Your activity: 6 p.v.

COVID-19: Neurologic complications and management of neurologic conditions

COVID-19: Neurologic complications and management of neurologic conditions
Authors:
Brett L Cucchiara, MD
Igor J Koralnik, MD
Section Editors:
José Biller, MD, FACP, FAAN, FAHA
Alejandro A Rabinstein, MD
Scott E Kasner, MD
Deputy Editor:
Richard P Goddeau, Jr, DO, FAHA
Literature review current through: Dec 2022. | This topic last updated: Apr 07, 2022.

INTRODUCTION — Coronaviruses are important human and animal pathogens. At the end of 2019, a novel coronavirus was identified as the cause of a cluster of pneumonia cases in Wuhan, a city in the Hubei Province of China. The disease rapidly spread, and, in February 2020, the World Health Organization designated the disease coronavirus disease 2019 (COVID-19) [1]. The World Health Organization declared COVID-19 a pandemic on March 11, 2020 [2]. The virus that causes COVID-19 is designated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).

Understanding of COVID-19 is evolving. Interim guidance has been issued by the World Health Organization and, in the United States, by the Centers for Disease Control and Prevention [2,3] and the National Institutes of Health COVID-19 Treatment Guidelines Panel [4]. Links to these and other related society guidelines are found elsewhere. (See 'Society guideline links' below.)

Neurologic complications in patients with COVID-19 are common in hospitalized patients [5-12]. More than 80 percent of hospitalized patients may have neurologic symptoms at some point during their disease course [12]. Rates vary by geographical location and patient characteristics. Myalgias, headache, encephalopathy, and dizziness may be most common, occurring in approximately one-third of patients in China, Europe, and the United States [5,8,12-14]. Neurologic symptoms such as dysgeusia or anosmia may be less common, but accurate ascertainment of symptoms may be limited in patients with severe cognitive or cardiorespiratory dysfunction [13,14]. Stroke, movement disorders, motor and sensory deficits, ataxia, and seizures appear uncommon [12,13]. Critically ill patients have a higher proportion of neurologic complications than patients with less severe illness [5,12]. As the frequency and severity of acute presentations of COVID-19 have decreased due to vaccination and social distancing strategies, some patients have reported persisting neurologic symptoms such as cognitive dysfunction, headache, and numbness [15].

This topic will review the neurologic complications of COVID-19 and the management of patients with neurologic conditions who develop COVID-19. Other aspects of COVID-19 are described separately:

(See "COVID-19: Epidemiology, virology, and prevention".)

(See "COVID-19: Clinical features".)

(See "COVID-19: Diagnosis".)

(See "COVID-19: Management in hospitalized adults".)

(See "COVID-19: Management of the intubated adult".)

(See "COVID-19: Clinical manifestations and diagnosis in children".)

(See "COVID-19: Evaluation and management of adults with persistent symptoms following acute illness ("Long COVID")".)

(See "COVID-19: Vaccine-induced immune thrombotic thrombocytopenia (VITT)".)

NEUROPATHOGENESIS — The underlying mechanisms of neurologic complications in patients with COVID-19 are diverse and, in some cases, multifactorial. Neurologic complications arise most frequently from systemic response to the infection [13,16]. Distinct mechanisms include:

Neurologic injury from systemic dysfunction – Hypoxemia, prevalent in patients with severe COVID-19, is likely to play a role in many patients with encephalopathy, as are metabolic derangements due to organ failure and medication effects. Neurochemical evidence of astrocytic and neuronal injury documented in plasma of patients with moderate and severe COVID-19 does not suggest a specific pathogenesis [17].

Neuropathologic case series of patients who succumbed to COVID-19 revealed acute hypoxic ischemic damage in nearly all patients, as well as the presence of hemorrhagic and bland infarcts, microglial activation with microglial nodules, and neuronophagia in many [16,18]. In other series, neuroimaging findings appeared consistent with a delayed posthypoxic leukoencephalopathy and are similar to those described in patients with acute respiratory distress syndrome (ARDS) unrelated to COVID-19 [19-21]. (See 'Evaluation and management' below and 'Encephalopathy' below.)

Renin-angiotensin system dysfunction – Maladaptive activity of the renin-angiotensin system (RAS) may be another relevant pathophysiologic mechanism of COVID-19 infection. SARS-CoV-2 utilizes angiotensin converting enzyme 2 (ACE2), a membrane-bound protein, as its point of entry into cells. ACE2 functions to convert angiotensin II into angiotensin-(1-7), which has vasodilator, antiproliferative, and antifibrotic properties [22,23]. By binding to ACE2, the SARS-CoV-2 virus may damage vascular endothelial cells by inhibiting mitochondrial function and endothelial nitric oxide synthetase activity resulting in secondary cardio- and cerebrovascular effects [24]. (See 'Cerebrovascular disease' below.)

Immune dysfunction – A dysregulated systemic immune response to SARS-CoV-2 has been implicated [25,26].

Proinflammatory state – Critically ill patients with COVID-19 often develop signs of severe systemic inflammation consistent with a cytokine release syndrome-like presentation that manifests with persistent fever, elevated inflammatory markers (eg, D-dimer, ferritin), and elevated proinflammatory cytokines [27,28]. Markers of inflammation (eg, peripheral tumor necrosis factor [TNF], TNF-alpha, and interleukin 6 [IL-6]) are elevated in patients with severe COVID-19 [29,30]. (See "COVID-19: Management in hospitalized adults", section on 'IL-6 pathway inhibitors (eg, tocilizumab)'.)

High levels of circulating proinflammatory cytokines can cause confusion and alteration of consciousness [9,31]. A single case series described five patients with delayed awakening after ventilation for COVID-19-related ARDS who underwent brain vessel wall magnetic resonance imaging (MRI), which revealed abnormal contrast enhancement in the vascular wall of the basal skull arteries, a finding interpreted as possible endotheliitis [32]. However, evidence of resolution of these imaging abnormalities after corticosteroid treatment was not presented nor was there pathological confirmation of inflammation. (See 'Encephalopathy' below.)

A proinflammatory state also may be associated with thrombophilia ("thromboinflammation"), increasing risk of stroke and other thrombotic events [33]. Complement activation may also lead to thrombotic microvascular injury in patients with severe COVID-19 [34]. (See 'Cerebrovascular disease' below.)

Cytokine release may also lead to brain injury by microglial activation and a systemic inflammatory response. In case series and reports, microglial nodules and neuronophagia were found in brain tissue without evidence of direct viral invasion [16,35]. Microglial activation to phagocytose hypoxic neurons has been seen with other viral infections.

Parainfectious and postinfectious triggers – The timing of symptoms relative to initial symptoms of COVID-19 infection suggests that Guillain-Barré occurs as a parainfectious rather than a postinfectious complication in most patients. In one case, weakness preceded the onset of fever and respiratory symptoms [36]. Other cases report a longer interval between the onset of viral illness and weakness, consistent with its occurrence as a postinfectious complication. (See 'Guillain-Barré syndrome' below.)

Direct viral invasion of the nervous system – Some reports provide evidence for direct viral invasion of the nervous system [37-39]. In postmortem case series, SARS-CoV-2 was detected in most brain specimens, but these findings were unrelated to the severity of neuropathological findings [16,37,38]. This suggests that neural injury may be due to a systemic inflammatory response triggered by the SARS-CoV-2 virus rather than the infection itself.

It is uncertain if SARS-CoV-2 directly infects the cerebral vessels. Autopsy studies have reported potential evidence of direct endothelial invasion by the SARS-CoV-2 virus with a possible associated endotheliitis in the lung, heart, kidney, liver, and small intestine [40,41]. However, this remains controversial, as structures on electron microscopy thought to represent viral particles in the endothelium of blood vessels in the kidney may actually be normal structures or artifacts [42,43]. Case reports suggest the possibility of multifocal ischemic and hemorrhagic lesions consistent with endothelial involvement, microthrombosis, or small vessel vasculitis, though pathology was not performed [44,45]. Neuropathological studies have not confirmed frank cerebral vasculitis with SARS-CoV-2 [16].

SMELL AND TASTE DISORDERS — Anosmia and dysgeusia have been reported as common early symptoms in patients with COVID-19, occurring in greater than 80 percent of patients in one series [46]. In a meta-analysis of 83 studies involving more than 27,000 patients, olfactory dysfunction was reported in 48 percent (95% CI 41.2-54.5) [47]. These symptoms may be an initial manifestation of COVID-19 and can occur in the absence of nasal congestion or discharge; however, these are rarely the only clinical manifestations of COVID-19. (See "COVID-19: Clinical features", section on 'Initial presentation'.)

MRI signal abnormalities in one or both olfactory bulbs have been described in patients with COVID-19, which can resolve on follow-up imaging [48-52]. In two autopsy cases, pathologic findings demonstrated inflammatory infiltrate and axonal injury in the olfactory tracts but could not determine whether direct viral damage was responsible [53]. Transient anosmia may be related to inflammatory changes in the sustentacular cells within the nasal epithelium rather than direct injury to the olfactory neurons [54]. In one MRI-based study of 20 patients with anosmia, edematous obstruction was identified in the olfactory cleft of the nasal cavities [55]. At one-month follow-up, olfactory function correlated with improvement of obstruction.

Robust data on long-term prognosis are lacking [56]. In one series, among the 33 percent of affected patients who had recovered olfactory function, the mean symptom duration was eight days [46]. In a survey of nonhospitalized patients with olfactory dysfunction from Italy, 83 percent reported complete recovery at a mean of 37 days after symptom onset [57]. Among 51 patients with anosmia who underwent objective olfactory testing, full recovery at four and eight months was reported in 84 and 96 percent, respectively [58]. In some patients, anosmia and dysgeusia may persist for several months, along with other neurologic or systemic symptoms after acute COVID-19 infection [59]. (See 'Persistent neurologic symptoms after COVID-19 infection' below.)

These symptoms can occur with other viral infections and other causes; an approach to evaluation is discussed separately. (See "Taste and olfactory disorders in adults: Evaluation and management".)

ENCEPHALOPATHY — Encephalopathy is common in critically ill patients with COVID-19. In a cohort study of 2088 patients with COVID-19 admitted to an intensive care unit, delirium was common, occurring in 55 percent [60]. In a study of 509 hospitalized COVID-19 patients, 31.8 percent had encephalopathy, and those patients were older than those without encephalopathy (66 versus 55 years), had a shorter time from symptom onset to hospitalization (6 versus 7 days), were more likely to be male, and were more likely to have risk factors (including a history of any neurologic disorder, cancer, cerebrovascular disease, chronic kidney disease, diabetes, dyslipidemia, heart failure, hypertension, or smoking) [12].

Encephalopathy may be the primary symptom of COVID-19. In a study of 817 older patients (median age 78 years) evaluated in the emergency department who were diagnosed with COVID-19 infection, encephalopathy was present in 28 percent [61]. Among those patients, 37 percent lacked typical COVID-19 symptoms such as fever or dyspnea. Risk factors for encephalopathy included older age, vision impairment, history of Parkinson disease or stroke, and prior psychoactive medication use.

Clinical, laboratory, and radiologic features — Patients with COVID-19 may develop prominent delirium and agitation requiring sedation; others manifest encephalopathy with somnolence and a decreased level of consciousness [5,6]. Corticospinal tract signs (eg, hyperreflexia, extensor plantar responses) are common; seizures are described along with encephalopathy in patients with COVID-19, just as they can occur in toxic-metabolic encephalopathy in other settings [62,63]. These signs and symptoms are described separately. (See "Diagnosis of delirium and confusional states", section on 'Clinical presentation'.)

In most cases, encephalopathy develops in patients who become critically ill. In exceptional cases, delirium may be an early, and even a presenting, feature [64,65]. Cases of transient global amnesia have also been reported [66,67]. It is unclear whether a prolonged confusional state may occur in COVID-19 in the absence of respiratory symptoms or hypoxia.

Patients with encephalopathy typically have no evidence of brain inflammation on neuroimaging studies or on cerebrospinal fluid (CSF) analysis, although there are exceptions.

MRI findings – A spectrum of neuroimaging abnormalities have been described in patients with COVID-19-related encephalopathy; some but not all of these findings indicate a specific, alternative diagnosis for the patient's mental state, such as stroke, encephalitis, reversible posterior leukoencephalopathy syndrome (RPLS), and others [6,19,52,68-70]. (See 'Other acute neurologic manifestations' below.)

