Skip to main content
Research Article
Originally Published 22 November 2019
Free Access

New Treatment Approaches to Modify the Course of Cerebral Small Vessel Diseases

See related articles, p 9, p 12, p 21, p 29, p 47
Cerebral small vessel diseases (cSVDs) are a common cause of stroke and an important contributor to age-related cognitive decline and risk for dementia. Yet there is surprisingly little information on how to prevent their progression.
The common pathologies underlying cSVDs are arteriolosclerosis caused by aging, hypertension, and other conventional vascular risk factors and cerebral amyloid angiopathy (CAA) caused by vascular deposition of β-amyloid. These diseases lead to varied and overlapping types of vascular brain injury such as lacunes, white matter hyperintensities (WMHs) of presumed vascular origin, and microbleeds. These radiological manifestations of cSVD are common in aging populations. For example, >20% of elderly >80 years of age have ≥1 lacunes. Thus, cSVD is a major public health problem.
In this review, we discuss current treatment, identify gaps in our knowledge, review current trials addressing these gaps, and highlight methodological considerations that are important in planning future trials in cSVD.

Where Are We Now? Current Treatment of cSVD

For patients with lacunar stroke, secondary prevention strategies are mostly inferred from studies of ischemic stroke in general, the majority of which did not specifically examine efficacy in lacunar stroke. The only large definitive phase 3 trial to focus exclusively on lacunar stroke patients, with magnetic resonance imaging (MRI) confirmation, is the SPS3 trial (Secondary Prevention of Small Subcortical Strokes) of 3020 patients. This trial showed that long-term dual antiplatelet therapy was not superior to aspirin alone1 and that more intensive blood pressure lowering (target systolic, <130 mm Hg) reduced recurrent strokes. This reduction did not reach statistical significance for total stroke (ie, ischemic and hemorrhagic combined; hazard ratio, 0.81 [95% CI, 0.64–1.03]) but did for hemorrhagic stroke alone (hazard ratio, 0.37 [95% CI, 0.15–0.95]).2 No trials similar to SPS3 have been performed to assess the effects of statins, smoking cessation, diabetes mellitus management, and lifestyle interventions on lacunar stroke.
Unfortunately, little reliable information on preventing lacunes is available from other clinical trials because unlike SPS3, they generally have not required MRI for stroke subtyping. Treatment approaches to lacunar stroke have recently been systematically reviewed.3 As many as half of patients with a lacunar syndrome do not have cSVD on more rigorous subtyping.4 A systematic review of only 2 prior studies reported single antiplatelet therapy is better than no therapy for preventing new stroke (ischemic and hemorrhagic combined; risk ratio, 0.77 [95% CI, 0.62–0.97]) or new ischemic stroke (risk ratio, 0.48 [95% CI, 0.30–0.78]) in patients with recent lacunar stroke, but cSVD subtyping within these studies was not optimal.5
For patients with diffuse WMHs with or without lacunes, there have been no large-scale, definitive trials. This includes no definitive trials in patients with covert signs of cSVD (such as confluent WMH) on neuroimaging,6 in patients with vascular cognitive impairment,7 or in patients with CAA.

Are New Treatments for cSVD Needed?

Given the success in preventing stroke in general, with important reductions in incidence and mortality, are new treatments to target cSVD progression even necessary? That they are is supported by the evidence that many patients with cSVD experience progression with cognitive and functional decline despite conventional stroke risk reduction.
In the SCANS study (St George’s Cognition and Neuroimaging in Stroke) of 99 patients with symptomatic lacunar stroke and confluent WMH receiving conventional medical therapy, 18% progressed to dementia over 5 years.8 Even though all patients were known to have stroke and receive routine stroke preventive care, significant radiological progression was detected for all cSVD-related markers except for enlarged perivascular spaces (specifically, changes in lacunar volume, WMH volume, mean diffusivity, and reduced fractional anisotropy on diffusion tensor imaging were seen).8–12 Only 3% had a symptomatic recurrent stroke, but 27% had new lacunes, with >3 new lacunes in 8%.8 Patients with new lacunes exhibited declines in executive function.8
Furthermore, the pathophysiology of cSVD is complex, involving multiple pathways (eg, blood-brain barrier permeability13) that are distinct from the thromboembolism, cholesterol dysmetabolism, and glucose dysregulation that are the primary targets for current ischemic stroke prevention. As an example, recent genetic studies have found genetic variants in thrombosis and hemostasis are associated with an increased the risk of both large artery and cardioembolic stroke but not with lacunar stroke.14,15 This raises the possibility that antiplatelet and anticoagulation therapy may have less effect in cSVD.

Insights From cSVD-Focused Substudies of Major Trials

An efficient approach to test the effect of vascular risk reduction on cSVD progression is to piggyback an MRI substudy onto large clinical trials. This provides access to large study populations but limits interventions to those being tested against cardiovascular disease in general (mostly caused by atherosclerosis or cardiac disease rather than small vessel disease) and that if lacunar stroke is used as an end point, then accurate subtyping of stroke end points is required.
To identify recent and ongoing cSVD trials, we systematically searched the PubMed and https://www.clinicaltrials.gov databases using keywords for cSVD, WMHs, and vascular cognitive impairment in April 2019, combined with expert knowledge and hand searching of reference lists.
cSVD-focused MRI substudies investigated the effects of a multidomain lifestyle intervention, comprehensive vascular care, antihypertensives, statins, and intensive glucose control (Table 1). A systematic review of 4 trials found that antihypertensives reduced WMH progression (standardized mean difference, −0.19 [95% CI, −0.32 to −0.06]).16 The recently published SPRINT-MIND trial (Systolic Blood Pressure Intervention Trial - Mind Substudy) found that intensive systolic blood pressure lowering to <120 mm Hg compared with <140 mm Hg reduced WMH progression17 (Table 1) and the combined end point of mild cognitive impairment and dementia (hazard ratio, 0.85 [95% CI, 0.74−0.97]).18
Table 1. Published Substudies of Cerebral Small Vessel Disease Progression Embedded Within Prevention Trials
TrialYearPopulationInterventionnDurationOutcome
PreDIVA192017CommunityNurse-led vascular care1953 yNo difference in WMH progression or new infarcts
EVA202010AD with WMHAspirin, vascular care652 yLess progression of WMH by visual rating score; no difference in lacunes
FINGER212019Older, at risk for cognitive declineMultidomain: diet, exercise, cognitive training, vascular care1222 yNo difference in WMH progression
SPRINT172018High blood pressureIntensive BP control (systolic <120 mm Hg)4544.0 yLess progression of WMH difference with intensive treatment; no difference in brain volume
SCOPE222007Older, hypertensiveCandesartan1332 yFewer patients with high WMH increase (fifth quintile) in candesartan vs placebo
PROGRESS232004StrokePerindopril plus indapamide6673.9 yNo difference in new infarcts
PROGRESS242005StrokePerindopril plus indapamide1923.0 yLess WMH increase
PROFESS252012Ischemic strokeTelmisartan77128 moNo difference in WMH progression
VITATOPS262012Ischemic stroke/TIAB vitamins3592 yNo difference in WMH progression or lacune incidence; less WMH increase if severe WMH at baseline
PROSPER272005Cardiovascular riskPravastatin, 40 mg daily53533 moNo difference in WMH progression or change in infarct volume
ROCAS282009TIASimvastatin, 20 mg daily2082 yNo difference in WMH progression; less WMH increase if severe WMH at baseline
ACCORD292011Diabetes mellitusIntensive glycemic control (HbA1c, <6.0%)50340 moLess decline in brain volume but higher increase in WMH volume
ACCORD indicates Action to Control Cardiovascular Risk in Diabetes; AD, Alzheimer disease; BP, blood pressure; EVA, Evaluation of Vascular Care in Alzheimer’s Disease; FINGER, Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability; HbA1c, hemoglobin A1c; PreDIVA, Prevention of Dementia by Intensive Vascular Care; PROFESS, Prevention Regimen for Effectively Avoiding Second Strokes; PROGRESS, Perindopril Protection Against Recurrent Stroke Study; PROSPER, Prospective Study of Pravastatin in the Elderly at Risk; ROCAS, Regression of Cerebral Artery Stenosis; SCOPE, Study on Cognition and Prognosis in the Elderly; SPRINT, Systolic Blood Pressure Intervention Trial; TIA, transient ischemic attack; VITATOPS, Vitamins to Prevent Stroke; and WMH, white matter hyperintensity.

