb deletion). Ann. Neurol 42, 799–802 (1997). [DOI] [PubMed] [Google Scholar]
- 263.Eksandh LB et al. Full-field ERG in patients with Batten/Spielmeyer-Vogt disease caused by mutations in the CLN3 gene. Ophthalmic Genet 21, 69–77 (2000). [PubMed] [Google Scholar]
- 264.Santavuori P, Vanhanen SL & Autti T Clinical and neuroradiological diagnostic aspects of neuronal ceroid lipofuscinoses disorders. Eur. J. Paediatr. Neurol 5 (Suppl. A), 157–161 (2001). [DOI] [PubMed] [Google Scholar]
- 265.Cialone J et al. Females experience a more severe disease course in Batten disease. J. Inherit. Metab. Dis 35, 549–555 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Boustany RM, Alroy J & Kolodny EH Clinical classification of neuronal ceroid-lipofuscinosis subtypes. Am. J. Med. Genet. Suppl 5, 47–58 (1988). [DOI] [PubMed] [Google Scholar]
- 267.Burneo JG et al. Adult-onset neuronal ceroid lipofuscinosis (Kufs disease) with autosomal dominant inheritance in Alabama. Epilepsia 44, 841–846 (2003). [DOI] [PubMed] [Google Scholar]
- 268.Holmberg V et al. Phenotype-genotype correlation in eight patients with Finnish variant late infantile NCL (CLN5). Neurology 55, 579–581 (2000). [DOI] [PubMed] [Google Scholar]
- 269.Canafoglia L et al. Electroclinical spectrum of the neuronal ceroid lipofuscinoses associated with CLN6 mutations. Neurology 85, 316–324 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Beesley C et al. CLN8 disease caused by large genomic deletions. Mol. Genet. Genomic Med 5, 85–91 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Allen NM et al. Variant late-infantile neuronal ceroid lipofuscinosis due to a novel heterozygous CLN8 mutation and de novo 8p23.3 deletion. Clin. Genet 81, 602–604 (2012). [DOI] [PubMed] [Google Scholar]
- 272.Reinhardt K et al. Novel CLN8 mutations confirm the clinical and ethnic diversity of late infantile neuronal ceroid lipofuscinosis. Clin. Genet 77, 79–85 (2010). [DOI] [PubMed] [Google Scholar]
- 273.Doccini S et al. Early infantile neuronal ceroid lipofuscinosis (CLN10 disease) associated with a novel mutation in CTSD. J. Neurol 263, 1029–1032 (2016). [DOI] [PubMed] [Google Scholar]
- 274.Varvagiannis K et al. Congenital neuronal ceroid lipofuscinosis with a novel CTSD gene mutation: a rare cause of neonatal-onset neurodegenerative disorder. Neuropediatrics 49, 150–153 (2018). [DOI] [PubMed] [Google Scholar]
- 275.Smith KR et al. Strikingly different clinicopathological phenotypes determined by progranulin-mutation dosage. Am. J. Hum. Genet 90, 1102–1107 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Di Fabio R et al. Pseudo-dominant inheritance of a novel CTSF mutation associated with type B Kufs disease. Neurology 83, 1769–1770 (2014). [DOI] [PubMed] [Google Scholar]
- 277.Smith KR et al. Cathepsin F mutations cause Type B Kufs disease, an adult-onset neuronal ceroid lipofuscinosis. Hum. Mol. Genet 22, 1417–1423 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Gupta P et al. Disruption of PPT1 or PPT2 causes neuronal ceroid lipofuscinosis in knockout mice. Proc. Natl Acad. Sci. USA 98, 13566–13571 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Tamaki SJ et al. Neuroprotection of host cells by human central nervous system stem cells in a mouse model of infantile neuronal ceroid lipofuscinosis. Cell Stem Cell 5, 310–319 (2009). [DOI] [PubMed] [Google Scholar]
- 280.Griffey M, Macauley SL, Ogilvie JM & Sands MS AAV2-mediated ocular gene therapy for infantile neuronal ceroid lipofuscinosis. Mol. Ther 12, 413–421 (2005). [DOI] [PubMed] [Google Scholar]
- 281.Wei H et al. Disruption of adaptive energy metabolism and elevated ribosomal p-S6K1 levels contribute to INCL pathogenesis: partial rescue by resveratrol. Hum. Mol. Genet 20, 1111–1121 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Roberts MS et al. Combination small molecule PPT1 mimetic and CNS-directed gene therapy as a treatment for infantile neuronal ceroid lipofuscinosis. J. Inherit. Metab. Dis 35, 847–857 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Rozenberg AJ, Lykken E, Spratt K, Miller TJ & Gray SJ Combination dosing of CLN1 gene therapy extends lifespan in a mouse model of infantile neuronal ceroid lipofuscinosis. Mol. Genet. Metab 123, S124 (2018). [Google Scholar]