In the aggregate, approximately half of neuroimaging studies in patients with COVID-19-related encephalopathy demonstrate an acute abnormality, the most common of which are acute ischemic stroke, cortical fluid-attenuated inversion recovery (FLAIR) signal abnormality, leptomeningeal enhancement (often subtle), and other manifestations of encephalitis [6,19,21,52,68,70,71].

As examples, one series reported results of MRI studies in 190 patients with severe COVID-19, most of whom had symptoms consistent with encephalopathy [68]. After excluding patients with ischemic stroke or chronic unrelated lesions, abnormalities were reported in 37 patients. Patterns of MRI abnormality included signal abnormality in the medial temporal lobe, multifocal white matter lesions visible on FLAIR and diffusion-weighted imaging with associated hemorrhage, and isolated white matter microhemorrhages. Hemorrhagic lesions were described exclusively in patients with ARDS. Another series described MRI abnormalities in 64 patients with COVID-19 and accompanying neurologic manifestations (mostly encephalopathy) [70]. Ischemic stroke was identified in 17 patients (27 percent), 10 of whom had focal or lateralizing signs on examination, which suggested possible stroke. MRI abnormalities in other patients included leptomeningeal enhancement in 17 percent and encephalitis in 13 percent; 46 percent of MRI studies were normal. Another case series described MRI findings in 115 hospitalized patients with COVID-19; 25 had cerebral microbleeds, often with concomitant leukoencephalopathy. These were most common in patients with more severe respiratory illness [21].

Cytotoxic lesions in the splenium of the corpus callosum have also been reported in adult patients with COVID-19-related encephalopathy [68,70,72,73] as well as in a few children with multisystem inflammatory syndrome in COVID-19 [69]. (See "COVID-19: Multisystem inflammatory syndrome in children (MIS-C) clinical features, evaluation, and diagnosis".)

Electroencephalography (EEG) findings – Patients with encephalopathy and COVID-19 who have undergone electroencephalography have typically demonstrated nonspecific findings [6,74,75].

Cerebrospinal fluid – Abnormal findings in the CSF are nonspecific and appear in a minority of cases, including patients presenting with acute neurologic symptoms [76]. Case series reported CSF analysis from 12 patients and revealed no white cells and negative reverse transcription polymerase chain reaction (RT-PCR) assays for SARS-CoV-2 in all [6,19]. A 2021 systemic review of individual reports and case series involving 430 patients identified SARS-CoV-2 in the CSF of only 17 patients (6 percent) [76]. Oligoclonal bands were found in 3 patients among the 132 patients tested.

Patients who have elevated white blood cell count in CSF should undergo further evaluation for encephalitis and other conditions. (See 'Other acute neurologic manifestations' below.)

Underlying causes and risk factors — The etiology of encephalopathy in patients with COVID-19 is often multifactorial. Critically ill patients with COVID-19 are subject to the same causes of encephalopathy as are other critically ill patients. Common causes of agitated or hypoactive delirium are varied and include toxic metabolic encephalopathy, medication effects, cerebrovascular disease, nonconvulsive seizures, and others as outlined in more detail in the table (table 1). Factors associated with a higher risk of delirium among patients with COVID-19 admitted to an intensive care unit included mechanical ventilation, vasopressor use, use of restraints, benzodiazepine or continuous opioid infusions, and lack of family visitation [60]. These underlying causes are described separately. (See "Acute toxic-metabolic encephalopathy in adults", section on 'Specific etiologies'.)

Other less common complications of COVID-19 may also produce altered mental status; these include ischemic or hemorrhagic stroke, encephalitis, posterior reversible encephalopathy syndrome (PRES), multisystem inflammatory syndrome, and postinfectious demyelinating disease. (See 'Cerebrovascular disease' below and 'Other acute neurologic manifestations' below.)

Evaluation and management — Patients with persistent encephalopathy that is not explained by sedating medications, hypoxemia, or other systemic factors may warrant further evaluation to rule out other causes. Patients with focal or lateralizing neurologic signs on examination should be evaluated with neuroimaging. For other patients, a decision to perform further testing depends on the clinical scenario and may include MRI with and without gadolinium, EEG to exclude subclinical seizures, and CSF sampling to rule out central nervous system infection. Patients with an elevated white blood cell count in CSF should undergo further evaluation for encephalitis and other conditions. The evaluation of patients with encephalopathy is described in detail separately. (See "Diagnosis of delirium and confusional states", section on 'Evaluation'.)

As with other causes of encephalopathy, the management is primarily directed toward the underlying disease, which is discussed separately. (See "COVID-19: Management in hospitalized adults" and "COVID-19: Management of the intubated adult".)

The symptomatic management of patients with COVID-19 and encephalopathy is the same as the management of other critically ill patients with delirium and is described separately. (See "Delirium and acute confusional states: Prevention, treatment, and prognosis", section on 'Management' and "Sedative-analgesic medications in critically ill adults: Selection, initiation, maintenance, and withdrawal".)

The role of glucocorticoids or other immunomodulatory therapies in the management of patients with COVID-19 and encephalopathy is uncertain. Case series have identified a small number of patients with severe encephalopathy who have improved neurologically after receiving glucocorticoids with or without plasma exchange [32,77]. In one series, mechanically ventilated patients with unresponsive wakefulness in whom sedation had been withdrawn were given methylprednisolone 1 g for five days followed by plasma exchange (5 to 10 sessions) [77]. Three of five patients improved neurologically within one week but two showed no improvement. However, glucocorticoids or other immunomodulatory therapies should not be considered routine therapeutic options for patients with COVID-19-related encephalopathy unless additional data emerge to help identify immunotherapy-responsive cases.

The role of glucocorticoids in the general management of severely ill patients with COVID-19 is discussed in further detail separately. (See "COVID-19: Management in hospitalized adults", section on 'Dexamethasone and other glucocorticoids'.)

Prognosis — Encephalopathy is a risk factor for poor outcome. In one study, hospitalized patients with COVID-19 and encephalopathy had longer lengths of stay, worse functional impairment at hospital discharge, and a higher 30-day mortality rate compared with those without encephalopathy (22 versus 3 percent) [12]. In another series, patients with normal MRI and CSF were most likely to recover earlier [78].

As with other critically ill patients, improvement in neurologic dysfunction may be delayed beyond the period when symptoms of the acute illness have resolved. Our clinical experience, as well as some published reports, suggest that some patients with a prolonged disorder of consciousness in the setting of severe COVID-19 may awaken up to several days following cessation of sedating medications and later recover [12,79-81]. In one series of 795 hospitalized patients with severe COVID-19 infection who were intubated for at least seven days, among the nearly 72 percent who eventually recovered consciousness, the median time to recovery of command-following was 30 days [82]. Variables associated with prolonged recovery included hypoxemia, duration of exposure to paralytic or sedative medications, age, and male sex. Approximately one-third of patients with encephalopathy and COVID-19 infection who subsequently recover remained subjectively cognitively impaired at the time of hospital discharge [6,12,83]. Thus, clinicians should exercise caution about withdrawal of life support measures in patients with encephalopathy in the absence of structural brain injury on neuroimaging or other evidence of futility.

CEREBROVASCULAR DISEASE

Epidemiology — − Stroke appears to be relatively infrequent in the setting of COVID-19 [84-87]. The incidence of ischemic stroke associated with COVID-19 in hospitalized patients has ranged from 0.4 to 2.7 percent, while the incidence of intracranial hemorrhage has ranged from 0.2 to 0.9 percent [5,88-99]. Cerebral venous thrombosis (CVT) has been reported in patients with COVID-19 infection [100-102]. A retrospective study of over 13,000 patients with COVID-19 identified 12 patients with CVT within 3 months, corresponding to an incidence of 8.8 per 10,000 patients [103]. In a systematic review involving 34,331 patients hospitalized with SARS-CoV-2 infection, the estimated frequency of CVT was 0.08 percent (95% CI 0.01-0.5) [104].

The risk of stroke may vary according to the severity of COVID-19. Early case series suggest that for patients with mild illness, the risk is <1 percent, while for patients in intensive care, the risk may be as high as 6 percent [5].

Most often, stroke occurs one to three weeks after onset of COVID-19 symptoms, although stroke has been the initial symptom leading to hospitalization in a minority of reported patients [90,92,105-107]. In a report that compared 86 patients with COVID-19 and imaging-confirmed stroke with 499 matched control patients who had stroke without COVID-19 one year before, COVID-19 was an independent risk factor for in-hospital stroke (odds ratio [OR] 20.9, 95% CI 10.4-42.0) [106].

The mean age of patients with COVID-19 and stroke appears similar to those without COVID-19. While some reports observed that ischemic stroke occurred in young patients with COVID-19 [100,108-110], including children [97,111,112], these appear to represent a minority of stroke cases associated with COVID-19. In a subsequent systematic analysis of 10 studies including 160 COVID-19 patients with ischemic stroke, the median age was 65 years [113]. In a United States national stroke registry that included 1143 patients with COVID-19 and acute stroke, the median age at presentation was 68 years compared with 71 years for patients without COVID-19 during the same period [114].

Risk factors and mechanisms

Traditional stroke risk factors – Most patients with ischemic stroke associated with COVID-19 are older patients with vascular risk factors [90,93,94,115]. Traditional stroke risk factors such as hypertension, hyperlipidemia, atrial fibrillation, and/or diabetes mellitus have been identified in these patients.

Hypercoagulability and proinflammatory state associated with infection – While several mechanisms of stroke related to COVID-19 have been postulated, thrombophilia associated with the virus or the host immune response appears to be one important mechanism, as suggested by elevated markers of hypercoagulability and inflammation [33].

Considerable evidence suggests that COVID-19 is associated with a hypercoagulable state. This is reflected in the extremely elevated D-dimer levels (a marker of clot turnover) observed in many patients over the course of the first few weeks of disease, particularly those who are more severely affected [116]. Such markedly elevated D-dimer levels appear to be present specifically in some patients with ischemic stroke as well [85,117]. D-dimer levels >10,000 ng/mL have been proposed to identify patients with cryptogenic stroke potentially attributable to COVID-19 hypercoagulability [118]. (See "COVID-19: Hypercoagulability".)

An association with antiphospholipid antibodies has also been observed with COVID-19, but the relative proportion of different antiphospholipid antibody subtypes and their pathogenicity are uncertain, and follow-up testing has been incomplete. In a series from Philadelphia, antiphospholipid antibodies testing was positive in six of eight patients with COVID-19 and stroke; all six had anticardiolipin antibodies only, while none were positive for beta-2 glycoprotein-1 antibodies or lupus anticoagulant [93]. Anticardiolipin antibodies appear to be relatively common in several other viral infectious diseases (such as HIV and hepatitis) but are not clearly correlated with an increased thrombotic risk with those infections [119].

Prior to COVID-19, some evidence suggested that serious infections could trigger acute stroke, potentially due to increased inflammation and consequent thrombosis [120-122]. For example, in the Cardiovascular Health Study, the risk of ischemic stroke increased following hospitalization for infection within the previous 30 days (OR 7.3, 95% CI 1.9-40.9) [121]. Influenza, sepsis, and minor respiratory and urinary tract infections were also associated with increased stroke risk in analyses of administrative datasets [123-125]. Coexisting high-risk features, such as valvular heart disease, congestive heart failure, renal failure, lymphoma, peripheral vascular disease, pulmonary circulatory disorders, and coagulopathy, may further increase the risk of stroke after sepsis [126].

COVID-19 appears to be associated with a higher risk of ischemic stroke compared with influenza. In a retrospective cohort study comparing patients with emergency department visits or hospitalizations for COVID-19 (n = 1916) or influenza (n = 1486), the incidence of ischemic stroke was higher among patients with COVID-19 (1.6 percent, versus 0.2 percent with influenza; adjusted odds ratio 7.6, 95% CI 2.3-25.2) [92].

Cardioembolism – Cardiac dysfunction associated with COVID-19 infection may also serve as a potential embolic stroke mechanism, either directly due to SARS-CoV-2 myocarditis or indirectly due to cardiac injury or dysfunction related to general critical illness. COVID-19 has been associated with several cardiac manifestations, including arrhythmia, heart failure, and myocardial infarction, many of which may predispose to cardioembolic stroke. (See "COVID-19: Evaluation and management of cardiac disease in adults" and "COVID-19: Myocardial infarction and other coronary artery disease issues".)

In one study of 100 consecutive COVID-19 patients evaluated with echocardiography, only 10 percent had any degree of left ventricular systolic dysfunction, suggesting that cardioembolism due to severe left heart failure is relatively uncommon [127].