Early Phase Trials to Prevent cSVD Progression

New approaches to preventing cSVD might target its causes, its consequences, or, for cognition, enhance cognitive reserve. In this review, we focus on interventions that reduce the progression of cSVD. Thus, we do not review trials of cognitive training, physical or cognitive rehabilitation after stroke, cognitive enhancing medications, or the safety of antithrombotics and anticoagulants.
The additional benefit to be gained from more aggressive conventional risk factor management is unclear; as summarized in a recent review: “vascular prophylaxis, as appropriate for large artery disease and cardioembolism, includes antithrombotics, and blood pressure and lipid lowering; however, these strategies may not be effective for small vessel disease, or are already used routinely so precluding further detailed study.”30 As outlined earlier, intensive blood pressure lowering may reduce cSVD and cognitive impairment. New trials to prevent cSVD progression should also look to different choices of agents (eg, different antihypertensive classes) or different pathways, in addition to stricter treatment targets.
Early phase trials to prevent cSVD are tabulated separately for patients with lacunar stroke (Table 2), progressive chronic cSVD (Table 3), and CAA (Table 4). Interventions can be divided into 5 broad categories: lifestyle and behavioral, optimizing conventional vascular risk reduction strategies or targets, novel drugs, remote ischemic conditioning (RIC), and interventions to reduce amyloid accumulation in CAA (Table 3).
Table 2. Completed and Ongoing Early Phase Trials Targeting Lacunar Stroke
InterventionName/RegistrationPopulationSizeKey OutcomesDurationResults or Expected Completion Date
Lifestyle and behavior
 High-intensity trainingHITPALS/NCT02731235History of lacunar stroke84WMH volume1 yEstimated completion: 2019
New drugs
 Cilostazol31ECLIPSE/NCT00741286Lacunar stroke130WMH volume90 dNo difference in WMH volume change
 Cilostazol, 100 mg BID, and isosorbide mononitrate, 25 mg BID32LACI-1/NCT02481323Lacunar stroke57MRI CO2 reactivity11 wkWell tolerated and safe. MRI substudy in 28 showed higher proportion with WMH decrease in cilostazol arms
 Cilostazol, 100 mg BID, and isosorbide mononitrate, 25 mg BIDLACI-2/NCT03451591Lacunar stroke400Combined stroke, myocardial infarction, death, cognitive impairment, dependency1 yEstimated completion: November 2020
RIC
 RICREM-PROTECT/NCT02169739Lacunar stroke60WMH volume1 yEstimated completion: December 2019
Registration numbers are for https://www.clinicaltrials.gov. ECLIPSE indicates Effect of Cilostazol in Acute Lacunar Infarction Based on Pulsatility Index of Transcranial Doppler; HITPALS, High Intensity Training in Patients With Lacunar Stroke; LACI, Lacunar Intervention; MRI, magnetic resonance imaging; REM-PROTECT, Remote Preconditioning Over Time to Empower Cerebral Tissue; RIC, remote ischemic conditioning; TCD, transcranial Doppler; and WMH, white matter hyperintensity.
Table 3. Completed and Ongoing Early Phase Trials Testing Interventions for Reducing Progression of Chronic Cerebral Small Vessel Disease
InterventionName/RegistrationPopulationSizeKey OutcomesDurationResults or Expected Completion Date
Lifestyle and behavior
 Aerobic dance trainingADTSVD/NCT02730065≥2 lacunes or early confluent or confluent WMH11030-min cognitive battery, TCD pulsatility index6 moEstimated completion: 2019
 Aerobic exercise33,34NCT01027858WMH with neurological signs and cognitive impairment70ADAS-Cog, WMH volume6 moCompared with placebo, better ADAS-Cog scores at 6 mo but the difference was not sustained at 6 mo; no change in WMH volume in substudy of 30
 Aerobic exerciseRISE-2/NCT02068391High WMH39CBF, gray matter density6 moFinal data, March 2018; not yet published
 Moderate-intensity aerobic exercise35AIBLSubjective memory complaints or MCI with ≥1 vascular risk factor98WMH volume2 yNo difference in WMH volume at 24 mo
 Resistance training twice weekly or once weekly36NCT00426881Cognitively not impaired with WMH on MRI54WMH volume, Stroop test1 yEnd-study WMH volume lower in the twice-weekly group compared with stretching
 Resistance trainingReshaping the Path of Vascular Cognitive Impairment/NCT02669394Subcortical ischemic vascular cognitive impairment88ADAS-Cog subscale plus; WMH volume; MRI DTI1 yEstimated completion: December 2020
Optimizing vascular risk reduction
 Telmisartan, 80 mg per day, and rosuvastatin, 10 mg per day37NAHypertensive732WMH volume; MMSE5 yTwo-by-two factorial design with placebo. No effect of telmisartan; rosuvastatin treated had less increase in WMH but interpretation complicated by an interaction between the rosuvastatin and telmisartan arms
 Target systolic <125 mm Hg38,39PRESERVEHypertensive, lacunar stroke with confluent WMH111DTI MRI; CBF by ASL MRI (n=62)12 wkNo difference in DTI white matter damage between intensive and standard BP lowering; CBF preserved at lower target in substudy
 Telemetric BP monitoringPROHIBIT ICH/NCT03863665Primary ICH with systolic >130 mm Hg112WMH volume1 yEstimated completion: December 2021
 Amlodipine, losartan, and atenololTREAT-SVDS/NCT03082014Lacunar stroke, cognitive impairment with WMH, or CADASIL105MRI-measured vascular reactivity to CO24 wk of treatment with each (crossover design)Estimated completion: March 2020
Pharmacological
 Allopurinol, 300 mg BIDXILO-FIST/NCT02122718Recent ischemic stroke or TIA464WMH volume2 yEstimated completion: September 2020
 DL-3-n-butylphthalide, 200 mg TID40NASubcortical ischemic vascular cognitive impairment281ADAS-Cog, CIBIC-plus6 moChange in ADAS-Cog and CIBIC-plus favored treatment arm
 DL-3-n-butylphthalide, 200 mg TIDNCT03906123Subcortical ischemic vascular dementia64WMH volume, CBF on ASL MRI, neuropsychological testing48 wkEstimated completion: December 2019
 Tadalafil41PASTIS/NCT02450253Lacunar stroke or TIA with MRI lacunes or WMH55CBF by ASL MRISingle doseCompleted but not yet published
RIC
 RIC42,43NCT01658306Lacunar infarction or generalized WMH36WMH volume, MoCA1 yReduced WMH volume compared with sham-RIC; significantly better visuospatial and executive function sections of the MoCA
Registration numbers are for https://www.clinicaltrials.gov. ADAS-Cog indicates Alzheimer’s Disease Assessment Scale-Cognitive; ADTSVD, Aerobic Dance Training in Small Vessel Disease; AIBL, Australian Imaging, Biomarkers and Lifestyle; ASL, arterial spin label; BP, blood pressure; CADASIL, Cerebral Autosomal Dominant Arteriopathy With Ischemic Leukoencephalopathy; CBF, cerebral blood flow; CIBIC-Plus, clinician’s interview-based impression of change plus caregiver input; DTI, diffusion tensor imaging; ICH, intracerebral hemorrhage; MCI, mild cognitive impairment; MMSE, Folstein Minimental Status Exam; MoCA, Montreal Cognitive Assessment; MRI, magnetic resonance imaging; NA, not applicable; PASTIS, Perfusion by Arterial Spin Labelling Following Single Dose Tadalafil in Small Vessel Disease; PRESERVE, How Intensively Should We Treat Blood Pressure in Established Cerebral Small Vessel Disease?; PROHIBIT ICH, Prevention of Hypertensive Injury to the Brain by Intensive Treatment in Intracerebral Haemorrhage; RIC, remote ischemic conditioning; RISE-2, Recovery Improved in Covert Stroke With Exercise; TCD, transcranial Doppler ultrasound; TIA, transient ischemic attack; TREAT-SVDS, Effects of Amlodipine and Other Blood Pressure Lowering Agents on Microvascular Function in Small Vessel Diseases; WMH, white matter hyperintensity; and XILO-FIST, Xanthine Oxidase Inhibition for the Improvement of Long-Term Outcomes Following Ischaemic Stroke and Transient Ischaemic Attack.
Table 4. Completed and Ongoing Early Phase Trials Testing Interventions for Reducing Progression of CAA
InterventionName/RegistrationPopulationSizeKey OutcomesDurationResults or Expected Completion Date
Ponezumab44NCT01821118CAA by Boston criteria36MRI visual BOLD activation, MRI microbleed count90 dNo difference in MRI BOLD activation or microbleed count
Tramiprosate45NCT00056238CAA by Boston criteria24MRI microbleed count12 wkNo difference in microbleed count
Registration numbers are for https://www.clinicaltrials.gov. BOLD indicates blood oxygen level-dependent; CAA, cerebral amyloid angiopathy; and MRI, magnetic resonance imaging.

