CADASIL, or Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy, is a rare genetic disorder characterized by the progressive thickening of small and medium-sized arteries in the brain due to mutations in the NOTCH3 gene on chromosome 19, leading to reduced blood flow, recurrent subcortical ischemic strokes, white matter lesions, and eventual vascular dementia.[1] This autosomal dominant condition is the most common monogenic form of cerebral small vessel disease, with an estimated prevalence of 2 to 5 cases per 100,000 individuals worldwide, though it may be underdiagnosed in some populations.[2] Pathologically, it involves the accumulation of granular osmiophilic material (GOM) in arterial smooth muscle cells, causing vessel wall degeneration and ischemia primarily in the subcortical regions.[1]Clinically, CADASIL typically manifests between the ages of 20 and 60 years, with an average onset in the 30s to 40s, though symptoms can vary widely even within families.[1] Common initial symptoms include migraines with aura, affecting up to 55% of patients, often severe and accompanied by transient neurological deficits.[1] This is frequently followed by recurrent lacunar infarcts and transient ischemic attacks in 60-85% of cases, mood disturbances or psychiatric symptoms in 25-30%, and progressive cognitive impairment leading to dementia in about 60% by midlife.[1] Other features may include apathy, depression, and pyramidal signs, with disability often progressing to dependency in the 60s for many, and life expectancy now estimated around 65-70 years depending on gender and management.[1][3]Diagnosis relies on a combination of clinical presentation, neuroimaging, and genetic confirmation, as there is no single definitive test.[1]Brain MRI characteristically reveals extensive T2/FLAIR hyperintensities in the anterior temporal poles and external capsule by age 30, distinguishing it from sporadic small vessel disease.[1] Skin or muscle biopsy can detect GOM deposits in about 50% of cases, but genetic testing for NOTCH3 mutations—over 280 of which have been identified—is the gold standard, confirming the diagnosis in nearly all affected individuals.[1][2] There is no disease-modifying treatment, so management focuses on secondary prevention through control of cardiovascular risk factors (e.g., hypertension, smoking cessation), symptomatic relief for migraines and mood disorders, and supportive care for cognitive and motor decline.[1] Ongoing research explores potential therapies targeting Notch3 signaling pathways. As of 2025, initiatives such as an $8 million grant to Mount Sinai for translational research and explorations of base editing and immunotherapy show promise in targeting NOTCH3 dysfunction.[2][4][5]
Background
Definition and Classification
CADASIL, or Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy, is a monogenic form of cerebral small vessel disease characterized by the progressive degeneration of small- to medium-sized arteries, primarily in the brain.[6] The acronym encapsulates its core features: "cerebral" refers to its predominant involvement of brain vasculature, "autosomal dominant" denotes the inheritance pattern, "arteriopathy" indicates the arterial pathology, "subcortical infarcts" highlights the location of ischemic events below the cerebral cortex, and "leukoencephalopathy" describes the associated white matter abnormalities.[7] This condition leads to recurrent subcortical strokes, cognitive decline, and other neurological manifestations such as migraines and mood disturbances.[8]Classified as an autosomal dominant hereditary strokedisorder, CADASIL falls within the broader category of cerebral small vessel diseases but is distinguished by its monogenic etiology, in contrast to sporadic forms of vascular dementia like those driven by hypertension or aging.[6] Unlike multifactorial sporadic small vessel diseases, CADASIL arises from a single genemutation, making it a prototypical hereditary cerebral microangiopathy that affects vascular smooth muscle cells and endothelial integrity.[9] This genetic basis underscores its role as a model for understanding inherited vascular pathologies, separate from acquired cerebrovascular conditions.[10]Key pathological hallmarks include the accumulation of granular osmiophilic material (GOM) deposits in arterial walls, which can be detected via electron microscopy in skin or brain biopsies, alongside neuroimaging evidence of subcortical infarcts and diffuse leukoencephalopathy.[6] The disease's arteriopathy results in narrowed and thickened vessel walls, impairing blood flow and causing ischemic damage predominantly in subcortical regions.[8] Historically, the identification of CADASIL unified several previously disparate familial conditions resembling Binswanger's disease, a sporadic subcortical arteriosclerotic encephalopathy, by revealing a shared genetic underpinning in affected kindreds.[10]
History and Discovery
The earliest descriptions of what is now recognized as CADASIL appeared in the mid-20th century, initially classified under broader terms like familial subcortical dementia or hereditary multi-infarct dementia. In 1955, neurologist Ludo van Bogaert reported two sisters exhibiting rapidly progressive subcortical encephalopathy resembling Binswanger's disease, characterized by dementia, gait disturbances, and white matter changes without typical risk factors for vascular disease.[11] Subsequent cases in the 1970s further highlighted the hereditary nature of the condition; for instance, in 1977, Sourander and Wålinder described a large Swedish family with autosomal dominant inheritance of recurrent strokes, migraines, and progressive dementia, termed hereditary multi-infarct dementia, distinguishing it from sporadic vascular dementias. Granular osmiophilic material (GOM) deposits were identified in pathological examinations of affected vessels during these early studies.