Coagulopathy and anticoagulation – Cases of spontaneous intraparenchymal and cortical subarachnoid hemorrhage have been reported with coagulopathy or anticoagulation [52]. Some of these hemorrhages may represent unrecognized ischemic events with subsequent hemorrhagic conversion. In one report of 3824 hospitalized patients with COVID-19, intracerebral hemorrhage was reported in 33 (0.9 percent) [91]. Based upon the radiologic appearance, the investigators inferred that approximately three-quarters of these may have resulted from hemorrhagic transformation of ischemic stroke. Another report of 278 hospitalized patients with COVID-19 who had neuroimaging reported intracerebral hemorrhage in 10 patients. In both studies, most of the patients with intracerebral hemorrhage had been treated with full-dose anticoagulation [52,91]. (See "COVID-19: Hypercoagulability", section on 'Bleeding'.)

Intracranial hemorrhage with COVID-19 has also been associated with use of extracorporeal membrane oxygenation (ECMO) [128-130]. In an international registry, 145 of 2346 (6 percent) patients on ECMO with COVID-19 had an intracranial hemorrhage as of early October 2020 [129]. Patients on ECMO are also at increased risk of brain ischemia, including due to air embolism. (See "COVID-19: Extracorporeal membrane oxygenation (ECMO)", section on 'Complications and outcomes of ECMO for treatment of COVID-19'.)

Diagnostic evaluation

COVID-19 screening – During the pandemic, we recommend testing all patients with suspected stroke for COVID-19 at the time of admission [131]. This recommendation is based on the observation that many patients presenting with stroke may test positive even when systemic signs of infection are absent [90,92,105,106]. This approach ensures that appropriate isolation measures are implemented for patients testing positive and allows early recognition should systemic symptoms of COVID-19 appear. In addition, the presence of acute COVID-19 in a patient with stroke has implications for the underlying mechanism causing stroke, the long-term risk of recurrence, and potentially for the choice of optimal therapy both in the short and long term.

Etiologic testing – Given the frequent association of stroke in COVID-19 with typical vascular risk factors and traditional stroke mechanisms, the initial diagnostic approach should otherwise be similar to the approach generally used for all patients with suspected stroke. Diagnostic testing to identify underlying stroke mechanism should include brain and neurovascular imaging and cardiac evaluation, with treatment appropriate to the identified mechanism. (See "Initial assessment and management of acute stroke" and "Neuroimaging of acute stroke".)

Routine testing recommended for all patients hospitalized with COVID-19 includes a complete blood count (CBC), platelet count, prothrombin time (PT), activated partial thromboplastin time (aPTT), fibrinogen, and D-dimer. For patients without a defined mechanism for ischemic stroke associated with COVID-19, our approach to testing for a hypercoagulable state is otherwise similar to the approach used for patients without COVID-19. (See "COVID-19: Hypercoagulability", section on 'Routine testing' and "Overview of the evaluation of stroke", section on 'Hypercoagulable studies'.)

Other aspects of the evaluation of adults hospitalized with COVID-19 are discussed in detail elsewhere. (See "COVID-19: Management in hospitalized adults", section on 'Evaluation'.)

Management issues — The management of ischemic or hemorrhagic stroke in patients under investigation or those who are positive for COVID-19 should follow the same standards of care as for patients without COVID-19 but with necessary precautions related to infection control [132-134]. (See "Initial assessment and management of acute stroke" and "Approach to reperfusion therapy for acute ischemic stroke".)

Thrombolytic and reperfusion therapies – Evaluation for intravenous thrombolytic therapy should be undertaken as with any stroke patient. While the safety of intravenous tissue plasminogen activator (tPA; alteplase) has not been specifically studied in the setting of COVID-19, anecdotal data do not suggest obvious safety concerns [89,135].

Similar to the approach to intravenous thrombolysis, patients with ischemic stroke and COVID-19 should be evaluated for mechanical thrombectomy, as for any patient with acute ischemic stroke. Small cohort studies of mechanical thrombectomy in patients with COVID-19 and acute large vessel occlusion have reported varying results [84,109,110]. There may be an increased risk of reocclusion after initial recanalization in patients with COVID-19, potentially related to hypercoagulability associated with the infection. (See "Mechanical thrombectomy for acute ischemic stroke" and 'Risk factors and mechanisms' above.)

Acute antithrombotic therapy – For patients with ischemic stroke and an unambiguous indication for full-dose anticoagulation (eg, atrial fibrillation, severe heart failure), early initiation is probably reasonable given the high thrombotic risk seen in patients with COVID-19, provided the bleeding risk is tolerable. For other patients with ischemic stroke, early use of aspirin is generally indicated, regardless of COVID-19 infection status.

An individual assessment of the severity of systemic illness, presence of other potential thrombotic events, and bleeding risk should be considered in deciding on optimal antithrombotic therapy. (See "COVID-19: Hypercoagulability", section on 'Supporting evidence'.)

Management for patients with vaccine-induced thrombotic thrombocytopenia – For patients who develop cerebral venous thrombosis with thrombocytopenia after COVID-19 vaccination, anticoagulation with a non-heparin agent (eg, a direct oral anticoagulant) and intravenous immune globulin treatment have been suggested [136]. This is discussed in greater detail separately. (See "COVID-19: Vaccine-induced immune thrombotic thrombocytopenia (VITT)", section on 'Management'.)

Treatment with ACE inhibitors and ARBs – Patients receiving angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) should continue treatment with these agents if there is no other reason for discontinuation (eg, hypotension, acute kidney injury, need for permissive hypertension in acute phase of ischemic stroke). Initial speculation that patients with COVID-19 who are receiving these agents may be at increased risk for adverse outcomes has not been supported by findings from observational studies. This is discussed in detail elsewhere. (See "COVID-19: Issues related to acute kidney injury, glomerular disease, and hypertension", section on 'Renin angiotensin system inhibitors'.)

General management – The general management of adults hospitalized with COVID-19, including COVID-19-specific therapy, is reviewed in detail separately. (See "COVID-19: Management in hospitalized adults".)

Secondary stroke prevention – Long-term treatment of vascular risk factors (eg, hypertension, diabetes, hyperlipidemia, atrial fibrillation, smoking) and appropriate use of antithrombotics should be undertaken, as with any stroke patient. (See "Overview of secondary prevention of ischemic stroke".)

Long-term risks – The incidence of stroke may be elevated beyond the acute period of COVID-19 infection. In a study of approximately 5.8 million United States veterans, the incidence of stroke at one year was higher in patients with prior COVID-19 infection than controls (hazard ratio 1.52, 95% CI 1.4-1.6) [137]. Cerebrovascular risk was higher for patients hospitalized with COVID-19 than for patients with less severe symptoms. The etiology of this finding is uncertain and may include a proinflammatory state or endothelial dysfunction following infection or suboptimal follow-up and management of vascular risk factors during the pandemic.

Severity and prognosis — Stroke associated with COVID-19 may be more severe than stroke without COVID-19 [117,138]. In a report from a hospital in New York City, the median National Institutes of Health Stroke Scale (NIHSS) score was greater for patients with stroke and COVID-19 compared with contemporary control patients with stroke but without COVID-19 (NIHSS score 19 versus 8) [90]. In another study, with a pooled sample of patients with COVID-19 from 28 sites in 16 countries, the NIHSS score was higher among 174 patients with stroke and COVID-19 compared with propensity-matched stroke patients without COVID-19 from one of the centers (NIHSS score 10 versus 6) [139]. In addition, mortality and disability after ischemic stroke were higher among those with than without COVID-19 [117]. In one analysis, in-hospital mortality among 160 patients with COVID-19 and stroke was 34 percent [113]. This finding could reflect greater stroke severity and/or greater comorbidity from respiratory and other systemic complications of COVID-19 [90,139].

Limited data suggest that, in North America, prognosis for stroke associated with COVID-19 is worse for Black American patients. One retrospective study reported outcomes for 69 patients (27 Black individuals and 42 from other racial backgrounds) with acute stroke associated with COVID-19 from 14 hospitals in the United States and Canada [140]. Stroke severity on admission, as determined by the mean NIHSS score, was similar for Black Americans and Canadians compared with other groups (16.3 versus 14.9) but mortality was higher (56 versus 29 percent).

Preventive measures — For patients with a history of stroke or those at high risk, physicians should emphasize the following practices and recommendations, along with appropriate risk modifications and other secondary stroke prevention measures:

Patients with stroke symptoms should be advised to seek emergency help just as they would prior to the pandemic. Acute stroke remains a potentially disabling and fatal illness, and patients should seek optimal medical care, which has been shown to improve stroke outcomes, even during the pandemic. Emergency wards and hospitals have rapidly become adept at separating patients with COVID-19 from other patients, practicing universal precautions against the spread of infection, and limiting the risk to their patients. Experience shows that high-quality stroke care can be delivered during the pandemic [141].

Preventive measures are discussed in detail separately. (See "COVID-19: Epidemiology, virology, and prevention", section on 'Prevention'.)

Patients with cerebrovascular disease should consider having extra medications on hand in case of the need to quarantine at home or in case of a disruption in supply chains.

Routine outpatient visits can often be conducted safely and effectively using telehealth [142,143]. Building on developments in the use of rapid outpatient evaluation for transient ischemic attack (TIA) patients, telehealth follow up of patients with TIA may be safe and effective [144].

The benefits of vaccination to prevent the morbidity and mortality associated with COVID-19 infection greatly outweigh the risk of vaccine-associated immune thrombotic thrombocytopenia (VITT) [145]. The risk of thromboembolism from COVID-19 infection appears higher than the risk of VITT [104,146]. This is discussed in greater detail separately. (See "COVID-19: Vaccines", section on 'Thrombosis with thrombocytopenia' and 'Vaccination against COVID-19' below.)

NEUROMUSCULAR DISEASE

Guillain-Barré syndrome — Rare cases of Guillain-Barré syndrome (GBS) have been reported after COVID-19 infection [36,78,147-155]. However, a potential causal association of COVID-19 with the risk of GBS remains uncertain. A cohort study from the United Kingdom failed to show a specific association between GBS and COVID-19 infection [156]. In this study, the incidence of GBS between March and May of 2020 was lower than the same months in the preceding four years. This reduction was attributed to the reduced transmission of other infective triggers from societal lockdown measures during this time. Among approximately 1200 patients with COVID-19 admitted over a one-month period to three northern Italy hospitals, only five cases of GBS were identified [147,154] .

Clinical features – Most patients with GBS and COVID-19 presented with progressive, ascending limb weakness evolving over one to four days [147]. The interval between the onset of viral illness and the development of muscle weakness is 5 to 16 days, similar to that observed for other viral infections associated with GBS [157]. In a report of 11 patients in the International GBS Outcome Study (IGOS) who developed GBS after COVID-19 infection, sensorimotor features were found in 73 percent including facial palsy in 64 percent [154]. Other reports suggest that the symptoms appear to progress more rapidly and be more severe than is typical for GBS; in one series, three of five patients required mechanical ventilation [147]. However, it was difficult to distinguish respiratory failure due to GBS from that due to COVID-19-related lung disease. Dysautonomic features were not observed in this series.

Miller Fisher syndrome [52,158] and other bulbar variant forms of GBS [159] have also been described in patients with COVID-19. These clinical syndromes and the differential diagnosis of GBS are described separately. (See "Guillain-Barré syndrome in adults: Pathogenesis, clinical features, and diagnosis".)

Diagnostic studies – GBS should be considered in the setting of progressive limb weakness and also when chest imaging findings are not commensurate with the respiratory insufficiency. The evaluation and diagnostic criteria for GBS are described separately. (See "Guillain-Barré syndrome in adults: Pathogenesis, clinical features, and diagnosis".)

In one series of five patients with COVID-19, the following diagnostic test features were observed [147]:

Cerebrospinal fluid was typical of other GBS patients with low or absent white cell count. Most patients have elevated protein level, although two of five patients in one series had normal levels. No cerebrospinal fluid (CSF) samples were positive for SARS-CoV-2.

Electrodiagnostic studies were consistent with either the axonal variant of GBS (preserved distal motor latencies and velocity, absent F-wave, and fibrillations) or with a demyelinating process (prolonged distal motor latencies and conduction block) [157].

MRI showed nerve root enhancement in some but not all patients.

Management and prognosis – Patients with GBS in the setting of COVID-19 should be managed as are other patients with GBS. (See "Guillain-Barré syndrome in adults: Treatment and prognosis".)

In one series, all patients received treatment with intravenous immune globulin (IVIG); two received a second course of IVIG and one started plasma exchange [147]. At least one patient was walking independently at discharge, but information regarding long-term outcomes for these patients is still pending.