Trials of Lifestyle and Behavior Interventions

Two completed and 4 ongoing small trials are testing the effects of aerobic exercise, resistance training, or high-intensity training on cognition and imaging measures of cSVD (Tables 2 and 3). Hypothesized mechanisms of effect include improved cardiovascular health, improved cerebrovascular reactivity, increased angiogenesis, neuroprotection, reduced inflammation, and increased sensitivity to insulin.46

Trials Optimizing Conventional Vascular Risk Reduction Strategies or Targets

There has been concern that in patients with extensive cSVD, in whom reduced cerebral blood flow and cerebral autoregulation has been demonstrated, intensive blood pressure lowering could decrease perfusion further increasing ischemic damage.47 That this is not the case is supported by the recent cerebral blood flow substudy within the PRESERVE trial (How Intensively Should We Treat Blood Pressure in Established Cerebral Small Vessel Disease?),38 which showed that intensive lowering (target systolic, <125 mm Hg) was associated with no reduction in cerebral blood flow, and more recently by the full PRESERVE results showing no increase in white matter damage assessed by diffusion tensor imaging MRI (Hugh Markus personal communication; Table 3).

Trials of New Drugs for cSVD

We identified 4 new drugs that are being tested: allopurinol, cilostazol, DL-3-n-butylphthalide, and tadalafil (Table 3).
Allopurinol—a xanthine oxidase inhibitor—reduces uric acid levels, oxidative stress, and inflammation and improves cerebral NO bioavailability.48
Cilostazol is a phosphodiesterase 3 inhibitor that inhibits platelet aggregation and improves endothelial dysfunction,49 making it an attractive potential treatment to test in cSVD. It is already approved in major markets for other indications and has been the subject of several large trials in ischemic stroke (not limited to cSVD) primarily in Asia, where in some it appeared to have lower bleeding risk than other antithrombotics.50,51 In animal models of hypoperfusion (bilateral carotid artery occlusion or spontaneously hypertensive stroke-prone rat), cilostazol reduced blood-brain barrier permeability, reduced white matter microglial activation, reduced white matter demyelination, and improved endothelial function.52–57
DL-3-n-butylphthalide, naturally found in celery oil, has antithrombotic, antiapoptotic, and antioxidant effects. In a rat bilateral carotid occlusion model, it improved angiogenesis, cerebral blood flow, and cognitive deficits.58
Tadalafil is a phosphodiesterase 5 inhibitor that is used to treat erectile dysfunction, benign prostatic hypertrophy, and pulmonary arterial hypertension. It vasodilates small arteries, mediated by increases in intracellular cyclic GMP in smooth muscle cells.59 A small trial is testing the effect of a single dose on cerebral blood flow.41

Trials of RIC

One completed and one ongoing trial are testing the effects of RIC (Table 3). In experimental animal models, inducing brief periods of ischemia-reperfusion can exert remote protective effects on distant organs such as the brain.60 In practice, this can be done through cycles of inflation and deflation of a blood pressure cuff. In a mouse model of cerebral hypoperfusion induced by bilateral carotid occlusion, chronic daily hindlimb RIC increased angiogenesis, increased cerebral blood flow, and preserved cerebral white matter myelin.61 cSVD may be an excellent scenario for preconditioning, as serial MRI studies suggest that patients with cSVD have frequent, recurrent silent microinfarcts.62 Another attractive feature of RIC is that it induces a broad range of humoral responses that promote cell survival and inhibit inflammatory and apoptotic pathways, potentially addressing mechanisms of cSVD progression that are independent of infarction.

Trials for Reducing β-Amyloid Accumulation in CAA

The amyloid aggregation inhibitor tramiprosate showed no safety concerns in a randomized controlled trial of 24 patients,45 but the drug is of uncertain value for Alzheimer disease63 and was not developed further for CAA. The anti–aβ-monoclonal antibody ponezumab also showed no safety concerns in a randomized controlled trial of 36 patients, but the drug failed to improve visual task–related functional MRI activation (a surrogate of vascular reactivity and the preplanned surrogate efficacy outcome), and there are no plans for further development.44

Other Possible Interventions

A recent framework for cSVD progression, supported by existing biomarker studies (predominantly neuroimaging based), proposes that early endothelial dysfunction leads to disruption of the blood-brain barrier with leakage of fluid and toxic plasma proteins into the vascular media and surrounding tissues, with secondary effects on vascular reactivity, pericyte function, oligodendrocyte proliferation, and perivascular fluid drainage pathways.13 A better understanding of the molecular pathways underlying these processes may lead to new targets for drug therapy. Similar processes appear to be at play in CAA and arteriosclerotic disease, but more research is needed on their similarities and differences. Current evidence implicates molecular processes related to endothelial dysfunction, NO synthesis, blood-brain barrier integrity, maintenance and repair of the extracellular matrix (eg, matrix metalloproteinases and their inhibitors), oxidative stress, mechanical stress, thrombosis, and inflammation.64 Proposed new therapeutic approaches could include endothelin antagonists, neurotrophins, peroxisome proliferator–activated receptor-γ agonists, and prostacyclin mimics.30

How Can We Optimize Trial Design for cSVD?