[12]The modern understanding of CADASIL emerged in the early 1990s through systematic genetic studies in French cohorts. In 1993, Elisabeth Tournier-Lasserve and colleagues performed linkage analysis on two unrelated large French families affected by recurrent subcortical strokes and leukoencephalopathy, mapping the causative gene to chromosome 19q12 and proposing the descriptive term "cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy" to reflect its inherited vascular pathology.[13] The acronym CADASIL was introduced in 1993 by Tournier-Lasserve et al., with Marie-Germaine Bousser contributing to its standardization in workshop proceedings. Subsequent studies refined the locus to 19p13, facilitating the identification of mutations in the NOTCH3 gene. A pivotal advancement occurred in 1996 when Anne Joutel and collaborators identified mutations in the NOTCH3 gene within the linked chromosomal region, confirming CADASIL as a monogenic disorder caused by cysteine-altering variants in this vascular receptor.[14] This discovery enabled targeted genetic testing, which expanded significantly in the 2000s as sequencing technologies improved and became commercially available, facilitating diagnosis in sporadic cases and international registries.[15] By the 2010s, research increasingly recognized the broad phenotypic variability of CADASIL, with studies documenting differences in age of onset, stroke frequency, and cognitive decline influenced by mutation location and modifiers, refining clinical expectations beyond uniform progression.[16]
Epidemiology
Prevalence and Incidence
CADASIL is estimated to affect 2 to 5 individuals per 100,000 population globally, though this figure likely underrepresents the true burden due to diagnostic challenges.[17] In Western Europe, prevalence rates range from 2 to 4 per 100,000 adults, based on population-based genetic screening studies.[9] Comparable estimates of 1.2 to 3.6 per 100,000 adults have been reported in East Asian populations, such as in Japan, where regional registries have identified clusters in areas like Kansai and Kanto.[18]Incidence data for CADASIL remain limited, as the condition follows an autosomal dominant inheritance pattern with near-complete penetrance by age 65, but new cases typically manifest with initial symptoms between ages 30 and 50.[9] The disease's onset is insidious, often beginning with migraines or mood disturbances, which contributes to delayed recognition.[2]Geographic variations in prevalence are influenced by founder effects, particularly in European populations where specific NOTCH3 mutations, such as the p.R133C variant, have led to higher localized rates; for instance, studies in Finland highlight enriched mutation frequencies due to historical genetic bottlenecks.[19] Emerging genomic research from the 2020s has expanded understanding in diverse ethnic groups, revealing similar prevalence ranges in non-European cohorts, including those from South Asia and the Middle East, though data from African and Latin American populations remain sparse.[20]Underreporting of CADASIL is widespread, primarily due to misdiagnosis as conditions like multiple sclerosis or Alzheimer's disease, especially before genetic testing for NOTCH3 mutations became routinely available and accessible after the early 2000s.[21] This has resulted in an underestimation of global incidence, with recent studies suggesting the true prevalence may be substantially higher upon broader screening.[22]
Risk Factors and Demographics
CADASIL is primarily a genetic disorder caused by mutations in the NOTCH3 gene, following an autosomal dominant inheritance pattern, which means that a single copy of the mutated gene from an affected parent confers the risk to offspring.[6] Each child of an individual with CADASIL has a 50% chance of inheriting the pathogenic variant and developing the disease.[6] Family history thus represents the primary non-modifiable risk factor for susceptibility.[6]The disease affects males and females equally in terms of incidence, with no significant sex-based differences in overall prevalence.[23] However, clinical manifestations show sex-specific patterns: migraine with aura occurs more frequently in women, often with an earlier onset before age 51.[23] In contrast, men experience a higher frequency of strokes before menopause and more severe post-stroke outcomes, such as elevated National Institutes of Health Stroke Scale scores, after age 51.[23] Median survival is shorter in men (approximately 65 years) compared to women (approximately 71 years), potentially contributing to higher stroke-related morbidity in older males.[24]Modifiable cardiovascular risk factors can exacerbate vascular damage and accelerate disease progression in CADASIL patients. Hypertension is associated with an increased risk of incident stroke (odds ratio 2.69) and should be aggressively managed.[25]Smoking similarly worsens outcomes, leading to strokes occurring about 10 years earlier in affected individuals.[26] Recent studies in the 2020s have linked hyperlipidemia (dyslipidemia) to faster phenotypic progression, with it present in a substantial proportion of patients alongside other vascular risks.[27][25]Beyond the primary NOTCH3 mutation, non-genetic modifiers influence severity. The APOE ε4 allele interacts with CADASIL pathology, independently increasing the risk of incident dementia (hazard ratio 10.70) and baseline dementia (odds ratio 3.51), particularly in patients with certain mutations like p.Arg544Cys.[28] Ethnic variations also affect mutation hotspots; for instance, Asian populations, including Chinese, Japanese, and Korean cohorts, show a higher prevalence of specific cysteine-altering NOTCH3 mutations such as p.R544C and p.R607C compared to Western populations.[29]
Clinical Presentation
Signs and Symptoms