The possible risk of GBS after vaccination against COVID-19 is discussed in greater detail separately. (See "COVID-19: Vaccines", section on 'Guillain-Barre syndrome' and 'Vaccination against COVID-19' below.)

Other acute neuromuscular syndromes

Myositis – Because myalgia and fatigue are common symptoms in COVID-19, some speculate that COVID-19 may be associated with a viral myositis; however, conclusive evidence of this is lacking [160]. In Wuhan, 11 percent of patients were reported to have evidence of muscle injury with elevated creatine kinase (CK; >200 units/L) and/or myalgia [5]. Myalgia was a common complaint in a series from Italy [161]. Three case reports have described rhabdomyolysis with CK >12,000 units/L [162-164]. In one case, muscle biopsy in one patient with COVID-19 and myopathy showed perivascular inflammation and deposition of myxovirus resistance protein A, a type I interferon-inducible protein [164]. Toxic effects of type I interferonopathies are seen in tissue in response to viral infection.

Focal and multifocal neuropathies – Several peripheral nerve and plexus syndromes have been reported in patients with COVID-19. These include:

Facial nerve palsy [155,165]

Ocular motor neuropathies [52,166]

Lower cranial neuropathy (vagus, accessory, and hypoglossal; Tapia syndrome) [167,168]

Multiple cranial neuropathies [158,166]

Neuralgic amyotrophy [169,170]

Critical illness neuropathy and myopathy – This complication tends to develop later in the course of COVID-19 infection than does parainfectious GBS [160,171]. (See "Neuromuscular weakness related to critical illness".)

Peripheral nerve injuries after prone positioning – Patients placed in prone positioning for COVID-19-related ARDS may develop peripheral nerve, typically brachial plexus, injuries [78,172,173]. In one study of 83 patients admitted to a rehabilitation facility after severe COVID-19 infection, 12 (14.5 percent) were diagnosed with a peripheral nerve injury, 11 of whom had been placed in prone positioning [172]. Nerve injuries were most frequently axonal and in the upper limb. (See "Prone ventilation for adult patients with acute respiratory distress syndrome", section on 'Complications'.)

OTHER ACUTE NEUROLOGIC MANIFESTATIONS — Isolated case reports have described the following syndromes in patients with COVID-19:

Meningoencephalitis – Both viral and apparent autoimmune meningoencephalitis have been reported in patients with COVID-19. These complications are rare. After a prodrome of headaches, fatigue, and fever for a few days, a 24-year-old male presented with generalized seizures and altered mental status. MRI revealed signal abnormality in the right mesial temporal lobe. Cerebrospinal fluid (CSF) analysis revealed 12 mononuclear cells and 2 polymorphonuclear cells; reverse transcription polymerase chain reaction (RT-PCR) analysis detected SARS-CoV-2 in the CSF [174]. A second case with viral detection by polymerase chain reaction (PCR) in the CSF was reported in a 41-year-old female who presented with seizure and altered mental status along with lymphocytic pleocytosis in the CSF [175].

Other cases of meningoencephalitis have been reported in patients in whom CSF was either negative for SARS-CoV-2 [78,176-180] or not tested [181-183]. It is possible that such cases do reflect viral infection with false-negative PCR testing [9]. One of these patients had evidence of viral particles in the brain on autopsy, and SARS-CoV-2 was detected in brain tissue by RT-PCR [180].

An alternative autoimmune mechanism has been postulated for these cases. Some patients have clinical syndromes and MRI findings that appear similar to autoimmune encephalitis [78]. One patient was found to have anti-N-methyl-d-aspartate (anti-NMDA) receptor antibodies [179]. Many of these patients appeared to respond to immunomodulatory treatment with glucocorticoids [78,177], plasma exchange [178], and/or intravenous immunoglobulin [179].

Rhombencephalitis – Parainfectious complications including brainstem encephalitis or isolated cerebellitis have been reported in adults and children with COVID-19 infection [184-188]. An inflammatory cause has been suggested. Some patients with fulminant disease and brainstem compression received external ventricular drainage for hydrocephalus and have been treated empirically with glucocorticoids and lopinavir-ritonavir with good short-term outcome [186].

Acute disseminated encephalomyelitis (ADEM) and acute hemorrhagic necrotizing encephalopathy – A few case reports have described patients with clinical and neuroimaging findings consistent with ADEM [9,78,189-191]. Some patients have had myelitis with or without brain involvement [78]. An additional case report describes a similar syndrome, acute necrotizing encephalopathy, in a patient with COVID-19 [191]. (See "Acute disseminated encephalomyelitis (ADEM) in adults".)

Increasing numbers of patients with hemorrhagic encephalomyelitis are reported [50,78,192]. A case report described a female in her late fifties who presented with fever, cough, and altered mental status; MRI revealed hemorrhagic lesions in bilateral thalami, medial temporal lobes, and subinsular lesions [193]. Another case was reported with predominant brainstem involvement in a 59-year-old female who also suffered from aplastic anemia [194]. In another series, four of nine patients with ADEM had hemorrhagic change on MRI [78]. (See "Acute disseminated encephalomyelitis (ADEM) in adults", section on 'Acute hemorrhagic leukoencephalitis'.)

Treatment with high-dose steroids, intravenous immunoglobulin, and/or plasma exchange has been attempted in these cases with variable outcomes; many patients die or have substantial neurologic morbidity, particularly those with hemorrhagic lesions [9,78,189-191].

Multisystem inflammatory syndrome in children – Some children with COVID-19 develop a multisystem inflammatory syndrome, similar to incomplete Kawasaki disease, which can include neurocognitive symptoms (headache, lethargy, confusion); in four patients with this syndrome, MRI revealed signal abnormality in the splenium of the corpus callosum [69]. (See "COVID-19: Multisystem inflammatory syndrome in children (MIS-C) clinical features, evaluation, and diagnosis".)

Seizures and status epilepticus – Seizures and status epilepticus have been reported in patients with severe COVID-19 infection [195,196]. In one series of 32 patients with COVID-19 who presented to the hospital with seizures, 40 percent had no history of epilepsy or other central nervous system diagnoses [197]. In rare instances, seizures have been the presenting symptom for patients without signs of infection who have tested positive for COVID-19 [197,198]. A systematic review of case series and reports identified 47 patients with COVID-19 who developed status epilepticus [199]. Most patients had preceding respiratory symptoms and no history of prior seizures. Neuroimaging was abnormal in approximately half of patients and 4 patients had a positive RT-PCR for SARS-CoV-2 in the CSF.

Generalized myoclonus – One report describes three patients (ages 63 to 88 years) who developed generalized myoclonus as an apparent post-infectious complication of COVID-19 [200]. Patients were not severely ill at the time myoclonus developed, and the myoclonus could not be explained by hypoxia, metabolic cause, or drug effect. Patients were treated symptomatically with levetiracetam, valproate, clonazepam, and/or propofol sedation and appeared to recover gradually with immunotherapy (methylprednisolone and/or plasma exchange).

Reversible posterior leukoencephalopathy syndrome (RPLS) – RPLS has been reported in patients with COVID-19 and may be due to hypertension and renal failure in some [50,52,94,96,201-205]. In one neuroimaging case series, findings consistent with RPLS were seen in more than 1 percent [52]. (See "Reversible posterior leukoencephalopathy syndrome".)

Reversible cerebral vasoconstriction syndrome (RCVS) – RCVS has been reported in adults and children with COVID-19 infection [206-208]. Corresponding features on brain imaging of patients with a severe syndrome included subarachnoid hemorrhagic, intracerebral hemorrhage, and ischemic stroke. (See "Reversible cerebral vasoconstriction syndrome".)

PERSISTENT NEUROLOGIC SYMPTOMS AFTER COVID-19 INFECTION — Patients recovering from a severe illness or after hospitalization may report prolonged neurologic symptoms. Likewise, some patients report symptoms attributed to COVID-19 that persist for weeks to months after the acute infection [209,210]. Among patients recovering from a severe COVID-19 infection who were hospitalized, fatigue, dyspnea, memory impairment, and myalgias were most common [209-212]. This is discussed in greater detail separately. (See "COVID-19: Evaluation and management of adults with persistent symptoms following acute illness ("Long COVID")", section on 'COVID-19 recovery'.)

In addition, patients with milder acute COVID-19 symptoms who never required hospitalization for pneumonia or hypoxemia may also report persistent neurologic and systemic symptoms [15,213]. In a survey of 180 nonhospitalized patients with COVID-19, more than 50 percent reported having at least one persistent symptom at a mean of 125 days after symptom onset [214]. Fatigue and anosmia were reported most frequently, occurring in 24 percent of symptomatic patients. In a prospective study of 100 nonhospitalized patients with COVID-19 with neurologic symptoms that persisted for at least six weeks, the most frequently reported were "brain fog" (81 percent), headache (68 percent), numbness/tingling (60 percent), dysgeusia (59 percent), anosmia (55 percent), and myalgias (55 percent) [15]. Patients showed impairments in quality-of-life (cognitive and fatigue) domains, attention, and working memory compared with controls. There was no correlation between time from disease onset and subjective impression of recovery. (See "COVID-19: Evaluation and management of adults with persistent symptoms following acute illness ("Long COVID")", section on 'Persistent symptoms'.)

The constellation of persistent symptoms after acute COVID-19 infection has been described by various terms including "long COVID," "post-COVID syndrome," "post-acute sequelae of SARS-CoV-2 infection," and "post-COVID conditions" [215]. This umbrella term covers a wide range of lingering consequences that remain four or more weeks after SARS-CoV-2 infection, which have been reported to range between 5 and 80 percent of patients [210,216-223].

Systemic symptoms such as fatigue, myalgias, and dyspnea are most common after COVID-19 infection (table 2). In addition, neurologic symptoms reported in case series to persist after COVID-19 infection include:

Headache [15,224,225]

Anosmia/dysgeusia [15,57,58,226,227]

Cognitive dysfunction [224,228-230]

Autonomic dysfunction [231-233]

Insomnia or other sleep impairments [210,234]

In a retrospective analysis based on the medical records of more than 270,000 patients with COVID-19, 37 percent had symptoms in the postacute period between 90 and 180 days [224]. In another analysis of 1438 patients from China who were hospitalized for COVID-19 infection, the incidence of cognitive impairment was 12 percent at one-year follow-up [230]. Some post-COVID conditions may share similarities with other post-viral syndromes [15,235-239]. The evaluation and management of patients with symptoms after acute COVID-19 illness are discussed in greater detail separately. (See "COVID-19: Evaluation and management of adults with persistent symptoms following acute illness ("Long COVID")".)

The pathophysiology and the role of the brain in the etiology of these symptoms are uncertain. Neuronal changes in limbic and other brain regions have been reported in patients with a prior COVID-19 infection. Among 785 participants of the United Kingdom Biobank study with baseline and follow-up imaging, structural changes were likelier in those following COVID-19 infection [240]. Repeat imaging was performed at a mean of 141 days following COVID-19 infection. Patients with COVID-19 infection were found to have greater reductions in functional connectivity and structural domains (gray matter thickness in the orbitofrontal and parahippocampal cortices and global brain size) compared with controls. Brain changes were also correlated with impaired performance on cognitive testing. In another study, elevated levels of biomarkers associated with neuronal dysfunction were reported in patients with persistent neurologic symptoms following either severe or mild COVID-19 infection [241]. The persistence of these findings and their functional significance over the long term is uncertain. Further study is warranted to assess the role of these findings in patients with post-COVID neurologic symptoms.

MANAGEMENT OF PATIENTS WITH NEUROLOGIC CONDITIONS — Despite the lack of high-quality data, patients with baseline disabling neurologic disease and those on immunosuppressive therapy should be particularly vigilant about infection control measures including vaccination, social distancing, and mask-wearing.

Vaccination against COVID-19 — Patients with some neurologic conditions, including those taking immunosuppressive medications, may be at high risk for severe illness from COVID-19 (table 3). Based on available data from the general population, we encourage vaccination against COVID-19 to those without a contraindication as soon as it is available to them, in agreement with guidelines from the American Association of Neuromuscular and Electrodiagnostic Medicine and the National Multiple Sclerosis Society [242,243]. COVID-19 vaccination has been associated with a reduced risk of neurologic complications of COVID-19 infection, including stroke [244].

Immunosuppressive medications may reduce the immunogenicity and effectiveness of vaccination against COVID-19. Strategies to optimize effectiveness of vaccination against COVID-19 infection in patients taking immunosuppressive therapy are discussed separately. (See "COVID-19: Vaccines", section on 'Immunocompromised individuals'.)