The absence of positive trials for cSVD predominantly reflects a lack of trials, not past failures. Standardized neuroimaging definitions of cSVD, important for patient selection and surrogate outcomes, have been developed only recently. Additionally, a better understanding of the basic disease mechanisms is emerging. These recent developments likely explain the expanding number of early phase trials on cSVD.

Choosing the Patient Group to Include

A modern view of cSVD pathology emphasizes that it is a diffuse, progressive disease with strokes emerging in some patients as intermittent, stochastic events. Many patients experience effects of progressive cSVD in the absence of symptomatic strokes. The implication for clinical trials is that one must consider broadening selection criteria and outcomes beyond stroke events to include the changes in cognition, mood, and gait that are frequent in cSVD.
At the same time, we need to ensure that patients in cSVD trials really do have cSVD as the underlying pathology. Clinical lacunar and cognitive syndromes have low accuracy for cSVD and thus should not be used as the sole entry criteria. Neuroimaging is essential, and ideally, trials should include MRI to confirm the diagnosis.
cSVD has different subtypes and manifestations that may differ in their pathophysiology and response to treatments. CAA is a specific subtype of cSVD that is caused by vascular β-amyloid deposition and confers the highest risk of subsequent bleeding. Trial selection criteria must identify the appropriate cSVD population to match the hypothesized mechanism of intervention.
Lacunar infarcts can have different pathogeneses and do not always occur in conjunction with diffuse cSVD. Lacunar infarcts can also occur due to embolism or branch atheromatous disease or can occur in isolation. For studies targeting the diffuse arteriopathy underlying arteriosclerotic cSVD, it would be reasonable to require additional evidence of diffuse cSVD, for example, by requiring either multiple lacunes or that a threshold of WMH burden is exceeded.
To enrich the trial with more progression events, patients with higher burden of disease can be selected at baseline. Simple MRI scores have been developed that combine different neuroimaging features of cSVD65 and may allow this. The optimal disease stage for intervention is unclear. Effective intervention at an earlier stage would preserve function in a less disabled state but would require larger sample sizes to achieve the desired number of end points. CADASIL (Cerebral Autosomal Dominant Arteriopathy With Ischemic Leukoencephalopathy) and other monogenic hereditary cSVDs offer the opportunity for more specific diagnosis and earlier stage intervention (eg, in asymptomatic mutation carriers), but their molecular pathophysiology differs from acquired arteriosclerotic disease.

Optimizing the Outcome Measure

Hard clinical end points such as recurrent stroke and dementia are ideal and are most likely to change practice if the trial results are positive. However, in small vessel disease, the recurrent stroke rate is relatively low, and the dementia incidence is also low over the next 2 to 3 years. Longer follow-up and large sample sizes are, therefore, required in definitive phase 3 trials using these end points, making them expensive and challenging to perform. This has led to increasing interest in the use of surrogate end points, particularly to screen new therapies before large phase 3 trials.
Cognitive testing has been most widely used, but there is no consensus on the optimal cognitive battery. The harmonization standards from the National Institutes of Health and Canadian Stroke Network suggested 30- and 60-minute test batteries,66 while other simple batteries sensitive to the prominent executive dysfunction and delayed processing speed seen in cSVD have also been proposed.67 However, all these batteries have been developed for diagnosis, and whether they are optimal for detection of change needs evaluating.
In practice, current cognitive batteries have been shown to be insensitive to change over the 2- to 3-year time period used in most clinical trials.68,69 However, in the SCANS cohort of lacunar stroke patients, cognitive decline could be detected after 5 years.68 This means large sample sizes are required for cognitive end points (Table I in the online-only Data Supplement). Practice effects are likely to be one of the reasons why cognitive tests are insensitive to change; an improvement in cognition over 2 and 3 years of follow-up, especially in memory domain, was seen on the SCANS cohort, consistent with this.68
Because of the large sample sizes required with cognitive testing, there is increasing interest in using MRI as a surrogate marker for phase 2 trials. Quantitative measures of change in WMH volume, brain volume, and diffusion tensor imaging metrics appear to be the most sensitive to potential treatment effects, with sample sizes of 120 to 150 per arm needed to detect a 30% treatment effect over 3 years,12 although estimated sample sizes are sensitive to the underlying assumptions70,71 (Table I in the online-only Data Supplement). Standardized imaging core laboratory protocols are critical to maintain control over image quality.72 The Harmonizing Brain Magnetic Resonance Imaging Methods for Vascular Contributions to Neurodegeneration initiative provides resources for standardizing MRI methods, available at www.harness-neuroimaging.org.73
Blood or cerebrospinal fluid biomarkers may play a role in the future. For example, neurofilament light correlates well with degree of cSVD74; whether it is a sensitive marker in longitudinal studies remains to be determined.
cSVD affects not only cognition but also behavior and mood, activities of daily living, and quality of life, and including these as end points may add sensitivity and clinical relevance, although this remains to be demonstrated.

Conclusions and Future Directions

Substudies of large cardiovascular and stroke prevention trials provide moderate-quality evidence that lowering blood pressure reduces the progression of WMH but were not large enough to link this reduction to improved cognition or reduced stroke risk. Otherwise, little is known about how to prevent progression of cSVD. There have been few trials of interventions to prevent cSVD progression, probably because it is difficult to reliably and accurately subtype cSVD on clinical grounds alone. However, the time is ripe for a new generation of cSVD trials. The advent of widespread MRI scanning with standardized lesion definitions75 enables more accurate phenotyping, for example, into acute or chronic lacunar stroke with or without diffuse cSVD, vascular cognitive impairment with diffuse cSVD, and CAA. New trials should exploit advanced quantitative imaging markers (WMH volume, diffusion tensor imaging metrics, cerebral blood flow, and cerebrovascular reactivity) as surrogate end points, as they are more sensitive to change over time than cognition or recurrent stroke. With these improved tools, the field is now much better positioned to convert new pathophysiological insights into new treatments.