CADASIL manifests through a range of neurological, cognitive, psychiatric, and other symptoms, primarily resulting from small vessel disease in the brain. The most prominent features include recurrent ischemic events, migraines, and progressive cognitive changes, often beginning in mid-adulthood.[6]Neurological symptoms in CADASIL frequently involve recurrent subcortical ischemic strokes, typically lacunar infarcts affecting deep brain structures, with over 85% of individuals experiencing at least one stroke. Transient ischemic attacks (TIAs) are also common, presenting as temporary episodes of neurological dysfunction such as weakness or speech difficulties. Pyramidal signs, including spasticity and hyperreflexia, emerge as motor abnormalities due to upper motor neuron involvement. Sensory disturbances, such as numbness or paresthesia, may occur alongside these, reflecting white matter and subcortical damage.[6][30][8]Migraines with aura affect approximately 40-55% of individuals with CADASIL, often onsetting in the 20s or 30s and preceding other symptoms. These migraines are typically more frequent and prolonged than in the general population, with aura symptoms including visual disturbances, sensory changes, or aphasia; hemiplegic variants, characterized by unilateral weakness, occur in some cases. Atypical aura features, such as prolonged duration or multiple aura types, are reported in up to 59% of those with migraine aura.[30][31][32]Cognitive impairments begin with early executive dysfunction, affecting planning, attention, and problem-solving, and are evident in about 40-50% of patients by age 50. Over time, these evolve into broader deficits in memory and global cognitive function, though initial presentations emphasize frontal-subcortical involvement.[30][33]Psychiatric symptoms include mood disorders such as depression and apathy, occurring in 20-30% of cases and often leading to social withdrawal. These may present early and independently of cognitive decline. Psychosis is rare but has been documented in isolated reports.[30][34]Other manifestations encompass epilepsy, with seizures reported in 5-10% of individuals, typically focal or generalized and linked to cortical involvement. Additional features can include parkinsonism with slow movements and tremors, as well as vision problems from optic nerve or retinal changes.[8][35][36]
Disease Progression and Stages
CADASIL typically unfolds in distinct phases, beginning with a prodromal period in the 20s to 40s characterized by initial manifestations such as migraines with aura (mean onset age 28 years, range 6-54 years) and mood disturbances including depression or apathy (mean onset age 32 years, range 20-59 years).[6] This early phase often precedes more overt neurological involvement, with symptoms varying in severity and not always leading immediately to diagnosis.[6]The disease advances to a stroke phase in the 40s to 50s, marked by recurrent subcortical ischemic events (mean onset age 47 years, range 20-76 years), which accumulate and contribute to stepwise neurological deterioration.[6] These events, averaging 2 to 5 over a lifetime, often result in progressive motor and cognitive impairments without complete recovery between episodes.[37] By the dementia phase in the 60s onward (mean onset age 62 years, range 45-80 years), severe cognitive decline predominates, leading to vascular dementia in most affected individuals.[6]Progression is highly variable, with a mean survival of approximately 20 years post-diagnosis, though recent cohorts show improved outcomes with medianage at death around 65-77 years depending on sex and era of diagnosis. Recent studies suggest that the clinical phenotype and prognosis of CADASIL are improving over time, possibly due to better control of cardiovascular risk factors.[6][3]Disability milestones include gait disturbance often requiring assistance by the 50s, with fewer than half of patients over 60 years able to walk unassisted and nearly 80% becoming fully dependent near death.[38]Wheelchair dependence commonly emerges in the 60s as cumulative subcortical infarcts exacerbate motor deficits.[38]Phenotypic variability influences the rate and severity of progression, with certain NOTCH3 mutations like p.Arg133Cys associated with more severe features, including earlier onset of strokes (range 28-71 years) and greater overall disability compared to other variants.[39] Complications from repeated infarcts frequently include pseudobulbar palsy and urinary incontinence, contributing to profound functional impairment in advanced stages.[6]
Pathophysiology
Genetic Basis
CADASIL is caused by pathogenic variants in the NOTCH3 gene, located on chromosome 19p13.12, which encodes a single-pass transmembrane receptor protein essential for cell-cell signaling, particularly in vascular smooth muscle cells and pericytes during vascular development and maintenance.[40] The NOTCH3 receptor consists of an extracellular domain with 34 epidermal growth factor-like (EGF-like) repeats, a negative regulatory region, a transmembrane domain, and an intracellular domain that mediates signaling upon ligand binding, influencing processes such as cell differentiation and proliferation.[41]Over 200 distinct NOTCH3 mutations have been identified as causative for CADASIL, with the vast majority being missense mutations that alter cysteine residues in the EGF-like repeats of the extracellular domain, leading to an odd number of cysteines and disrupting proper protein folding and multimerization.[42] These mutations are highly stereotyped, almost exclusively affecting the 34 EGF-like domains (exons 2-24), and result in the accumulation of granular osmiophilic material in affected tissues.[43] Common examples include p.R169C in exon 4 and p.R544C in exon 11, though the mutation spectrum varies by population.[19]The disorder follows an autosomal dominant inheritance pattern with high but incomplete age-dependent penetrance, such that most individuals develop clinical or radiological features by age 65, though a minority (~20%) may remain asymptomatic.[6]De novo mutations occur rarely, accounting for less than 5% of cases, and most affected individuals inherit the variant from an affected parent.[44]Genotype-phenotype correlations exist, with mutations in exon 4, such as p.R169C, often associated with a more severe phenotype including earlier onset of strokes and cognitive decline compared to those in other exons.[45] Recent advances in next-generation sequencing (NGS) have facilitated the identification of novel NOTCH3 variants, expanding the known genetic spectrum and improving diagnostic yield in atypical or sporadic cases as of 2025.[46]