The benefits of vaccination to prevent the morbidity and mortality associated with COVID-19 infection greatly outweigh the risks including the risks of vaccine-induced thrombotic thrombocytopenia (VITT) and Guillain-Barré syndrome (GBS) [145,245,246]. The risk of thromboembolism from COVID-19 infection appears higher than the risk of VITT [104,146]. Similarly, the risk of GBS appears higher in the setting of COVID-19 infection than after vaccination [155,247]. Because of the possible increased risk of GBS associated with the adenovirus vector Ad26.COV2.S (Janssen/Johnson & Johnson) and ChAdOx1 nCoV-19/AZD1222 (AstraZeneca) COVID-19 vaccines, for patients with a history of GBS, we suggest other available COVID-19 vaccines until additional data emerge. (See "COVID-19: Vaccines", section on 'Specific safety concerns' and "COVID-19: Vaccine-induced immune thrombotic thrombocytopenia (VITT)" and "Guillain-Barré syndrome in adults: Treatment and prognosis", section on 'Subsequent immunizations'.)

Vaccine-associated immune thrombotic thrombocytopenia – Cases of thrombotic events with thrombocytopenia, including cerebral venous thrombosis (CVT) with and without hemorrhage, have been reported in patients immunized with the adenovirus-vector ChAdOx1 nCoV-19/AZD122 (AstraZeneca COVID-19) and Ad26.COV2.S (Janssen COVID-19) vaccines [248-259]. In a cohort study of 95 patients in the United Kingdom with CVT occurring after COVID-19 vaccination, those with VITT were younger (47 versus 57 years) and less likely to have other thromboembolic risk factors (34 versus 56 percent) than those with CVT not attributed to VITT [260]. However, patients with VITT had more extensive intra- and extracranial thromboses and a higher mortality rate than those without (29 versus 4 percent). DNA from adenovirus vectors may bind to platelet factor 4 and trigger the production of autoantibodies in some individuals [136]. This autoimmune VITT syndrome occurs between 5 and 30 days post-vaccination. (See "COVID-19: Vaccine-induced immune thrombotic thrombocytopenia (VITT)".)

Guillain-Barré syndrome – Cases of GBS have been observed with the adenovirus vector Ad26.COV2.S (Janssen/Johnson & Johnson) and ChAdOx1 nCoV-19/AZD1222 (AstraZeneca) COVID-19 vaccines in the United States and Europe [261-264], although a causal link has not been established. In the United States, 123 cases occurring within six weeks of immunization with the Ad26.COV2.S vaccine were reported among 13.2 million administered doses [264]. The estimated rate of post-vaccination GBS was higher than the background rate at 8.4 versus 2 per 100,000 person-years (rate ratio 4.18, 95% CI 3.47-4.98). The median time to symptom onset after vaccination was 13 days, and the rates of hospitalization and respiratory failure were 94 and 14 percent, respectively. This finding has not been reported with other COVID-19 vaccines [265]. This possible risk is discussed in greater detail separately. (See "COVID-19: Vaccines", section on 'Guillain-Barre syndrome'.)

Further information regarding vaccination against COVID-19, including guidance on vaccination dosing and boosters for patients who may be immunocompromised due to active immunosuppressive therapy (table 4), is discussed elsewhere. (See "COVID-19: Vaccines", section on 'Approach to vaccination in the United States' and "COVID-19: Vaccines", section on 'Immunocompromised individuals'.)

Managing immunosuppressive therapy — Patients with neurologic disease who are treated with immunosuppressive therapy do not appear to be at increased risk of COVID-19 infection. In most cases, immunosuppressive therapy should be continued and should be discontinued only if severe COVID-19 infection develops [266-270]. The role of switching agents or amending treatment protocols may be appropriate for some patients and is determined by assessing individual risks.

Multiple sclerosis – Patients with multiple sclerosis treated with B cell-depleting anti-CD20 or sphingosine-1-phosphate receptor disease-modifying therapies (DMT) may have a lower antibody response to SARS-CoV-2 infection than those treated with other DMTs. Patients with multiple sclerosis should be given vaccination prior to starting an anti-CD20 DMT, if possible. For patients already on an anti-CD20 DMT, timing the infusion in selected stable patients to occur several weeks after vaccination may be used to improve humoral response and possibly vaccine effectiveness [271]. Decisions should be individualized based on disease severity and activity. DMTs for multiple sclerosis are discussed in greater detail separately. (See "Disease-modifying therapies for multiple sclerosis: Pharmacology, administration, and adverse effects".)

In a cohort of 119 patients with multiple sclerosis or neuromyelitis optica spectrum disorder and COVID-19 infection, the rate of seroconversion was lower in patients taking an anti-CD20 DMT than those taking another DMT (48 versus 86 percent) [272]. The interval between anti-CD20 infusion and COVID-19 infection was longer in patients who developed an antibody response compared with those who did not (mean 3.7 months versus 1.9 months). In addition, a reduced humoral response to vaccination against SARS-CoV-2 has been reported in patients treated with ocrelizumab [273-275]. However, T cell response was retained in these patients, suggesting some efficacy of vaccination. Additional data are needed on the effects of these immunotherapies on vaccination.

Myasthenia gravis – Immunosuppressive medications for patients with myasthenia gravis are typically continued to minimize the risk of neuromuscular deterioration. Alternative treatments may also be considered for some patients who develop COVID-19 while taking immunosuppressive therapy. As examples, immunoglobulin therapy, complement inhibitor therapy, and plasma exchange are not expected to increase the risk of COVID-19; however, such treatments are not appropriate in all patients and indiscriminate switching to these treatments is not advised [269].

In small observational studies, the effect of COVID-19 infection on patients with myasthenia gravis is variable. Respiratory failure and exacerbations have been reported in some cases [276-278]. However, other patients with myasthenia gravis well controlled with immunosuppressive agents who developed a COVID-19 infection had a mild course [279-281]. In a cohort of 93 patients with myasthenia gravis and COVID-19 infection, factors associated with a severe course (eg, hospitalization requiring respiratory support) included lower premorbid forced vital capacity, poorer functional status, older age, and higher-dose oral glucocorticoid treatment [282]. A physician-reported registry has been established to collect outcome data in patients with myasthenia gravis who develop a COVID-19 infection [276].

Additional advice specific to disease-modifying treatment of myasthenia gravis is presented separately. (See "Overview of the treatment of myasthenia gravis", section on 'Guidance during COVID-19 pandemic'.)

Risk for more severe COVID-19 illness — Because neurologic disease may worsen the prognosis of COVID-19, vaccination and other preventive measures are particularly important in these patients. (See "COVID-19: Epidemiology, virology, and prevention", section on 'Prevention'.)

Cerebrovascular disease – Several risk factors have been associated with the risk of severe infection with SARS-CoV-2 (table 3). Patients with a history of cardiovascular disease, including stroke, appear to have worse outcomes when infected with SARS-CoV-2 [212,283-287]. There are several potential reasons for this. Patients with cerebrovascular disease often have other cardiovascular and metabolic risk factors that make them susceptible to worse outcomes from COVID-19; hypertension, obesity, and diabetes all are associated with a more aggressive course and higher mortality among COVID-19 patients [288]. The pandemic has manifested many of the same disparities by race, ethnicity, education, and income that are seen with cerebrovascular disease [105,289]. Thus, the same cardiovascular conditions, including obesity and diabetes, that predispose minority and under-resourced populations to stroke likely make them more susceptible to complications of COVID-19 [290]. In addition, the upregulation of angiotensin-converting enzyme 2 (ACE2) in those on angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs) could lead to a more severe infection in patients taking those medications, although observational studies have not borne this out. (See "COVID-19: Clinical features", section on 'Risk factors for severe illness'.)

Epilepsy – Patients with a history of epilepsy appear to be at an elevated risk of poor outcome after COVID-19 infection. In a meta-analysis of 13 observational studies including more than 57,000 patients hospitalized with COVID-19 infection, the risk of poor outcome was higher in patients with epilepsy than those without (odds ratio 1.71, 95% CI 1.11-2.59) [291]. This association may be due to morbidity of seizures and status epilepticus, adverse effects attributed to antiseizure medications, or worsened control of epilepsy due to reduced health care utilization. (See 'Health care utilization' below.)

Neuromuscular weakness – Patients with baseline cardiac or respiratory dysfunction, those with severe neuromuscular weakness, and those with bulbar weakness due to other debilitating neurologic disease (eg, amyotrophic lateral sclerosis, multiple sclerosis) are likely to have a more severe course and also may not return to their prior baseline [160,267,292,293]. This is based on observations in acute infections other than COVID-19 but are likely to apply in this setting as well. One registry of patients with multiple sclerosis found an association between more severe disability (as measured by the Expanded Disability Status Scale [EDSS]) and more severe COVID-19 infection; age and obesity were also risk factors in patients with multiple sclerosis as they are in the general population [294].

Multiple sclerosis – The hospitalization rate may be elevated for patients with multiple sclerosis taking B cell-depleting DMTs. In a multinational cohort study that included 1683 patients with multiple sclerosis, those taking either ocrelizumab or rituximab were likelier to be hospitalized than those taking other disease-modifying therapies (adjusted odds ratio [aOR] 1.75, 95% CI 1.29-2.38; aOR 2.76, 95% CI 1.87-4.07) [295]. Both ocrelizumab and rituximab were also associated with higher risk of admission to an intensive care unit, but neither was associated with an elevated risk of death.

Patients with neurologic disorders may be at higher risk for readmission after hospitalization for COVID-19 infection. Among 509 patients initially hospitalized with COVID-19, hospital reevaluation including readmission within the following four months occurred in 22 percent [296]. Older age, medical comorbid conditions, and preexisting neurologic disorders were associated risk factors.

An increased risk of COVID-19 and higher mortality have been suggested for patients with dementia [297] and also those with epilepsy [291,298,299].

COVID-19-specific therapy — The indications for and approach to COVID-19-specific therapy in symptomatic patients vary by clinical setting and risk profile for severe illness. Some patients are at risk for severe illness due to neurologic conditions such as those with dementia or neuroinflammatory disorders treated with immunosuppressive medications (table 3). The approach to COVID-19-specific therapy is discussed separately. (See "COVID-19: Management in hospitalized adults", section on 'COVID-19-specific therapy' and "COVID-19: Management of adults with acute illness in the outpatient setting", section on 'Treatment with COVID-19-specific therapies'.)

Health care utilization — Telemedicine is being increasingly used during the pandemic to manage outpatients with chronic neurologic disease. In a survey of 143 patients with epilepsy, reduced availability of ambulatory care occurred in approximately one-third of patients and was associated with exacerbation of seizures [300]. Patients with neurologic diseases (such as epilepsy and migraine) should develop rescue treatment plans that can be administered at home, if appropriate [301]. (See "Telemedicine for adults", section on 'Telemedicine during COVID-19 pandemic'.)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: COVID-19 – Index of guideline topics".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or email these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: COVID-19 overview (The Basics)" and "Patient education: COVID-19 and pregnancy (The Basics)" and "Patient education: COVID-19 and children (The Basics)" and "Patient education: COVID-19 vaccines (The Basics)")

SUMMARY AND RECOMMENDATIONS

Neurologic manifestations of COVID-19 infection – Neurologic manifestations occur in approximately half of hospitalized COVID-19 patients. Myalgias, headache, and encephalopathy may be most common.

Encephalopathy is common in critically ill patients with COVID-19 occurring in approximately 30 to 55 percent of patients. Common causes include toxic metabolic encephalopathy, medication effects, cerebrovascular disease, and nonconvulsive seizures. (See 'Encephalopathy' above.)

Stroke has been associated with COVID-19 in approximately 1 to 3 percent of hospitalized patients, with higher rates in those with more severe COVID-19. Several stroke subtypes may occur, including ischemic stroke, intracranial hemorrhage, and cerebral venous sinus thrombosis. In addition to traditional stroke mechanisms, potential mechanisms of ischemic stroke related to COVID-19 include hypercoagulability, inflammation, renin-angiotensin-aldosterone system dysfunction, and cardiac dysfunction. (See 'Epidemiology' above.)

Cases of Guillain-Barré syndrome and related syndromes have been described in patients with COVID-19. In general, the evaluation and management of such patients is similar to those not associated with the pandemic. (See 'Guillain-Barré syndrome' above and "Guillain-Barré syndrome in adults: Pathogenesis, clinical features, and diagnosis" and "Guillain-Barré syndrome in adults: Treatment and prognosis".)

Rare neurologic manifestations of COVID-19 include meningoencephalitis, cerebellitis, acute disseminated encephalomyelitis, multisystem inflammatory syndrome, seizures, generalized myoclonus, and reversible posterior leukoencephalopathy. (See 'Other acute neurologic manifestations' above.)