Supplemental Material

File (str_stroke-2018-024150_supp1.pdf)

References

1.
Benavente OR, Hart RG, McClure LA, Szychowski JM, Coffey CS, Pearce LA, et al; SPS3 Investigators. Effects of clopidogrel added to aspirin in patients with recent lacunar stroke. N Engl J Med. 2012;367:817–825. doi: 10.1056/NEJMoa1204133
2.
Benavente OR, Coffey CS, Conwit R, Hart RG, McClure LA, Pearce LA, et al; SPS3 Study Group. Blood-pressure targets in patients with recent lacunar stroke: the SPS3 randomised trial. Lancet. 2013;382:507–515. doi: 10.1016/S0140-6736(13)60852-1
3.
Das AS, Regenhardt RW, Feske SK, Gurol ME. Treatment approaches to lacunar stroke. J Stroke Cerebrovasc Dis. 2019;28:2055–2078. doi: 10.1016/j.jstrokecerebrovasdis.2019.05.004
4.
Asdaghi N, Jeerakathil T, Hameed B, Saini M, McCombe JA, Shuaib A, et al. Oxfordshire community stroke project classification poorly differentiates small cortical and subcortical infarcts. Stroke. 2011;42:2143–2148. doi: 10.1161/STROKEAHA.111.613752
5.
Kwok CS, Shoamanesh A, Copley HC, Myint PK, Loke YK, Benavente OR. Efficacy of antiplatelet therapy in secondary prevention following lacunar stroke: pooled analysis of randomized trials. Stroke. 2015;46:1014–1023. doi: 10.1161/STROKEAHA.114.008422
6.
Smith EE, Saposnik G, Biessels GJ, Doubal FN, Fornage M, Gorelick PB, et al; American Heart Association Stroke Council; Council on Cardiovascular Radiology and Intervention; Council on Functional Genomics and Translational Biology; and Council on Hypertension. Prevention of stroke in patients with silent cerebrovascular disease: a scientific statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2017;48:e44–e71. doi: 10.1161/STR.0000000000000116
7.
Smith EE, Cieslak A, Barber P, Chen J, Chen YW, Donnini I, et al. Therapeutic strategies and drug development for vascular cognitive impairment. J Am Heart Assoc. 2017;6:e005568. doi: 10.1161/JAHA.117.005568
8.
Zeestraten EA, Lawrence AJ, Lambert C, Benjamin P, Brookes RL, Mackinnon AD, et al. Change in multimodal MRI markers predicts dementia risk in cerebral small vessel disease. Neurology. 2017;89:1869–1876. doi: 10.1212/WNL.0000000000004594
9.
Benjamin P, Trippier S, Lawrence AJ, Lambert C, Zeestraten E, Williams OA, et al. Lacunar infarcts, but not perivascular spaces, are predictors of cognitive decline in cerebral small-vessel disease. Stroke. 2018;49:586–593. doi: 10.1161/STROKEAHA.117.017526
10.
Zeestraten EA, Benjamin P, Lambert C, Lawrence AJ, Williams OA, Morris RG, et al. Application of diffusion tensor imaging parameters to detect change in longitudinal studies in cerebral small vessel disease. PLoS One. 2016;11:e0147836. doi: 10.1371/journal.pone.0147836
11.
Lambert C, Benjamin P, Zeestraten E, Lawrence AJ, Barrick TR, Markus HS. Longitudinal patterns of leukoaraiosis and brain atrophy in symptomatic small vessel disease. Brain. 2016;139(pt 4):1136–1151. doi: 10.1093/brain/aww009
12.
Benjamin P, Zeestraten E, Lambert C, Ster IC, Williams OA, Lawrence AJ, et al. Progression of MRI markers in cerebral small vessel disease: sample size considerations for clinical trials. J Cereb Blood Flow Metab. 2016;36:228–240. doi: 10.1038/jcbfm.2015.113
13.
Wardlaw JM, Smith C, Dichgans M. Small vessel disease: mechanisms and clinical implications. Lancet Neurol. 2019;18:684–696. doi: 10.1016/S1474-4422(19)30079-1
14.
Williams FM, Carter AM, Hysi PG, Surdulescu G, Hodgkiss D, Soranzo N, et al; EuroCLOT Investigators; Wellcome Trust Case Control Consortium 2; MOnica Risk, Genetics, Archiving and Monograph; MetaStroke; International Stroke Genetics Consortium. Ischemic stroke is associated with the ABO locus: the EuroCLOT study. Ann Neurol. 2013;73:16–31. doi: 10.1002/ana.23838
15.
Malik R, Chauhan G, Traylor M, Sargurupremraj M, Okada Y, Mishra A, et al; AFGen Consortium; Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium; International Genomics of Blood Pressure (iGEN-BP) Consortium; INVENT Consortium; STARNET; BioBank Japan Cooperative Hospital Group; COMPASS Consortium; EPIC-CVD Consortium; EPIC-InterAct Consortium; International Stroke Genetics Consortium (ISGC); METASTROKE Consortium; Neurology Working Group of the CHARGE Consortium; NINDS Stroke Genetics Network (SiGN); UK Young Lacunar DNA Study; MEGASTROKE Consortium. Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes. Nat Genet. 2018;50:524–537. doi: 10.1038/s41588-018-0058-3
16.
van Middelaar T, Argillander TE, Schreuder FHBM, Deinum J, Richard E, Klijn CJM. Effect of antihypertensive medication on cerebral small vessel disease: a systematic review and meta-analysis. Stroke. 2018;49:1531–1533. doi: 10.1161/STROKEAHA.118.021160
17.
Nasrallah IM, Pajewski NM, Auchus AP, Chelune G, Cheung AK, Cleveland ML, et al; Sprint Mind Investigators for the SPRINT Research Group. Association of intensive vs standard blood pressure control with cerebral white matter lesions. JAMA. 2019;322:524–534. doi: 10.1001/jama.2019.10551
18.
Williamson JD, Pajewski NM, Auchus AP, Bryan RN, Chelune G, Cheung AK, et al; Sprint Mind Investigators for the SPRINT Research Group. Effect of intensive vs standard blood pressure control on probable dementia: a randomized clinical trial. JAMA. 2019;321:553–561. doi: 10.1001/jama.2018.21442
19.
van Dalen JW, Moll van Charante EP, Caan MWA, Scheltens P, Majoie CBLM, Nederveen AJ, et al. Effect of long-term vascular care on progression of cerebrovascular lesions: magnetic resonance imaging substudy of the PreDIVA trial (Prevention of Dementia by Intensive Vascular Care). Stroke. 2017;48:1842–1848. doi: 10.1161/STROKEAHA.117.017207
20.
Richard E, Gouw AA, Scheltens P, van Gool WA. Vascular care in patients with alzheimer disease with cerebrovascular lesions slows progression of white matter lesions on MRI: the Evaluation of Vascular Care in Alzheimer’s Disease (EVA) study. Stroke. 2010;41:554–556. doi: 10.1161/STROKEAHA.109.571281
21.
Stephen R, Liu Y, Ngandu T, Antikainen R, Hulkkonen J, Koikkalainen J, et al; FINGER Study Group. Brain volumes and cortical thickness on MRI in the Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER). Alzheimers Res Ther. 2019;11:53. doi: 10.1186/s13195-019-0506-z
22.
Firbank MJ, Wiseman RM, Burton EJ, Saxby BK, O’Brien JT, Ford GA. Brain atrophy and white matter hyperintensity change in older adults and relationship to blood pressure. Brain atrophy, WMH change and blood pressure. J Neurol. 2007;254:713–721. doi: 10.1007/s00415-006-0238-4
23.
Hasegawa Y, Yamaguchi T, Omae T, Woodward M, Chalmers J; PROGRESS CT Substudy Investigators. Effects of perindopril-based blood pressure lowering and of patient characteristics on the progression of silent brain infarct: the Perindopril Protection Against Recurrent Stroke Study (PROGRESS) CT Substudy in Japan. Hypertens Res. 2004;27:147–156. doi: 10.1291/hypres.27.147
24.