Cellular and Molecular Mechanisms
The mutant NOTCH3 protein in CADASIL accumulates abnormally within vascular smooth muscle cells (VSMCs), leading to the formation of granular osmiophilic material (GOM) aggregates, which are electron-dense deposits visible under electron microscopy. These aggregates consist of the extracellular domain of NOTCH3 and are a hallmark pathological feature, disrupting normal protein homeostasis and triggering proteotoxic stress in affected cells. Studies have shown that both mutant and wild-type NOTCH3 can co-aggregate, exacerbating the accumulation and contributing to progressive cellular damage in small arteries.[47][48]This protein dysfunction results in the degeneration of VSMCs, a primary component of arterial walls, causing loss of vascular integrity and structural remodeling. VSMC degeneration is accompanied by thickening of the basement membrane due to excessive deposition of extracellular matrix proteins, such as collagen and laminin, which narrows the vessel lumen and impairs vasodilation. These changes lead to reduced cerebral blood flow through chronic hypoperfusion, particularly in small penetrating arteries, and contribute to endothelial dysfunction by altering barrier properties and nitric oxide signaling. Endothelial cells exhibit reduced expression of tight junction proteins, promoting leakage and further vascular instability.[41][49]At the molecular level, mutations in NOTCH3 disrupt canonical Notch signaling, a pathway critical for VSMC differentiation, proliferation, and survival. The impaired signaling fails to maintain proper cell fate decisions, leading to dedifferentiation and apoptosis of VSMCs, while also affecting interactions with neighboring endothelial cells. Recent 2025 research highlights the involvement of oxidative stress and inflammation in amplifying these effects; elevated reactive oxygen species (ROS) from mitochondrial dysfunction induce endoplasmic reticulum (ER) stress, activating unfolded protein response (UPR) pathways that promote inflammatory cytokine release, such as IL-6 and TNF-α, further driving vascular pathology. These mechanisms link NOTCH3 dysfunction to a pro-inflammatory milieu that sustains tissue damage. Emerging 2025 studies also implicate NOTCH3 mutations in glymphatic system dysfunction, potentially exacerbating amyloid pathology, and in inducing clonal hematopoiesis, broadening the systemic impact of the disease.[50][5][51][52][53]In the brain, these cellular alterations manifest as white matter hyperintensities on MRI, resulting from chronic hypoperfusion and ischemia in periventricular and deep white matter regions due to the widespread arteriolopathy. Subcortical infarcts arise from occlusion or rupture of affected small vessels, compounded by the loss of autoregulatory capacity in cerebral circulation. The cumulative impact disrupts oligodendrocyte function and myelin integrity, contributing to the demyelination observed in CADASIL-affected brains.[54][55]
Diagnosis
Clinical Evaluation
Clinical evaluation of CADASIL begins with a thorough medical and family history to identify patterns suggestive of this autosomal dominant disorder. Patients often present with recurrent ischemic strokes or transient ischemic attacks before the age of 50, alongside a family pedigree demonstrating vertical transmission across generations, including early-onset migraines with aura, cognitive decline, or psychiatric symptoms in multiple affected relatives.[6] Emphasis is placed on documenting the absence of traditional vascular risk factors such as hypertension, diabetes, or hyperlipidemia, which helps distinguish CADASIL from sporadic small vessel disease.[1]The neurological examination focuses on assessing cognitive function, motor capabilities, and psychiatric status to detect early multisystem involvement. Cognitive screening tools like the Montreal Cognitive Assessment (MoCA) are commonly employed to quantify subtle impairments in executive function and memory, often yielding scores below 26 in affected individuals even prior to overt dementia.[56] Motor evaluation reveals pyramidal signs such as brisk reflexes, spasticity, or pseudobulbar palsy, while gait disturbances may indicate subcortical involvement; psychiatric assessment screens for mood disorders, including depression and apathy, which can precede neurological symptoms by years.[6]Differential diagnosis requires ruling out other causes of young-onset stroke and leukoencephalopathy, such as sporadic small vessel disease, Fabry disease, or mitochondrial encephalomyopathies like MELAS. Neuroimaging standards, such as those outlined by the STRIVE consortium, help characterize the subcortical vascular changes and absence of cortical infarcts in CADASIL, aiding differentiation of its cognitive impairment from other dementias.[1] Conditions like multiple sclerosis or inflammatory vasculopathies are considered if inflammatory markers or atypical symptoms are present, but the autosomal dominant family history strongly favors CADASIL.[56]Red flags prompting suspicion of CADASIL include lacunar strokes in individuals under 50 without conventional risk factors and a disproportionate prevalence of mood disorders, such as major depressive episodes, affecting 20-40% of cases and often appearing early.[6][34] These features, combined with subtle psychiatric manifestations, underscore the need for prompt evaluation in families with unexplained neurological morbidity.[56]