- 284.Bible E, Gupta P, Hofmann SL & Cooper JD Regional and cellular neuropathology in the palmitoyl protein thioesterase-1 null mutant mouse model of infantile neuronal ceroid lipofuscinosis. Neurobiol. Dis 16, 346–359 (2004). [DOI] [PubMed] [Google Scholar]
- 285.Kielar C et al. Successive neuron loss in the thalamus and cortex in a mouse model of infantile neuronal ceroid lipofuscinosis. Neurobiol. Dis 25, 150–162 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Macauley SL et al. Cerebellar pathology and motor deficits in the palmitoyl protein thioesterase 1-deficient mouse. Exp. Neurol 217, 124–135 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Meng Y et al. A basic ApoE-based peptide mediator to deliver proteins across the blood-brain barrier: long-term efficacy, toxicity, and mechanism. Mol. Ther 25, 1531–1543 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Miller JN, Kovács AD & Pearce DA The novel Cln1(R151X) mouse model of infantile neuronal ceroid lipofuscinosis (INCL) for testing nonsense suppression therapy. Hum. Mol. Genet 24, 185–196 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Thada V, Miller JN, Kovacs AD & Pearce DA Tissue-specific variation in nonsense mutant transcript level and drug-induced read-through efficiency in the Cln1(R151X) mouse model of INCL. J. Cell. Mol. Med 20, 381–385 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Xu S et al. Large-volume intrathecal enzyme delivery increases survival of a mouse model of late infantile neuronal ceroid lipofuscinosis. Mol. Ther 19, 1842–1848 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Passini MA et al. Intracranial delivery of CLN2 reduces brain pathology in a mouse model of classical late infantile neuronal ceroid lipofuscinosis. J. Neurosci 26, 1334–1342 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Lin L & Lobel P Production and characterization of recombinant human CLN2 protein for enzyme-replacement therapy in late infantile neuronal ceroid lipofuscinosis. Biochem. J 357, 49–55 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Geraets RD et al. A tailored mouse model of CLN2 disease: a nonsense mutant for testing personalized therapies. PLOS ONE 12, e0176526 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Katz ML et al. A mouse gene knockout model for juvenile ceroid-lipofuscinosis (Batten disease). J. Neurosci. Res 57, 551–556 (1999). [PubMed] [Google Scholar]
- 295.Pontikis CC et al. Late onset neurodegeneration in the Cln3−/− mouse model of juvenile neuronal ceroid lipofuscinosis is preceded by low level glial activation. Brain Res 1023, 231–242 (2004). [DOI] [PubMed] [Google Scholar]
- 296.Pontikis CC, Cotman SL, MacDonald ME & Cooper JD Thalamocortical neuron loss and localized astrocytosis in the Cln3Deltaex7/8 knock-in mouse model of Batten disease. Neurobiol. Dis 20, 823–836 (2005). [DOI] [PubMed] [Google Scholar]
- 297.Katz ML, Johnson GS, Tullis GE & Lei B Phenotypic characterization of a mouse model of juvenile neuronal ceroid lipofuscinosis. Neurobiol. Dis 29, 242–253 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Osorio NS et al. Neurodevelopmental delay in the Cln3Deltaex7/8 mouse model for Batten disease. Genes Brain Behav 8, 337–345 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Weimer JM et al. Cerebellar defects in a mouse model of juvenile neuronal ceroid lipofuscinosis. Brain Res 1266, 93–107 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Schultz ML et al. Modulating membrane fluidity corrects Batten disease phenotypes in vitro and in vivo. Neurobiol. Dis 115, 182–193 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Mitchison HM, Lim MJ & Cooper JD Selectivity and types of cell death in the neuronal ceroid lipofuscinoses. Brain Pathol 14, 86–96 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Weimer JM et al. Visual deficits in a mouse model of Batten disease are the result of optic nerve degeneration and loss of dorsal lateral geniculate thalamic neurons. Neurobiol. Dis 22, 284–293 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Weimer JM et al. Alterations in striatal dopamine catabolism precede loss of substantia nigra neurons in a mouse model of juvenile neuronal ceroid lipofuscinosis. Brain Res 1162, 98–112 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Sappington RM, Pearce DA & Calkins DJ Optic nerve degeneration in a murine model of juvenile ceroid lipofuscinosis. Invest. Ophthalmol. Vis. Sci 44, 3725–3731 (2003). [DOI] [PubMed] [Google Scholar]