Dufouil C, Chalmers J, Coskun O, Besançon V, Bousser MG, Guillon P, et al; PROGRESS MRI Substudy Investigators. Effects of blood pressure lowering on cerebral white matter hyperintensities in patients with stroke: the PROGRESS (Perindopril Protection Against Recurrent Stroke Study) Magnetic Resonance Imaging Substudy. Circulation. 2005;112:1644–1650. doi: 10.1161/CIRCULATIONAHA.104.501163
25.
Weber R, Weimar C, Blatchford J, Hermansson K, Wanke I, Möller-Hartmann C, et al; PRoFESS Imaging Substudy Group. Telmisartan on top of antihypertensive treatment does not prevent progression of cerebral white matter lesions in the prevention regimen for effectively avoiding second strokes (PRoFESS) MRI substudy. Stroke. 2012;43:2336–2342. doi: 10.1161/STROKEAHA.111.648576
26.
Cavalieri M, Schmidt R, Chen C, Mok V, de Freitas GR, Song S, et al; VITATOPS Trial Study Group. B vitamins and magnetic resonance imaging-detected ischemic brain lesions in patients with recent transient ischemic attack or stroke: the Vitamins to Prevent Stroke (VITATOPS) MRI-substudy. Stroke. 2012;43:3266–3270. doi: 10.1161/STROKEAHA.112.665703
27.
ten Dam VH, van den Heuvel DM, van Buchem MA, Westendorp RG, Bollen EL, Ford I, et al; PROSPER Study Group. Effect of pravastatin on cerebral infarcts and white matter lesions. Neurology. 2005;64:1807–1809. doi: 10.1212/01.WNL.0000161844.00797.73
28.
Mok VC, Lam WW, Fan YH, Wong A, Ng PW, Tsoi TH, et al. Effects of statins on the progression of cerebral white matter lesion: post hoc analysis of the ROCAS (Regression of Cerebral Artery Stenosis) study. J Neurol. 2009;256:750–757. doi: 10.1007/s00415-009-5008-7
29.
Launer LJ, Miller ME, Williamson JD, Lazar RM, Gerstein HC, Murray AM, et al; ACCORD MIND Investigators. Effects of intensive glucose lowering on brain structure and function in people with type 2 diabetes (ACCORD MIND): a randomised open-label substudy. Lancet Neurol. 2011;10:969–977. doi: 10.1016/S1474-4422(11)70188-0
30.
Bath PM, Wardlaw JM. Pharmacological treatment and prevention of cerebral small vessel disease: a review of potential interventions. Int J Stroke. 2015;10:469–478. doi: 10.1111/ijs.12466
31.
Han SW, Song TJ, Bushnell CD, Lee SS, Kim SH, Lee JH, et al. Cilostazol decreases cerebral arterial pulsatility in patients with mild white matter hyperintensities: subgroup analysis from the Effect of Cilostazol in Acute Lacunar Infarction Based on Pulsatility Index of Transcranial Doppler (ECLIPse) study. Cerebrovasc Dis. 2014;38:197–203. doi: 10.1159/000365840
32.
Blair GW, Appleton JP, Flaherty K, Doubal F, Sprigg N, Dooley R, et al. Tolerability, safety and intermediary pharmacological effects of cilostazol and isosorbide mononitrate, alone and combined, in patients with lacunar ischaemic stroke: the Lacunar Intervention-1 (LACI-1) trial, a randomised clinical trial. EClinicalMedicine. 2019;11:34–43. doi: 10.1016/j.eclinm.2019.04.001
33.
Liu-Ambrose T, Best JR, Davis JC, Eng JJ, Lee PE, Jacova C, et al. Aerobic exercise and vascular cognitive impairment: a randomized controlled trial. Neurology. 2016;87:2082–2090. doi: 10.1212/WNL.0000000000003332
34.
Dao E, Barha CK, Best JR, Hsiung GY, Tam R, Liu-Ambrose T. The effect of aerobic exercise on white matter hyperintensity progression may vary by sex. Can J Aging. 2019;38:236–244. doi: 10.1017/S0714980818000582
35.
Venkatraman VK, Sanderson A, Cox KL, Ellis KA, Steward C, Phal PM, et al. Effect of a 24-month physical activity program on brain changes in older adults at risk of Alzheimer’s disease: the AIBL active trial. Neurobiol Aging. 2019;pii:S0197–S4580(19)30155-1. doi: 10.1016/j.neurobiolaging.2019.02.030
36.
Bolandzadeh N, Tam R, Handy TC, Nagamatsu LS, Hsu CL, Davis JC, et al. Resistance training and white matter lesion progression in older women: exploratory Analysis of a 12-Month Randomized Controlled Trial. J Am Geriatr Soc. 2015;63:2052–2060. doi: 10.1111/jgs.13644
37.
Zhang H, Cui Y, Zhao Y, Dong Y, Duan D, Wang J, et al. Effects of sartans and low-dose statins on cerebral white matter hyperintensities and cognitive function in older patients with hypertension: a randomized, double-blind and placebo-controlled clinical trial. Hypertens Res. 2019;42:717–729. doi: 10.1038/s41440-018-0165-7
38.
Croall ID, Tozer DJ, Moynihan B, Khan U, O’Brien JT, Morris RG, et al; PRESERVE Study Team. Effect of standard vs intensive blood pressure control on cerebral blood flow in small vessel disease: the PRESERVE randomized clinical trial. JAMA Neurol. 2018;75:720–727. doi: 10.1001/jamaneurol.2017.5153
39.
Croall ID, Lohner V, Moynihan B, Khan U, Hassan A, O’Brien JT, et al. Using DTI to assess white matter microstructure in cerebral small vessel disease (SVD) in multicentre studies. Clin Sci (Lond). 2017;131:1361–1373. doi: 10.1042/CS20170146
40.
Jia J, Wei C, Liang J, Zhou A, Zuo X, Song H, et al. The effects of DL-3-n-butylphthalide in patients with vascular cognitive impairment without dementia caused by subcortical ischemic small vessel disease: a multicentre, randomized, double-blind, placebo-controlled trial. Alzheimers Dement. 2016;12:89–99. doi: 10.1016/j.jalz.2015.04.010
41.
Pauls MMH, Clarke N, Trippier S, Betteridge S, Howe FA, Khan U, et al. Perfusion by Arterial Spin Labelling Following Single Dose Tadalafil in Small Vessel Disease (PASTIS): study protocol for a randomised controlled trial. Trials. 2017;18:229. doi: 10.1186/s13063-017-1973-9
42.
Mi T, Yu F, Ji X, Sun Y, Qu D. The interventional effect of remote ischemic preconditioning on cerebral small vessel disease: a pilot randomized clinical trial. Eur Neurol. 2016;76:28–34. doi: 10.1159/000447536
43.
Wang Y, Meng R, Song H, Liu G, Hua Y, Cui D, et al. Remote ischemic conditioning may improve outcomes of patients with cerebral small-vessel disease. Stroke. 2017;48:3064–3072. doi: 10.1161/STROKEAHA.117.017691
44.
Leurent C, Goodman JA, Zhang Y, He P, Polimeni JR, Gurol ME, et al; Ponezumab Trial Study Group. Immunotherapy with ponezumab for probable cerebral amyloid angiopathy. Ann Clin Transl Neurol. 2019;6:795–806. doi: 10.1002/acn3.761
45.
Greenberg SM, Rosand J, Schneider AT, Creed Pettigrew L, Gandy SE, Rovner B, et al. A phase 2 study of tramiprosate for cerebral amyloid angiopathy. Alzheimer Dis Assoc Disord. 2006;20:269–274. doi: 10.1097/01.wad.0000213845.28624.f4
46.
Kennedy G, Hardman RJ, Macpherson H, Scholey AB, Pipingas A. How does exercise reduce the rate of age-associated cognitive decline? A review of potential mechanisms. J Alzheimers Dis. 2017;55:1–18. doi: 10.3233/JAD-160665
47.
Birns J, Markus H, Kalra L. Blood pressure reduction for vascular risk: is there a price to be paid? Stroke. 2005;36:1308–1313. doi: 10.1161/01.STR.0000165901.38039.5f
48.
Dawson J, Broomfield N, Dani K, Dickie DA, Doney A, Forbes K, et al. Xanthine Oxidase Inhibition for the Improvement of Long-Term Outcomes Following Ischaemic Stroke and Transient Ischaemic Attack (XILO-FIST) - protocol for a randomised double blind placebo-controlled clinical trial. Eur Stroke J. 2018;3:281–290. doi: 10.1177/2396987318771426
49.