Imaging and Genetic Testing
Magnetic resonance imaging (MRI) is a cornerstone for diagnosing CADASIL, revealing characteristic symmetric white matter hyperintensities (WMHs) predominantly in the periventricular regions, deep white matter, and notably the anterior temporal poles, with involvement of the anterior temporal lobes observed in approximately 90% of cases and offering high specificity for the condition.[57][58] These WMHs appear as hyperintense lesions on T2-weighted and fluid-attenuated inversion recovery (FLAIR) sequences and are typically bilateral and symmetrical, distinguishing CADASIL from other small vessel diseases.[57] Lacunar infarcts, often multiple and located in subcortical areas, are also common on MRI and correlate strongly with cognitive impairment in CADASIL patients.[59][60]Advanced MRI techniques, such as susceptibility-weighted imaging (SWI), are valuable for detecting cerebral microbleeds, which are frequent in CADASIL and appear as hypointense foci in subcortical, brainstem, and thalamic regions, aiding in assessing disease burden and hemorrhagic risk.[61]Genetic testing provides definitive confirmation of CADASIL through sequencing of the NOTCH3 gene, with methods including traditional Sanger sequencing or next-generation sequencing (NGS), the latter offering faster and more comprehensive analysis of exons and splice sites.[62] These tests detect pathogenic variants, such as missense mutations, with a sensitivity exceeding 95% for establishing the diagnosis in symptomatic individuals.[63] Pre-symptomatic screening in at-risk family members follows guidelines similar to those for Huntington disease, emphasizing genetic counseling, psychological support, and testing only after age 18, typically recommended when clinical suspicion arises from family history.[64]Skin biopsy, though less commonly performed since the advent of reliable genetic testing, can support diagnosis via electron microscopy, which reveals granular osmiophilic material (GOM) deposits in the basal lamina of vascular smooth muscle cells in dermal arterioles.[65]Diagnostic criteria for CADASIL integrate clinical features, imaging abnormalities, and genetic or histopathological findings, building on early proposals like those from Sourander et al. (1993) and modified in subsequent guidelines to prioritize NOTCH3 mutations alongside characteristic MRI patterns for high-confidence diagnosis.[56]
Treatment and Management
Symptomatic Therapies
Symptomatic therapies for CADASIL aim to alleviate specific manifestations such as ischemic events, headaches, cognitive decline, mood disorders, and seizures, though no treatments modify the underlying disease progression. Management is tailored to individual symptoms, drawing from general neurological guidelines adapted for the vascular fragility in CADASIL, with caution to avoid therapies that increase hemorrhage risk. These approaches focus on acute relief and chronic symptom control, often requiring multidisciplinary input from neurologists, psychiatrists, and rehabilitation specialists.Stroke management in CADASIL primarily involves antiplatelet therapy for secondary prevention following ischemic events, as these patients are at high risk for recurrent subcortical infarcts. Aspirin or clopidogrel is commonly prescribed, with aspirin monotherapy considered safe for preventing recurrent strokes in symptomatic cases, though evidence for primary prevention is lacking. Anticoagulants are generally avoided due to the elevated risk of intracerebral hemorrhage associated with white matter lesions and microbleeds in CADASIL. Acute stroke care follows adapted protocols, with thrombolysis considered on a case-by-case basis due to uncertain safety and efficacy in CADASIL.[56] Endovascular interventions are used judiciously given the small-vessel pathology.Migraine treatment in CADASIL addresses both acute attacks and prophylaxis, with options selected to minimize vascular and cognitive side effects. For acute relief, triptans can be used cautiously, as they demonstrate similar efficacy and side effect profiles to the general migraine population without increased ischemic risk. Prophylactic agents include topiramate, which is preferred over beta-blockers due to reports of potential worsening of cognitive symptoms with the latter; beta-blockers should be avoided for prophylaxis due to potential worsening of cognitive symptoms, with alternatives including topiramate or acetazolamide.[56] Non-pharmacologic trigger avoidance complements these therapies, though detailed preventive strategies are covered elsewhere.Cognitive and psychiatric symptoms are managed supportively, recognizing the subcortical dementia and mood disturbances common in CADASIL. Cholinesterase inhibitors like donepezil show limited efficacy, with randomized trials demonstrating no significant improvement in cognitive scores for patients with vascular cognitive impairment due to CADASIL. For depression, which affects up to 30% of patients, selective serotonin reuptake inhibitors (SSRIs) are recommended as first-line therapy, offering good tolerability and symptom relief in most cases. Cognitive behavioral therapy (CBT) is incorporated for both depressive symptoms and mild cognitive challenges, providing non-pharmacologic support to enhance coping and daily functioning.Seizures, occurring in approximately 5-10% of CADASIL patients often secondary to infarcts, are controlled with antiepileptic drugs selected for minimal cognitive and vascular interactions. Levetiracetam is a preferred agent, administered at low doses (e.g., 500-1250 mg daily) to reduce seizure frequency while limiting side effects like sedation or mood alterations.