Persisting neurologic symptoms after COVID-19 infection – Some patients report symptoms attributed to COVID-19 that persist for weeks to months after the acute infection. Other patients with milder acute COVID-19 symptoms who never required hospitalization may also report persistent neurologic and systemic symptoms. The constellation of persistent symptoms after acute COVID-19 infection that remain four or more weeks has been reported to range between 5 and 80 percent of patients. (See 'Persistent neurologic symptoms after COVID-19 infection' above and "COVID-19: Evaluation and management of adults with persistent symptoms following acute illness ("Long COVID")".)

Management of patients with neurologic conditions – Patients with neurologic disease who are treated with immunosuppressive therapy are not at increased risk of COVID-19, and such medical treatment should be continued in uninfected patients.

If patients on such medications become infected with SARS-CoV-2, we advise an individualized approach that considers the risk associated with the specific medication, the severity of the underlying neurologic illness, and the severity of COVID-19 illness. (See 'Management of patients with neurologic conditions' above.)

Patients with a history of cardiovascular disease, including stroke, appear to be at risk of worse outcomes due to COVID-19; this likely applies to other patients with debilitating neurologic disease. Such patients should be specifically advised to adhere to preventive measures. (See 'Management of patients with neurologic conditions' above and "COVID-19: Epidemiology, virology, and prevention", section on 'Prevention'.)

Vaccination for patients with neurologic conditions – We encourage vaccination against COVID-19 to those without a contraindication as soon as it is available to them because patients with some neurologic conditions may be at high risk for severe illness from COVID-19. (See 'Management of patients with neurologic conditions' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Mitchell SV Elkind, MD, MS, FAAN, MD, who contributed to earlier versions of this topic review.