Lee SJ, Lee JS, Choi MH, Lee SE, Shin DH, Hong JM. Cilostazol improves endothelial function in acute cerebral ischemia patients: a double-blind placebo controlled trial with flow-mediated dilation technique. BMC Neurol. 2017;17:169. doi: 10.1186/s12883-017-0950-y
50.
Toyoda K, Uchiyama S, Yamaguchi T, Easton JD, Kimura K, Hoshino H, et al; CSPS.com Trial Investigators. Dual antiplatelet therapy using cilostazol for secondary prevention in patients with high-risk ischaemic stroke in Japan: a multicentre, open-label, randomised controlled trial. Lancet Neurol. 2019;18:539–548. doi: 10.1016/S1474-4422(19)30148-6
51.
Kim BJ, Lee EJ, Kwon SU, Park JH, Kim YJ, Hong KS, et al; PICASSO Investigators. Prevention of cardiovascular events in Asian patients with ischaemic stroke at high risk of cerebral haemorrhage (PICASSO): a multicentre, randomised controlled trial. Lancet Neurol. 2018;17:509–518. doi: 10.1016/S1474-4422(18)30128-5
52.
Edrissi H, Schock SC, Cadonic R, Hakim AM, Thompson CS. Cilostazol reduces blood brain barrier dysfunction, white matter lesion formation and motor deficits following chronic cerebral hypoperfusion. Brain Res. 2016;1646:494–503. doi: 10.1016/j.brainres.2016.06.036
53.
Ohtomo R, Bannai T, Ohtomo G, Shindo A, Tomimoto H, Tsuji S, et al. Cilostazol alleviates white matter degeneration caused by chronic cerebral hypoperfusion in mice: implication of its mechanism from gene expression analysis. Neurosci Lett. 2018;662:247–252. doi: 10.1016/j.neulet.2017.10.045
54.
Fujita Y, Lin JX, Takahashi R, Tomimoto H. Cilostazol alleviates cerebral small-vessel pathology and white-matter lesions in stroke-prone spontaneously hypertensive rats. Brain Res. 2008;1203:170–176. doi: 10.1016/j.brainres.2008.01.103
55.
Watanabe T, Zhang N, Liu M, Tanaka R, Mizuno Y, Urabe T. Cilostazol protects against brain white matter damage and cognitive impairment in a rat model of chronic cerebral hypoperfusion. Stroke. 2006;37:1539–1545. doi: 10.1161/01.STR.0000221783.08037.a9
56.
Lee JH, Park SY, Shin YW, Hong KW, Kim CD, Sung SM, et al. Neuroprotection by cilostazol, a phosphodiesterase type 3 inhibitor, against apoptotic white matter changes in rat after chronic cerebral hypoperfusion. Brain Res. 2006;1082:182–191. doi: 10.1016/j.brainres.2006.01.088
57.
Honda F, Imai H, Ishikawa M, Kubota C, Shimizu T, Fukunaga M, et al. Cilostazol attenuates gray and white matter damage in a rodent model of focal cerebral ischemia. Stroke. 2006;37:223–228. doi: 10.1161/01.STR.0000196977.76702.6d
58.
Xiong Z, Lu W, Zhu L, Zeng L, Shi C, Jing Z, et al. Dl-3-n-Butylphthalide treatment enhances hemodynamics and ameliorates memory deficits in rats with chronic cerebral hypoperfusion. Front Aging Neurosci. 2017;9:238. doi: 10.3389/fnagi.2017.00238
59.
Pauls MM, Moynihan B, Barrick TR, Kruuse C, Madigan JB, Hainsworth AH, et al. The effect of phosphodiesterase-5 inhibitors on cerebral blood flow in humans: a systematic review. J Cereb Blood Flow Metab. 2018;38:189–203. doi: 10.1177/0271678X17747177
60.
Hess DC, Blauenfeldt RA, Andersen G, Hougaard KD, Hoda MN, Ding Y, et al. Remote ischaemic conditioning-a new paradigm of self-protection in the brain. Nat Rev Neurol. 2015;11:698–710. doi: 10.1038/nrneurol.2015.223
61.
Khan MB, Hafez S, Hoda MN, Baban B, Wagner J, Awad ME, et al. Chronic remote ischemic conditioning is cerebroprotective and induces vascular remodeling in a VCID model. Transl Stroke Res. 2018;9:51–63. doi: 10.1007/s12975-017-0555-1
62.
Conklin J, Silver FL, Mikulis DJ, Mandell DM. Are acute infarcts the cause of leukoaraiosis? Brain mapping for 16 consecutive weeks. Ann Neurol. 2014;76:899–904. doi: 10.1002/ana.24285
63.
Aisen PS, Gauthier S, Ferris SH, Saumier D, Haine D, Garceau D, et al. Tramiprosate in mild-to-moderate Alzheimer’s disease - a randomized, double-blind, placebo-controlled, multi-centre study (the Alphase Study). Arch Med Sci. 2011;7:102–111. doi: 10.5114/aoms.2011.20612
64.
Regenhardt RW, Das AS, Lo EH, Caplan LR. Advances in understanding the pathophysiology of lacunar stroke: a review. JAMA Neurol. 2018;75:1273–1281. doi: 10.1001/jamaneurol.2018.1073
65.
Staals J, Makin SD, Doubal FN, Dennis MS, Wardlaw JM. Stroke subtype, vascular risk factors, and total MRI brain small-vessel disease burden. Neurology. 2014;83:1228–1234. doi: 10.1212/WNL.0000000000000837
66.
Hachinski V, Iadecola C, Petersen RC, Breteler MM, Nyenhuis DL, Black SE, et al. National Institute of Neurological Disorders and Stroke-Canadian Stroke Network vascular cognitive impairment harmonization standards. Stroke. 2006;37:2220–2241. doi: 10.1161/01.STR.0000237236.88823.47
67.
Brookes RL, Hollocks MJ, Tan RY, Morris RG, Markus HS. Brief screening of vascular cognitive impairment in patients with cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy without dementia. Stroke. 2016;47:2482–2487. doi: 10.1161/STROKEAHA.116.013761
68.
Lawrence AJ, Brookes RL, Zeestraten EA, Barrick TR, Morris RG, Markus HS. Pattern and rate of cognitive decline in cerebral small vessel disease: a Prospective Study. PLoS One. 2015;10:e0135523. doi: 10.1371/journal.pone.0135523
69.
Pearce LA, McClure LA, Anderson DC, Jacova C, Sharma M, Hart RG, et al; SPS3 Investigators. Effects of long-term blood pressure lowering and dual antiplatelet treatment on cognitive function in patients with recent lacunar stroke: a secondary analysis from the SPS3 randomised trial. Lancet Neurol. 2014;13:1177–1185. doi: 10.1016/S1474-4422(14)70224-8
70.
Chappell FM, Del Carmen Valdés Hernández M, Makin SD, Shuler K, Sakka E, Dennis MS, et al. Sample size considerations for trials using cerebral white matter hyperintensity progression as an intermediate outcome at 1 year after mild stroke: results of a prospective cohort study. Trials. 2017;18:78. doi: 10.1186/s13063-017-1825-7
71.
Schmidt R, Berghold A, Jokinen H, Gouw AA, van der Flier WM, Barkhof F, et al; LADIS Study Group. White matter lesion progression in LADIS: frequency, clinical effects, and sample size calculations. Stroke. 2012;43:2643–2647. doi: 10.1161/STROKEAHA.112.662593
72.
Wiseman SJ, Meijboom R, Valdés Hernández MDC, Pernet C, Sakka E, Job D, et al. Longitudinal multi-centre brain imaging studies: guidelines and practical tips for accurate and reproducible imaging endpoints and data sharing. Trials. 2019;20:21. doi: 10.1186/s13063-018-3113-6
73.
Smith EE, Biessels GJ, De Guio F, de Leeuw FE, Duchesne S, Düring M, et al. Harmonizing brain magnetic resonance imaging methods for vascular contributions to neurodegeneration. Alzheimers Dement (Amst). 2019;11:191–204. doi: 10.1016/j.dadm.2019.01.002
74.
Duering M, Konieczny MJ, Tiedt S, Baykara E, Tuladhar AM, Leijsen EV, et al. Serum neurofilament light chain levels are related to small vessel disease burden. J Stroke. 2018;20:228–238. doi: 10.5853/jos.2017.02565
75.
Wardlaw JM, Smith EE, Biessels GJ, Cordonnier C, Fazekas F, Frayne R, et al; Standards for Reporting Vascular Changes on Neuroimaging (STRIVE v1). Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol. 2013;12:822–838. doi: 10.1016/S1474-4422(13)70124-8