Preventive and Supportive Care
Preventive and supportive care for individuals with CADASIL focuses on mitigating modifiable vascular risk factors to potentially slow disease progression and enhancing quality of life through lifestyle modifications and multidisciplinary support.[6] Strict management of blood pressure is recommended, targeting levels below 130/80 mmHg to reduce the risk of ischemic events, as hypertension has been associated with increased stroke incidence in CADASIL patients.[56] Statins are advised for patients with dyslipidemia to control cholesterol levels, following general guidelines for cerebrovascular disease prevention.[6]Smoking cessation is strongly encouraged, as tobacco use exacerbates vascular damage and elevates stroke risk in this population.[8]Lifestyle interventions play a key role in supportive management, with adoption of a Mediterranean-style diet—rich in fruits, vegetables, whole grains, and healthy fats—recommended to support vascular health, though ongoing trials are evaluating its specific impact on CADASIL progression.[66] Regular aerobic exercise, such as walking or swimming for at least 150 minutes per week, is advised to maintain cardiovascular fitness and mobility, helping to counteract the gait disturbances common in later stages.[67]Genetic counseling is essential for affected individuals and their families, providing information on the autosomal dominant inheritance pattern and reproductive options, including preimplantation genetic diagnosis, to inform family planning decisions.[6]Supportive care emphasizes a multidisciplinary approach involving neurologists, physiotherapists, occupational therapists, and speech-language pathologists to address functional impairments. Physiotherapy is particularly beneficial for improving balance and gait in patients experiencing motor deficits, while speech therapy can assist those with dysphagia to prevent complications like aspiration.[68] Emotional and psychological support, including counseling, is recommended to manage the psychosocial burden of the disease and support family caregivers.[8]Regular monitoring is crucial for early detection of progression, with annual brain MRI recommended to assess white matter changes and lacunar infarcts, guiding adjustments to care plans.[69] Cognitive assessments, such as the Mini-Mental State Examination or more comprehensive batteries, should be performed yearly to track executive function and processing speed declines, enabling timely interventions.[67] For female carriers, 2020s guidelines highlight the need for close monitoring during pregnancy and the puerperium due to a potential increased risk of neurologic events, though pregnancy itself does not appear to substantially elevate stroke risk; multidisciplinary obstetric and neurological oversight is advised.[70][6]
Prognosis and Societal Impact
Long-term Outcomes
CADASIL significantly reduces life expectancy, with a median age at death of approximately 65 years in men and 71 years in women. More recent analyses report an overall median age at death around 68 years, with faster progression in males. Mortality is substantial following symptom onset, with most patients succumbing 10 to 20 years after initial clinical manifestations, and approximately 50% mortality within 20 years post-onset based on longitudinal cohort data.[71][6][72][12]The disease leads to profound disability in the majority of patients, with nearly 80% becoming completely dependent on caregivers immediately prior to death and common endpoints including a bedridden state and severe dementia. The median age for becoming bedridden is around 64 years, and by age 60, about 60% of patients are either deceased, demented, or bedridden.[38][73][74]Quality of life deteriorates markedly due to progressive neurological impairment, with emotional disturbances such as depression and apathy strongly associated with reduced patient well-being and elevated caregiver burden. The economic toll includes frequent early retirement, often in the fifth or sixth decade, driven by cognitive decline and physical limitations, contributing to substantial financial strain on families. Recent 2025 studies utilizing the modified Rankin Scale demonstrate steady progression of disability over 10 years, with transitions to higher dependence scores reflecting worsening functional status.[75][76][77]Outcomes are influenced by mutation severity, where more pathogenic NOTCH3 variants correlate with earlier onset and greater disease burden. Early intervention, including diagnosis before age 40, is linked to improved prognosis through timely preventive measures, as evidenced by lower stroke and dementia risks in more recently diagnosed cohorts. A 2025 longitudinal study of 555 patients reported a significant increase in the mean age of stroke onset over time, from 46.5 years in those diagnosed between 2001 and 2010 to 52.3 years in those diagnosed between 2011 and 2023, attributed to enhanced preventive care.[78][3][79]
Historical Cases and Awareness
The discovery of CADASIL stemmed from studies of a large French family in the early 1990s, where researchers Marie-Germaine Bousser and Elisabeth Tournier-Lasserve observed a hereditary pattern of recurrent strokes and dementia across multiple generations, leading to the identification of the underlying genetic basis and the NOTCH3 gene mutation in 1996.[80][81] This family's case, involving over 40 affected members, provided critical evidence of the autosomal dominant inheritance and vascular pathology, prompting international collaboration that confirmed similar patterns in other pedigrees.[82]In the 2000s, anonymous patient registries played a key role in advancing clinical understanding and trial recruitment, such as the UK CADASIL register established in 2002 at a regional neurosciences center, which enrolled genetically confirmed cases to track progression and inform studies on prognostic factors.