  1. World Health Organization. Director-General's remarks at the media briefing on 2019-nCoV on 11 February 2020. http://www.who.int/dg/speeches/detail/who-director-general-s-remarks-at-the-media-briefing-on-2019-ncov-on-11-february-2020 (Accessed on February 12, 2020).
  2. World Health Organization. Novel Coronavirus (2019-nCoV) technical guidance. https://www.who.int/emergencies/diseases/novel-coronavirus-2019/technical-guidance (Accessed on June 19, 2022).
  3. Centers for Disease Control and Prevention. 2019 Novel coronavirus, Wuhan, China. Information for Healthcare Professionals. https://www.cdc.gov/coronavirus/2019-nCoV/hcp/index.html (Accessed on June 19, 2022).
  4. National Institutes of Health. Coronavirus Disease 2019 (COVID-19) Treatment Guidelines. https://covid19treatmentguidelines.nih.gov/ (Accessed on August 08, 2022).
  5. Mao L, Jin H, Wang M, et al. Neurologic Manifestations of Hospitalized Patients With Coronavirus Disease 2019 in Wuhan, China. JAMA Neurol 2020; 77:683.
  6. Helms J, Kremer S, Merdji H, et al. Neurologic Features in Severe SARS-CoV-2 Infection. N Engl J Med 2020; 382:2268.
  7. Montalvan V, Lee J, Bueso T, et al. Neurological manifestations of COVID-19 and other coronavirus infections: A systematic review. Clin Neurol Neurosurg 2020; 194:105921.
  8. Romero-Sánchez CM, Díaz-Maroto I, Fernández-Díaz E, et al. Neurologic manifestations in hospitalized patients with COVID-19: The ALBACOVID registry. Neurology 2020; 95:e1060.
  9. Koralnik IJ, Tyler KL. COVID-19: A Global Threat to the Nervous System. Ann Neurol 2020; 88:1.
  10. Xiong W, Mu J, Guo J, et al. New onset neurologic events in people with COVID-19 in 3 regions in China. Neurology 2020; 95:e1479.
  11. Herman C, Mayer K, Sarwal A. Scoping review of prevalence of neurologic comorbidities in patients hospitalized for COVID-19. Neurology 2020; 95:77.
  12. Liotta EM, Batra A, Clark JR, et al. Frequent neurologic manifestations and encephalopathy-associated morbidity in Covid-19 patients. Ann Clin Transl Neurol 2020; 7:2221.
  13. Pezzini A, Padovani A. Lifting the mask on neurological manifestations of COVID-19. Nat Rev Neurol 2020; 16:636.
  14. Chou SH, Beghi E, Helbok R, et al. Global Incidence of Neurological Manifestations Among Patients Hospitalized With COVID-19-A Report for the GCS-NeuroCOVID Consortium and the ENERGY Consortium. JAMA Netw Open 2021; 4:e2112131.
  15. Graham EL, Clark JR, Orban ZS, et al. Persistent neurologic symptoms and cognitive dysfunction in non-hospitalized Covid-19 "long haulers". Ann Clin Transl Neurol 2021; 8:1073.
  16. Thakur KT, Miller EH, Glendinning MD, et al. COVID-19 neuropathology at Columbia University Irving Medical Center/New York Presbyterian Hospital. Brain 2021; 144:2696.
  17. Kanberg N, Ashton NJ, Andersson LM, et al. Neurochemical evidence of astrocytic and neuronal injury commonly found in COVID-19. Neurology 2020; 95:e1754.
  18. Solomon IH, Normandin E, Bhattacharyya S, et al. Neuropathological Features of Covid-19. N Engl J Med 2020; 383:989.
  19. Kandemirli SG, Dogan L, Sarikaya ZT, et al. Brain MRI Findings in Patients in the Intensive Care Unit with COVID-19 Infection. Radiology 2020; 297:E232.
  20. Radmanesh A, Derman A, Lui YW, et al. COVID-19-associated Diffuse Leukoencephalopathy and Microhemorrhages. Radiology 2020; 297:E223.
  21. Agarwal S, Jain R, Dogra S, et al. Cerebral Microbleeds and Leukoencephalopathy in Critically Ill Patients With COVID-19. Stroke 2020; 51:2649.
  22. Strawn WB, Ferrario CM, Tallant EA. Angiotensin-(1-7) reduces smooth muscle growth after vascular injury. Hypertension 1999; 33:207.
  23. Ye M, Wysocki J, William J, et al. Glomerular localization and expression of Angiotensin-converting enzyme 2 and Angiotensin-converting enzyme: implications for albuminuria in diabetes. J Am Soc Nephrol 2006; 17:3067.
  24. Lei Y, Zhang J, Schiavon CR, et al. SARS-CoV-2 Spike Protein Impairs Endothelial Function via Downregulation of ACE 2. Circ Res 2021; 128:1323.
  25. Pilotto A, Padovani A, ENCOVID-BIO Network. Reply to the Letter "COVID-19-Associated Encephalopathy and Cytokine-Mediated Neuroinflammation". Ann Neurol 2020; 88:861.
  26. Muccioli L, Pensato U, Cani I, et al. COVID-19-Associated Encephalopathy and Cytokine-Mediated Neuroinflammation. Ann Neurol 2020; 88:860.
  27. Channappanavar R, Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol 2017; 39:529.
  28. Huang KJ, Su IJ, Theron M, et al. An interferon-gamma-related cytokine storm in SARS patients. J Med Virol 2005; 75:185.
  29. Chen G, Wu D, Guo W, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 2020; 130:2620.
  30. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020; 395:497.
  31. Fotuhi M, Mian A, Meysami S, Raji CA. Neurobiology of COVID-19. J Alzheimers Dis 2020; 76:3.
  32. Pugin D, Vargas MI, Thieffry C, et al. COVID-19-related encephalopathy responsive to high-dose glucocorticoids. Neurology 2020; 95:543.
  33. Connors JM, Levy JH. Thromboinflammation and the hypercoagulability of COVID-19. J Thromb Haemost 2020; 18:1559.
  34. Magro C, Mulvey JJ, Berlin D, et al. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: A report of five cases. Transl Res 2020; 220:1.
  35. Al-Dalahmah O, Thakur KT, Nordvig AS, et al. Neuronophagia and microglial nodules in a SARS-CoV-2 patient with cerebellar hemorrhage. Acta Neuropathol Commun 2020; 8:147.
  36. Zhao H, Shen D, Zhou H, et al. Guillain-Barré syndrome associated with SARS-CoV-2 infection: causality or coincidence? Lancet Neurol 2020; 19:383.
  37. Matschke J, Lütgehetmann M, Hagel C, et al. Neuropathology of patients with COVID-19 in Germany: a post-mortem case series. Lancet Neurol 2020; 19:919.
  38. Meinhardt J, Radke J, Dittmayer C, et al. Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nat Neurosci 2021; 24:168.
  39. Song E, Zhang C, Israelow B, et al. Neuroinvasion of SARS-CoV-2 in human and mouse brain. bioRxiv 2020.
  40. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020; 395:1417.
  41. Ackermann M, Verleden SE, Kuehnel M, et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N Engl J Med 2020; 383:120.
  42. Goldsmith CS, Miller SE, Martines RB, et al. Electron microscopy of SARS-CoV-2: a challenging task. Lancet 2020; 395:e99.
  43. Varga Z, Flammer AJ, Steiger P, et al. Electron microscopy of SARS-CoV-2: a challenging task - Authors' reply. Lancet 2020; 395:e100.
  44. Hanafi R, Roger PA, Perin B, et al. COVID-19 Neurologic Complication with CNS Vasculitis-Like Pattern. AJNR Am J Neuroradiol 2020; 41:1384.
  45. Keller E, Brandi G, Winklhofer S, et al. Large and Small Cerebral Vessel Involvement in Severe COVID-19: Detailed Clinical Workup of a Case Series. Stroke 2020; 51:3719.
  46. Lechien JR, Chiesa-Estomba CM, De Siati DR, et al. Olfactory and gustatory dysfunctions as a clinical presentation of mild-to-moderate forms of the coronavirus disease (COVID-19): a multicenter European study. Eur Arch Otorhinolaryngol 2020; 277:2251.
  47. Saniasiaya J, Islam MA, Abdullah B. Prevalence of Olfactory Dysfunction in Coronavirus Disease 2019 (COVID-19): A Meta-analysis of 27,492 Patients. Laryngoscope 2021; 131:865.
  48. Aragão MFVV, Leal MC, Cartaxo Filho OQ, et al. Anosmia in COVID-19 Associated with Injury to the Olfactory Bulbs Evident on MRI. AJNR Am J Neuroradiol 2020; 41:1703.
  49. Li CW, Syue LS, Tsai YS, et al. Anosmia and olfactory tract neuropathy in a case of COVID-19. J Microbiol Immunol Infect 2021; 54:93.
  50. Coolen T, Lolli V, Sadeghi N, et al. Early postmortem brain MRI findings in COVID-19 non-survivors. Neurology 2020; 95:e2016.
  51. Laurendon T, Radulesco T, Mugnier J, et al. Bilateral transient olfactory bulb edema during COVID-19-related anosmia. Neurology 2020; 95:224.
  52. Lin E, Lantos JE, Strauss SB, et al. Brain Imaging of Patients with COVID-19: Findings at an Academic Institution during the Height of the Outbreak in New York City. AJNR Am J Neuroradiol 2020; 41:2001.
  53. Kirschenbaum D, Imbach LL, Ulrich S, et al. Inflammatory olfactory neuropathy in two patients with COVID-19. Lancet 2020; 396:166.
  54. Bilinska K, Jakubowska P, Von Bartheld CS, Butowt R. Expression of the SARS-CoV-2 Entry Proteins, ACE2 and TMPRSS2, in Cells of the Olfactory Epithelium: Identification of Cell Types and Trends with Age. ACS Chem Neurosci 2020; 11:1555.
  55. Eliezer M, Hamel AL, Houdart E, et al. Loss of smell in patients with COVID-19: MRI data reveal a transient edema of the olfactory clefts. Neurology 2020; 95:e3145.
  56. Meng X, Deng Y, Dai Z, Meng Z. COVID-19 and anosmia: A review based on up-to-date knowledge. Am J Otolaryngol 2020; 41:102581.
  57. Paderno A, Mattavelli D, Rampinelli V, et al. Olfactory and Gustatory Outcomes in COVID-19: A Prospective Evaluation in Nonhospitalized Subjects. Otolaryngol Head Neck Surg 2020; 163:1144.
  58. Renaud M, Thibault C, Le Normand F, et al. Clinical Outcomes for Patients With Anosmia 1 Year After COVID-19 Diagnosis. JAMA Netw Open 2021; 4:e2115352.
  59. Augustin M, Schommers P, Stecher M, et al. Post-COVID syndrome in non-hospitalised patients with COVID-19: a longitudinal prospective cohort study. Lancet Reg Health Eur 2021; 6:100122.
  60. Pun BT, Badenes R, Heras La Calle G, et al. Prevalence and risk factors for delirium in critically ill patients with COVID-19 (COVID-D): a multicentre cohort study. Lancet Respir Med 2021; 9:239.
  61. Kennedy M, Helfand BKI, Gou RY, et al. Delirium in Older Patients With COVID-19 Presenting to the Emergency Department. JAMA Netw Open 2020; 3:e2029540.
  62. Somani S, Pati S, Gaston T, et al. De Novo Status Epilepticus in patients with COVID-19. Ann Clin Transl Neurol 2020; 7:1240.
  63. Lyons S, O'Kelly B, Woods S, et al. Seizure with CSF lymphocytosis as a presenting feature of COVID-19 in an otherwise healthy young man. Seizure 2020; 80:113.
  64. Beach SR, Praschan NC, Hogan C, et al. Delirium in COVID-19: A case series and exploration of potential mechanisms for central nervous system involvement. Gen Hosp Psychiatry 2020; 65:47.
  65. Alkeridy WA, Almaghlouth I, Alrashed R, et al. A Unique Presentation of Delirium in a Patient with Otherwise Asymptomatic COVID-19. J Am Geriatr Soc 2020; 68:1382.
  66. Nishizawa T, Kawakami A, Taguchi T, Osugi Y. Transient global amnesia with bilateral hippocampal lesions during the COVID-19 global outbreak. J Gen Fam Med 2021; 22:154.
  67. Hussein HM. Transient Global Amnesia as a Possible First Manifestation of COVID-19. Neurohospitalist 2021; 11:84.
  68. Kremer S, Lersy F, de Sèze J, et al. Brain MRI Findings in Severe COVID-19: A Retrospective Observational Study. Radiology 2020; 297:E242.
  69. Abdel-Mannan O, Eyre M, Löbel U, et al. Neurologic and Radiographic Findings Associated With COVID-19 Infection in Children. JAMA Neurol 2020; 77:1440.
  70. Kremer S, Lersy F, Anheim M, et al. Neurologic and neuroimaging findings in patients with COVID-19: A retrospective multicenter study. Neurology 2020; 95:e1868.
  71. Larvie M, Lev MH, Hess CP. More on Neurologic Features in Severe SARS-CoV-2 Infection. N Engl J Med 2020; 382:e110.
  72. Klironomos S, Tzortzakakis A, Kits A, et al. Nervous System Involvement in Coronavirus Disease 2019: Results from a Retrospective Consecutive Neuroimaging Cohort. Radiology 2020; 297:E324.
  73. Chougar L, Shor N, Weiss N, et al. Retrospective Observational Study of Brain MRI Findings in Patients with Acute SARS-CoV-2 Infection and Neurologic Manifestations. Radiology 2020; 297:E313.
  74. Gélisse P, Rossetti AO, Genton P, et al. How to carry out and interpret EEG recordings in COVID-19 patients in ICU? Clin Neurophysiol 2020; 131:2023.
  75. Vespignani H, Colas D, Lavin BS, et al. Report on Electroencephalographic Findings in Critically Ill Patients with COVID-19. Ann Neurol 2020; 88:626.
  76. Lewis A, Frontera J, Placantonakis DG, et al. Cerebrospinal fluid in COVID-19: A systematic review of the literature. J Neurol Sci 2021; 421:117316.
  77. Cao A, Rohaut B, Le Guennec L, et al. Severe COVID-19-related encephalitis can respond to immunotherapy. Brain 2020; 143:e102.
  78. Paterson RW, Brown RL, Benjamin L, et al. The emerging spectrum of COVID-19 neurology: clinical, radiological and laboratory findings. Brain 2020; 143:3104.
  79. Fischer D, Threlkeld ZD, Bodien YG, et al. Intact Brain Network Function in an Unresponsive Patient with COVID-19. Ann Neurol 2020; 88:851.
  80. Abdo WF, Broerse CI, Grady BP, et al. Prolonged Unconsciousness Following Severe COVID-19. Neurology 2021; 96:e1437.
  81. Fischer D, Snider SB, Barra ME, et al. Disorders of Consciousness Associated With COVID-19: A Prospective Multimodal Study of Recovery and Brain Connectivity. Neurology 2022; 98:e315.
  82. Waldrop G, Safavynia SA, Barra ME, et al. Prolonged Unconsciousness is Common in COVID-19 and Associated with Hypoxemia. Ann Neurol 2022; 91:740.
  83. Rogers JP, Chesney E, Oliver D, et al. Psychiatric and neuropsychiatric presentations associated with severe coronavirus infections: a systematic review and meta-analysis with comparison to the COVID-19 pandemic. Lancet Psychiatry 2020; 7:611.
  84. Oxley TJ, Mocco J, Majidi S, et al. Large-Vessel Stroke as a Presenting Feature of Covid-19 in the Young. N Engl J Med 2020; 382:e60.
  85. Beyrouti R, Adams ME, Benjamin L, et al. Characteristics of ischaemic stroke associated with COVID-19. J Neurol Neurosurg Psychiatry 2020; 91:889.
  86. Bekelis K, Missios S, Ahmad J, et al. Ischemic Stroke Occurs Less Frequently in Patients With COVID-19: A Multicenter Cross-Sectional Study. Stroke 2020; 51:3570.
  87. Qureshi AI, Baskett WI, Huang W, et al. Acute Ischemic Stroke and COVID-19: An Analysis of 27 676 Patients. Stroke 2021; 52:905.
  88. Klok FA, Kruip MJHA, van der Meer NJM, et al. Confirmation of the high cumulative incidence of thrombotic complications in critically ill ICU patients with COVID-19: An updated analysis. Thromb Res 2020; 191:148.
  89. Lodigiani C, Iapichino G, Carenzo L, et al. Venous and arterial thromboembolic complications in COVID-19 patients admitted to an academic hospital in Milan, Italy. Thromb Res 2020; 191:9.
  90. Yaghi S, Ishida K, Torres J, et al. SARS-CoV-2 and Stroke in a New York Healthcare System. Stroke 2020; 51:2002.
  91. Dogra S, Jain R, Cao M, et al. Hemorrhagic stroke and anticoagulation in COVID-19. J Stroke Cerebrovasc Dis 2020; 29:104984.
  92. Merkler AE, Parikh NS, Mir S, et al. Risk of Ischemic Stroke in Patients With Coronavirus Disease 2019 (COVID-19) vs Patients With Influenza. JAMA Neurol 2020.
  93. Rothstein A, Oldridge O, Schwennesen H, et al. Acute Cerebrovascular Events in Hospitalized COVID-19 Patients. Stroke 2020; 51:e219.
  94. Requena M, Olivé-Gadea M, Muchada M, et al. COVID-19 and Stroke: Incidence and Etiological Description in a High-Volume Center. J Stroke Cerebrovasc Dis 2020; 29:105225.