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Versions

You are viewing the most recent version of this article.

History

Received: 10 July 2019
Revision received: 18 September 2019
Accepted: 2 October 2019
Published online: 22 November 2019
Published in print: January 2020

Permissions

Request permissions for this article.

Keywords

  1. cerebral small vessel diseases
  2. dementia, vascular
  3. leukoaraiosis
  4. risk factors
  5. stroke
  6. lacunar

Subjects

Authors

Affiliations

Eric E. Smith, MD, MPH [email protected]
From the Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Alberta, Canada (E.E.S.)
Hugh S. Markus, FMedSci
Department of Clinical Neurosciences, Cambridge University, United Kingdom (H.S.M.).

Notes

The online-only Data Supplement is available with this article at Supplemental Material.
Correspondence to Eric E. Smith, MD, MPH, University of Calgary, Room 2941, Health Sciences Bldg, 3330 Hospital Dr NW, Calgary, AB T2N 4N1, Canada. Email [email protected]

Disclosures

Dr Smith reports consulting for Alnylam Pharmaceuticals and Portola Pharmaceuticals. The other author reports no conflicts.

Sources of Funding

Dr Smith is supported by the Katthy Taylor Chair in Vascular Dementia from the University of Calgary. He has received funding from the Canadian Institutes of Health Research and Brain Canada. H.S. Markus is supported by the National Institutes of Health Research (NIHR) Senior Investigator award. His research is supported by infrastructural support from the Cambridge University Hospital Trust NIHR Biomedical Research Centre. He has received grant funding the Medical Research Council, British Heart Foundation, Alzheimer’s Research UK, and Stroke Association and personal fees from BIBA Medical Publishing. The views expressed are those of the authors and not necessarily those of the NIHR or the Department of Health and Social Care.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. The added value of b0-DWI analysis in the diagnosis of cavitating lacunes when T2-weighted spin-echo is unavailable, European Journal of Radiology, 183, (111924), (2025).https://doi.org/10.1016/j.ejrad.2025.111924
    Crossref
  2. The emerging role of nitric oxide in the synaptic dysfunction of vascular dementia, Neural Regeneration Research, 20, 2, (402-415), (2024).https://doi.org/10.4103/NRR.NRR-D-23-01353
    Crossref
  3. Cognitive Impairment in Cerebral Small Vessel Disease Is Associated with Corpus Callosum Microstructure Changes Based on Diffusion MRI, Diagnostics, 14, 16, (1838), (2024).https://doi.org/10.3390/diagnostics14161838
    Crossref
  4. Diffusion tensor imaging in cerebral small vessel disease applications: opportunities and challenges, Frontiers in Neuroscience, 18, (2024).https://doi.org/10.3389/fnins.2024.1473462
    Crossref
  5. Development and validation of a nomogram predictive model for cerebral small vessel disease: a comprehensive retrospective analysis, Frontiers in Neurology, 14, (2024).https://doi.org/10.3389/fneur.2023.1340492
    Crossref
  6. Quantifying neuroinflammation within deep gray matter in small vessel disease using diffusion tensor based free-water imaging: a longitudinal study, Frontiers in Aging Neuroscience, 16, (2024).https://doi.org/10.3389/fnagi.2024.1361436
    Crossref
  7. Progress of Clinical Research on Silent Brain Infarction in Traditional Chinese Medicine and Western Medicine, Advances in Clinical Medicine, 14, 03, (1666-1672), (2024).https://doi.org/10.12677/acm.2024.143892
    Crossref
  8. European stroke organisation (ESO) guideline on cerebral small vessel disease, part 2, lacunar ischaemic stroke, European Stroke Journal, 9, 1, (5-68), (2024).https://doi.org/10.1177/23969873231219416
    Crossref
  9. Does Thrombosis Play a Causal Role in Lacunar Stroke and Cerebral Small Vessel Disease?, Stroke, 55, 4, (934-942), (2024)./doi/10.1161/STROKEAHA.123.044937
    Abstract
  10. Advances in Cerebral Small Vessel Disease: Sandra E. Black Lecture to the Canadian Neurological Sciences Federation, Canadian Journal of Neurological Sciences / Journal Canadien des Sciences Neurologiques, 52, 1, (1-8), (2024).https://doi.org/10.1017/cjn.2024.26
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

New Treatment Approaches to Modify the Course of Cerebral Small Vessel Diseases
Stroke
  • Vol. 51
  • No. 1

Purchase access to this journal for 24 hours

Stroke
  • Vol. 51
  • No. 1
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response