[83] These registries facilitated multicenter trials, including a 2004 prospective study of 80 patients that evaluated stroke patterns and disability, contributing data for sample size calculations in future therapeutic research.[84] By anonymizing participant data, such efforts encouraged broader enrollment from affected families wary of genetic disclosure.Awareness efforts gained momentum with the establishment of the CADASIL Foundation in 2005 as a nonprofit to provide resources and support for patients and families, addressing the scarcity of information at the time.[85] The World Stroke Organization incorporated CADASIL into its educational modules on diagnosis and treatment by the early 2010s, enhancing recognition among clinicians through online academy content.[86] In 2025, media coverage highlighted advances in genetic screening, including a BBC report on a UK study showing delayed stroke onset with specialized care and a Mount Sinai grant announcement for research accelerating drug discovery via genetic data.[87][4]Societal impacts include stigma associated with hereditary dementia, where positive genetic test results have led to family estrangement or blame in some cases, exacerbating emotional burdens beyond physical symptoms.[88] Policy responses have included orphan drug designations in the US, such as FDA grants for therapies targeting CADASIL since the 2010s, providing incentives for rare disease research without full market approval yet.[89] Similar designations in the EU support development for conditions affecting fewer than 5 in 10,000 individuals.[9]Cultural depictions of CADASIL remain rare in literature and film, with no prominent portrayals identified, reflecting its obscurity as a monogenic disorder.[90] Awareness initiatives emphasize family planning education, including presymptomatic testing protocols that address ethical concerns like high dropout rates due to psychological distress, promoting informed reproductive decisions.[91]
Research and Future Directions
Current Studies on Variability
Recent studies from 2020 to 2025 have increasingly focused on phenotypic variability in CADASIL, revealing interactions between genetic mutations, environmental factors, and modifier genes that influence disease severity and onset. Cohorts such as the Australian CADASIL (AusCADASIL) study, launched in 2023 (NCT06148051), aim to enroll 150 participants from diverse ethnic backgrounds to examine how lifestyle and environmental exposures, including vascular risk factors like hypertension and smoking, modulate NOTCH3 mutation effects on clinical presentation.[92] Recent 2025 studies suggest gene-environment interactions, such as chronic exposure to oxidative stress or inflammation, exacerbate vascular smooth muscle cell (VSMC) dysfunction in CADASIL, leading to varied rates of cognitive decline and stroke incidence across patients.[93] Modifier genes like APOE have been implicated, with a 2023 study of individuals with NOTCH3 variants showing that the APOE ε2 allele is associated with more severe cognitive impairment, potentially through accelerated amyloid deposition in cerebral vessels.[94] Similarly, the APOE ε4 allele was linked in earlier seminal work to earlier stroke onset in patients under 50 years, an effect amplified by smoking, underscoring the role of lipid metabolism in phenotypic modulation.[95]Imaging biomarkers have advanced through longitudinal MRI trials, providing insights into lesion progression and variability prediction. A 2025 prospective study using 7T MRI in 22 CADASIL patients tracked white matter changes over two years, showing that lower baseline BOLD-CVR magnitude and higher dispersion are linked to increased WMH progression at specific locations.[96] Artificial intelligence models, including a 2024 two-stage convolutional neural network (CNN) validated on 652 MRI scans from 132 CADASIL patients, achieve high agreement (R=0.928–0.995) for automated WMH segmentation, facilitating precise measurement of longitudinal changes and identifying progression patterns tied to mutation type and age.[97] These AI-driven approaches, integrated with clinical data, predict cognitive trajectories with substantial sensitivity, highlighting temporal lobe atrophy as a key variability marker.Epidemiological research has expanded via multi-ethnic databases to address non-European mutation spectra and phenotypic differences. The Global CADASIL Consortium (GCC), established in 2023, promotes international collaboration on CADASIL research. Studies indicate phenotypic differences across ethnicities, though cysteine-sparing mutations remain uncommon globally and do not predominate in non-Europeans.[98][99] A 2022 analysis of 446 patients from international registries confirmed that mutation location in the NOTCH3 gene, combined with ethnic-specific vascular risk profiles, explains aspects of variability in stroke-free survival.[100] These efforts, including the AusCADASIL cohort, emphasize the need for inclusive databases to uncover how genetic diversity influences disease penetrance beyond European founder mutations.Basic science investigations using animal models continue to elucidate mechanisms of VSMC apoptosis underlying variability. Notch3 knockout mice exhibit progressive VSMC degeneration without granular osmiophilic material (GOM) deposits, mirroring human CADASIL arteriopathy and showing increased apoptosis rates in cerebral arteries by 18 months compared to wild-type controls. A 2011 knock-in model with the Arg170Cys mutation demonstrated GOM accumulation in VSMCs and pericytes, leading to microinfarcts in aged mice, with apoptosis linked to impaired Notch3 signaling and endothelial dysfunction. Recent studies in transgenic mice have revealed that mutant NOTCH3 aggregates trigger endoplasmic reticulum stress, contributing to VSMC apoptosis under hemodynamic stress, providing a mechanistic basis for observed human phenotypic differences.