- 305.Kovacs AD et al. Age-dependent therapeutic effect of memantine in a mouse model of juvenile Batten disease. Neuropharmacology 63, 769–775 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Kleine Holthaus SM, Smith AJ, Mole SE & Ali RR Gene therapy approaches to treat the neurodegeneration and visual failure in neuronal ceroid lipofuscinoses. Adv. Exp. Med. Biol 1074, 91–99 (2018). [DOI] [PubMed] [Google Scholar]
- 307.Rosato FE et al. Selective arterial stimulation of secretin in localization of gastrinomas. Surg. Gynecol. Obstet 171, 196–200 (1990). [PubMed] [Google Scholar]
- 308.Morgan JP et al. A murine model of variant late infantile ceroid lipofuscinosis recapitulates behavioral and pathological phenotypes of human disease. PLOS ONE 8, e78694 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309.Bronson RT, Lake BD, Cook S, Taylor S & Davisson MT Motor neuron degeneration of mice is a model of neuronal ceroid lipofuscinosis (Batten’s disease). Ann. Neurol 33, 381–385 (1993). [DOI] [PubMed] [Google Scholar]
- 310.Chang B et al. Retinal degeneration in motor neuron degeneration: a mouse model of ceroid lipofuscinosis. Invest. Ophthalmol. Vis. Sci 35, 1071–1076 (1994). [PubMed] [Google Scholar]
- 311.Messer A, Manley K & Plummer JA An early-onset congenic strain of the motor neuron degeneration (mnd) mouse. Mol. Genet. Metab 66, 393–397 (1999). [DOI] [PubMed] [Google Scholar]
- 312.Fujita K et al. Increase of glial fibrillary acidic protein fragments in the spinal cord of motor neuron degeneration mutant mouse. Brain Res 785, 31–40 (1998). [DOI] [PubMed] [Google Scholar]
- 313.Kuronen M et al. Galactolipid deficiency in the early pathogenesis of neuronal ceroid lipofuscinosis model Cln8mnd: implications to delayed myelination and oligodendrocyte maturation. Neuropathol. Appl. Neurobiol 38, 471–486 (2012). [DOI] [PubMed] [Google Scholar]
- 314.Bolivar VJ, Scott Ganus J & Messer A The development of behavioral abnormalities in the motor neuron degeneration (mnd) mouse. Brain Res 937, 74–82 (2002). [DOI] [PubMed] [Google Scholar]
- 315.Bertamini M et al. Mitochondrial oxidative metabolism in motor neuron degeneration (mnd) mouse central nervous system. Eur. J. Neurosci 16, 2291–2296 (2002). [DOI] [PubMed] [Google Scholar]
- 316.Elger B et al. Optimized synthesis of AMPA receptor antagonist ZK 187638 and neurobehavioral activity in a mouse model of neuronal ceroid lipofuscinosis. ChemMedChem 1, 1142–1148 (2006). [DOI] [PubMed] [Google Scholar]
- 317.Katz ML, Rice LM & Gao CL Dietary carnitine supplements slow disease progression in a putative mouse model for hereditary ceroid-lipofuscinosis. J. Neurosci. Res 50, 123–132 (1997). [DOI] [PubMed] [Google Scholar]
- 318.Zeman RJ, Peng H & Etlinger JD Clenbuterol retards loss of motor function in motor neuron degeneration mice. Exp. Neurol 187, 460–467 (2004). [DOI] [PubMed] [Google Scholar]
- 319.Koch S et al. Morphologic and functional correlates of synaptic pathology in the cathepsin D knockout mouse model of congenital neuronal ceroid lipofuscinosis. J. Neuropathol. Exp. Neurol 70, 1089–1096 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.Koike M et al. Cathepsin D deficiency induces lysosomal storage with ceroid lipofuscin in mouse CNS neurons. J. Neurosci 20, 6898–6906 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321.Partanen S et al. Synaptic changes in the thalamocortical system of cathepsin D-deficient mice: a model of human congenital neuronal ceroid-lipofuscinosis. J. Neuropathol. Exp. Neurol 67, 16–29 (2008). [DOI] [PubMed] [Google Scholar]
- 322.Shevtsova Z et al. CNS-expressed cathepsin D prevents lymphopenia in a murine model of congenital neuronal ceroid lipofuscinosis. Am. J. Pathol 177, 271–279 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]