Emerging Therapies and Trials
Research into emerging therapies for CADASIL focuses on addressing the underlying NOTCH3 mutations and their downstream effects on vascular smooth muscle cells (VSMCs), with several approaches in preclinical development and early clinical testing as of 2025.[55]Gene therapies targeting NOTCH3 represent a promising avenue to correct the genetic defect at its source. CRISPR-based editing, particularly using adeninebase editors, has been demonstrated in preclinical models to correct specific point mutations in NOTCH3 within patient-derived blood vessel organoids, restoring normal protein function and reducing aggregate formation without off-target effects.[101] Similarly, antisense oligonucleotides (ASOs) designed to induce exon skipping have shown potential to exclude mutant epidermal growth factor-like repeat domains from the NOTCH3 protein, thereby correcting the odd number of cysteine residues that lead to misfolding and toxicity in mouse models and cell lines.[102] These nucleic acid-based strategies remain in preclinical stages, with ongoing efforts to optimize delivery to cerebral vasculature for future translation.[55]Small molecule inhibitors modulating the Notch pathway are under investigation to mitigate the loss-of-function signaling and toxic gain-of-function aggregates caused by NOTCH3 mutations. Gamma-secretase inhibitors, which block the proteolytic cleavage required for Notch activation, have been explored in preclinical CADASIL models to reduce mutant protein accumulation, though their impact on restoring signaling remains under study due to the dual pathology in the disease.[103] As of 2025, no dedicated Phase I/II trials for gamma-secretase inhibitors in CADASIL are reported, but related Notch pathway modulators, such as Rho kinase (ROCK) inhibitors targeting ER stress-induced vasculopathy, have shown vascular protective effects in animal models.[103] Additionally, tocotrienols, a class of vitamin E-derived small molecules with antioxidant properties, are being tested in a Phase II trial (NCT04658823) to assess their ability to slow white matter hyperintensity progression and cognitive decline in CADASIL patients. As of November 2025, the trial remains ongoing with no published results.[104]Stem cell approaches aim to replace dysfunctional VSMCs, which are central to CADASIL pathology. Induced pluripotent stem cell (iPSC)-derived VSMCs from CADASIL patients have been used to model disease mechanisms, revealing impaired contractility and increased synthetic phenotype, and preclinical studies suggest that transplanting corrected iPSC-VSMCs could restore vascular integrity in mouse models.[105] Early safety data from European preclinical efforts, including EU-funded projects modeling vascular organoids, indicate feasibility for VSMC replacement without tumorigenicity, though no active clinical trials for stem cell therapy in CADASIL have reached human testing as of 2025.[106] These approaches leverage patient-specific iPSCs to bypass ethical concerns and enable personalized correction via gene editing prior to differentiation.[107]Clinical trials for emerging therapies in CADASIL are limited but expanding, with several interventional studies registered as of 2025, primarily focusing on neuroprotective and anti-inflammatory agents to delay cognitive decline. The CERebrolysin In CADASIL trial (NCT05755997), a Phase II study initiated in 2023, evaluates the neurotrophic peptide Cerebrolysin—an anti-inflammatory compound—for its risk-benefit profile in genetically confirmed patients, aiming to assess improvements in cognitive function and stroke prevention over 12 months.[108] Similarly, the AMCAD trial, a multicenter Phase II study of adrenomedullin (a vasodilatory peptide with anti-inflammatory effects), is assessing safety and efficacy in reducing ischemic events in CADASIL patients, with interim data suggesting potential stabilization of cerebral blood flow.[109] These trials build on preclinical evidence of inflammation's role in NOTCH3 aggregate exacerbation, providing foundational data for larger Phase III evaluations expected by 2027.[110] The 2025 CADASIL International Research Updates webinar highlighted progress in global cohorts and emerging therapies.[111]