  95. Cantador E, Núñez A, Sobrino P, et al. Incidence and consequences of systemic arterial thrombotic events in COVID-19 patients. J Thromb Thrombolysis 2020; 50:543.
  96. Hernández-Fernández F, Sandoval Valencia H, Barbella-Aponte RA, et al. Cerebrovascular disease in patients with COVID-19: neuroimaging, histological and clinical description. Brain 2020; 143:3089.
  97. LaRovere KL, Riggs BJ, Poussaint TY, et al. Neurologic Involvement in Children and Adolescents Hospitalized in the United States for COVID-19 or Multisystem Inflammatory Syndrome. JAMA Neurol 2021; 78:536.
  98. Coronado Munoz A, Tasayco J, Morales W, et al. High incidence of stroke and mortality in pediatric critical care patients with COVID-19 in Peru. Pediatr Res 2022; 91:1730.
  99. Leasure AC, Khan YM, Iyer R, et al. Intracerebral Hemorrhage in Patients With COVID-19: An Analysis From the COVID-19 Cardiovascular Disease Registry. Stroke 2021; 52:e321.
  100. Sweid A, Hammoud B, Bekelis K, et al. Cerebral ischemic and hemorrhagic complications of coronavirus disease 2019. Int J Stroke 2020; 15:733.
  101. Hughes C, Nichols T, Pike M, et al. Cerebral Venous Sinus Thrombosis as a Presentation of COVID-19. Eur J Case Rep Intern Med 2020; 7:001691.
  102. Tu TM, Goh C, Tan YK, et al. Cerebral Venous Thrombosis in Patients with COVID-19 Infection: a Case Series and Systematic Review. J Stroke Cerebrovasc Dis 2020; 29:105379.
  103. Al-Mufti F, Amuluru K, Sahni R, et al. Cerebral Venous Thrombosis in COVID-19: A New York Metropolitan Cohort Study. AJNR Am J Neuroradiol 2021; 42:1196.
  104. Baldini T, Asioli GM, Romoli M, et al. Cerebral venous thrombosis and severe acute respiratory syndrome coronavirus-2 infection: A systematic review and meta-analysis. Eur J Neurol 2021; 28:3478.
  105. Khatana SAM, Groeneveld PW. Health Disparities and the Coronavirus Disease 2019 (COVID-19) Pandemic in the USA. J Gen Intern Med 2020; 35:2431.
  106. Katz JM, Libman RB, Wang JJ, et al. Cerebrovascular Complications of COVID-19. Stroke 2020; 51:e227.
  107. Katsoularis I, Fonseca-Rodríguez O, Farrington P, et al. Risk of acute myocardial infarction and ischaemic stroke following COVID-19 in Sweden: a self-controlled case series and matched cohort study. Lancet 2021; 398:599.
  108. Majidi S, Fifi JT, Ladner TR, et al. Emergent Large Vessel Occlusion Stroke During New York City's COVID-19 Outbreak: Clinical Characteristics and Paraclinical Findings. Stroke 2020; 51:2656.
  109. Escalard S, Maïer B, Redjem H, et al. Treatment of Acute Ischemic Stroke due to Large Vessel Occlusion With COVID-19: Experience From Paris. Stroke 2020; 51:2540.
  110. Wang A, Mandigo GK, Yim PD, et al. Stroke and mechanical thrombectomy in patients with COVID-19: technical observations and patient characteristics. J Neurointerv Surg 2020; 12:648.
  111. Appavu B, Deng D, Dowling MM, et al. Arteritis and Large Vessel Occlusive Strokes in Children After COVID-19 Infection. Pediatrics 2021; 147.
  112. Mirzaee SMM, Gonçalves FG, Mohammadifard M, et al. Focal Cerebral Arteriopathy in a Pediatric Patient with COVID-19. Radiology 2020; 297:E274.
  113. Fridman S, Bres Bullrich M, Jimenez-Ruiz A, et al. Stroke risk, phenotypes, and death in COVID-19: Systematic review and newly reported cases. Neurology 2020; 95:e3373.
  114. Srivastava PK, Zhang S, Xian Y, et al. Acute Ischemic Stroke in Patients With COVID-19: An Analysis From Get With The Guidelines-Stroke. Stroke 2021; 52:1826.
  115. de Havenon A, Ney JP, Callaghan B, et al. Characteristics and Outcomes Among US Patients Hospitalized for Ischemic Stroke Before vs During the COVID-19 Pandemic. JAMA Netw Open 2021; 4:e2110314.
  116. Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 2020; 395:1054.
  117. Perry RJ, Smith CJ, Roffe C, et al. Characteristics and outcomes of COVID-19 associated stroke: a UK multicentre case-control study. J Neurol Neurosurg Psychiatry 2021; 92:242.
  118. Esenwa C, Cheng NT, Luna J, et al. Biomarkers of Coagulation and Inflammation in COVID-19-Associated Ischemic Stroke. Stroke 2021; 52:e706.
  119. García-Carrasco M, Galarza-Maldonado C, Mendoza-Pinto C, et al. Infections and the antiphospholipid syndrome. Clin Rev Allergy Immunol 2009; 36:104.
  120. Elkind MS. Why now? Moving from stroke risk factors to stroke triggers. Curr Opin Neurol 2007; 20:51.
  121. Elkind MS, Carty CL, O'Meara ES, et al. Hospitalization for infection and risk of acute ischemic stroke: the Cardiovascular Health Study. Stroke 2011; 42:1851.
  122. Cowan LT, Alonso A, Pankow JS, et al. Hospitalized Infection as a Trigger for Acute Ischemic Stroke: The Atherosclerosis Risk in Communities Study. Stroke 2016; 47:1612.
  123. Boehme AK, Luna J, Kulick ER, et al. Influenza-like illness as a trigger for ischemic stroke. Ann Clin Transl Neurol 2018; 5:456.
  124. Boehme AK, Ranawat P, Luna J, et al. Risk of Acute Stroke After Hospitalization for Sepsis: A Case-Crossover Study. Stroke 2017; 48:574.
  125. Smeeth L, Thomas SL, Hall AJ, et al. Risk of myocardial infarction and stroke after acute infection or vaccination. N Engl J Med 2004; 351:2611.
  126. Shao IY, Elkind MSV, Boehme AK. Risk Factors for Stroke in Patients With Sepsis and Bloodstream Infections. Stroke 2019; 50:1046.
  127. Szekely Y, Lichter Y, Taieb P, et al. Spectrum of Cardiac Manifestations in COVID-19: A Systematic Echocardiographic Study. Circulation 2020; 142:342.
  128. Zahid MJ, Baig A, Galvez-Jimenez N, Martinez N. Hemorrhagic stroke in setting of severe COVID-19 infection requiring Extracorporeal Membrane Oxygenation (ECMO). J Stroke Cerebrovasc Dis 2020; 29:105016.
  129. Extracorporeal Life Support Organization. ECMO in COVID-19. Available at: https://www.elso.org/Registry/FullCOVID19RegistryDashboard.aspx (Accessed on October 01, 2020).
  130. Usman AA, Han J, Acker A, et al. A Case Series of Devastating Intracranial Hemorrhage During Venovenous Extracorporeal Membrane Oxygenation for COVID-19. J Cardiothorac Vasc Anesth 2020; 34:3006.
  131. Barlinn K, Siepmann T, Pallesen LP, et al. Universal laboratory testing for SARS-CoV-2 in hyperacute stroke during the COVID-19 pandemic. J Stroke Cerebrovasc Dis 2020; 29:105061.
  132. Dafer RM, Osteraas ND, Biller J. Acute Stroke Care in the Coronavirus Disease 2019 Pandemic. J Stroke Cerebrovasc Dis 2020; 29:104881.
  133. Leira EC, Russman AN, Biller J, et al. Preserving stroke care during the COVID-19 pandemic: Potential issues and solutions. Neurology 2020; 95:124.
  134. Wira CR, Goyal M, Southerland AM, et al. Pandemic Guidance for Stroke Centers Aiding COVID-19 Treatment Teams. Stroke 2020; 51:2587.
  135. Co COC, Yu JRT, Laxamana LC, David-Ona DIA. Intravenous Thrombolysis for Stroke in a COVID-19 Positive Filipino Patient, a Case Report. J Clin Neurosci 2020; 77:234.
  136. Furie KL, Cushman M, Elkind MSV, et al. Diagnosis and Management of Cerebral Venous Sinus Thrombosis With Vaccine-Induced Immune Thrombotic Thrombocytopenia. Stroke 2021; 52:2478.
  137. Xie Y, Xu E, Bowe B, Al-Aly Z. Long-term cardiovascular outcomes of COVID-19. Nat Med 2022; 28:583.
  138. Katsanos AH, Palaiodimou L, Zand R, et al. The Impact of SARS-CoV-2 on Stroke Epidemiology and Care: A Meta-Analysis. Ann Neurol 2021; 89:380.
  139. Ntaios G, Michel P, Georgiopoulos G, et al. Characteristics and Outcomes in Patients With COVID-19 and Acute Ischemic Stroke: The Global COVID-19 Stroke Registry. Stroke 2020; 51:e254.
  140. Dmytriw AA, Phan K, Schirmer C, et al. Ischaemic stroke associated with COVID-19 and racial outcome disparity in North America. J Neurol Neurosurg Psychiatry 2020; 91:1362.
  141. Rudilosso S, Laredo C, Vera V, et al. Acute Stroke Care Is at Risk in the Era of COVID-19: Experience at a Comprehensive Stroke Center in Barcelona. Stroke 2020; 51:1991.
  142. Roy B, Nowak RJ, Roda R, et al. Teleneurology during the COVID-19 pandemic: A step forward in modernizing medical care. J Neurol Sci 2020; 414:116930.
  143. AHA/ASA Stroke Council Leadership. Temporary Emergency Guidance to US Stroke Centers During the Coronavirus Disease 2019 (COVID-19) Pandemic: On Behalf of the American Heart Association/American Stroke Association Stroke Council Leadership. Stroke 2020; 51:1910.
  144. Chang BP, Rostanski S, Willey J, et al. Safety and Feasibility of a Rapid Outpatient Management Strategy for Transient Ischemic Attack and Minor Stroke: The Rapid Access Vascular Evaluation-Neurology (RAVEN) Approach. Ann Emerg Med 2019; 74:562.
  145. Mahase E. AstraZeneca vaccine: Blood clots are "extremely rare" and benefits outweigh risks, regulators conclude. BMJ 2021; 373:n931.
  146. Thrombosis with thrombocytopenia syndrome following Janssen COVID-19 vaccine https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2021-04-23/03-COVID-Shimabukuro-508.pdf (Accessed on May 02, 2021).
  147. Toscano G, Palmerini F, Ravaglia S, et al. Guillain-Barré Syndrome Associated with SARS-CoV-2. N Engl J Med 2020; 382:2574.
  148. Scheidl E, Canseco DD, Hadji-Naumov A, Bereznai B. Guillain-Barré syndrome during SARS-CoV-2 pandemic: A case report and review of recent literature. J Peripher Nerv Syst 2020; 25:204.
  149. Sedaghat Z, Karimi N. Guillain Barre syndrome associated with COVID-19 infection: A case report. J Clin Neurosci 2020; 76:233.
  150. Virani A, Rabold E, Hanson T, et al. Guillain-Barré Syndrome associated with SARS-CoV-2 infection. IDCases 2020; 20:e00771.
  151. Alberti P, Beretta S, Piatti M, et al. Guillain-Barré syndrome related to COVID-19 infection. Neurol Neuroimmunol Neuroinflamm 2020; 7.
  152. Abu-Rumeileh S, Abdelhak A, Foschi M, et al. Guillain-Barré syndrome spectrum associated with COVID-19: an up-to-date systematic review of 73 cases. J Neurol 2021; 268:1133.
  153. Fragiel M, Miró Ò, Llorens P, et al. Incidence, clinical, risk factors and outcomes of Guillain-Barré in Covid-19. Ann Neurol 2021; 89:598.
  154. Luijten LWG, Leonhard SE, van der Eijk AA, et al. Guillain-Barré syndrome after SARS-CoV-2 infection in an international prospective cohort study. Brain 2021; 144:3392.
  155. Li X, Raventós B, Roel E, et al. Association between covid-19 vaccination, SARS-CoV-2 infection, and risk of immune mediated neurological events: population based cohort and self-controlled case series analysis. BMJ 2022; 376:e068373.
  156. Keddie S, Pakpoor J, Mousele C, et al. Epidemiological and cohort study finds no association between COVID-19 and Guillain-Barré syndrome. Brain 2021; 144:682.
  157. Li X, Wang Y, Wang H, Wang Y. SARS-CoV-2-associated Guillain-Barré syndrome is a para-infectious disease. QJM 2021; 114:625.
  158. Gutiérrez-Ortiz C, Méndez-Guerrero A, Rodrigo-Rey S, et al. Miller Fisher syndrome and polyneuritis cranialis in COVID-19. Neurology 2020; 95:e601.
  159. Juliao Caamaño DS, Alonso Beato R. Facial diplegia, a possible atypical variant of Guillain-Barré Syndrome as a rare neurological complication of SARS-CoV-2. J Clin Neurosci 2020; 77:230.
  160. Guidon AC, Amato AA. COVID-19 and neuromuscular disorders. Neurology 2020; 94:959.
  161. Carfì A, Bernabei R, Landi F, Gemelli Against COVID-19 Post-Acute Care Study Group. Persistent Symptoms in Patients After Acute COVID-19. JAMA 2020; 324:603.
  162. Suwanwongse K, Shabarek N. Rhabdomyolysis as a Presentation of 2019 Novel Coronavirus Disease. Cureus 2020; 12:e7561.
  163. Jin M, Tong Q. Rhabdomyolysis as Potential Late Complication Associated with COVID-19. Emerg Infect Dis 2020; 26:1618.
  164. Manzano GS, Woods JK, Amato AA. Covid-19-Associated Myopathy Caused by Type I Interferonopathy. N Engl J Med 2020; 383:2389.
  165. Goh Y, Beh DLL, Makmur A, et al. Pearls & Oy-sters: Facial nerve palsy in COVID-19 infection. Neurology 2020; 95:364.
  166. Dinkin M, Gao V, Kahan J, et al. COVID-19 presenting with ophthalmoparesis from cranial nerve palsy. Neurology 2020; 95:221.
  167. Decavel P, Petit C, Tatu L. Tapia syndrome at the time of the COVID-19 pandemic: Lower cranial neuropathy following prolonged intubation. Neurology 2020; 95:312.
  168. Todisco M, Alfonsi E, Arceri S, et al. Isolated bulbar palsy after SARS-CoV-2 infection. Lancet Neurol 2021; 20:169.
  169. Cacciavillani M, Salvalaggio A, Briani C. Pure sensory neuralgic amyotrophy in COVID-19 infection. Muscle Nerve 2021; 63:E7.
  170. Siepmann T, Kitzler HH, Lueck C, et al. Neuralgic amyotrophy following infection with SARS-CoV-2. Muscle Nerve 2020; 62:E68.
  171. Madia F, Merico B, Primiano G, et al. Acute myopathic quadriplegia in patients with COVID-19 in the intensive care unit. Neurology 2020; 95:492.
  172. Malik GR, Wolfe AR, Soriano R, et al. Injury-prone: peripheral nerve injuries associated with prone positioning for COVID-19-related acute respiratory distress syndrome. Br J Anaesth 2020; 125:e478.
  173. Fernandez CE, Franz CK, Ko JH, et al. Imaging Review of Peripheral Nerve Injuries in Patients with COVID-19. Radiology 2021; 298:E117.
  174. Moriguchi T, Harii N, Goto J, et al. A first case of meningitis/encephalitis associated with SARS-Coronavirus-2. Int J Infect Dis 2020; 94:55.
  175. Huang YH, Jiang D, Huang JT. SARS-CoV-2 Detected in Cerebrospinal Fluid by PCR in a Case of COVID-19 Encephalitis. Brain Behav Immun 2020; 87:149.
  176. Bernard-Valnet R, Pizzarotti B, Anichini A, et al. Two patients with acute meningoencephalitis concomitant with SARS-CoV-2 infection. Eur J Neurol 2020; 27:e43.
  177. Pilotto A, Odolini S, Masciocchi S, et al. Steroid-Responsive Encephalitis in Coronavirus Disease 2019. Ann Neurol 2020; 88:423.
  178. Dogan L, Kaya D, Sarikaya T, et al. Plasmapheresis treatment in COVID-19-related autoimmune meningoencephalitis: Case series. Brain Behav Immun 2020; 87:155.
  179. Panariello A, Bassetti R, Radice A, et al. Anti-NMDA receptor encephalitis in a psychiatric Covid-19 patient: A case report. Brain Behav Immun 2020; 87:179.
  180. Paniz-Mondolfi A, Bryce C, Grimes Z, et al. Central nervous system involvement by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). J Med Virol 2020; 92:699.
  181. Duong L, Xu P, Liu A. Meningoencephalitis without respiratory failure in a young female patient with COVID-19 infection in Downtown Los Angeles, early April 2020. Brain Behav Immun 2020; 87:33.
  182. McAbee GN, Brosgol Y, Pavlakis S, et al. Encephalitis Associated with COVID-19 Infection in an 11-Year-Old Child. Pediatr Neurol 2020; 109:94.
  183. Martin PJ, Felker M, Radhakrishnan R. MR Imaging Findings in a Neonate With COVID -19-Associated Encephalitis. Pediatr Neurol 2021; 119:48.
  184. Fadakar N, Ghaemmaghami S, Masoompour SM, et al. A First Case of Acute Cerebellitis Associated with Coronavirus Disease (COVID-19): a Case Report and Literature Review. Cerebellum 2020; 19:911.
  185. Povlow A, Auerbach AJ. Acute Cerebellar Ataxia in COVID-19 Infection: A Case Report. J Emerg Med 2021; 60:73.
  186. Sharma S, Ruparelia J, Bhaskar S, et al. Acute Fulminant Cerebellitis in Children with COVID-19 Infection: A Rare But Treatable Complication. Pediatr Neurol 2021; 119:45.
  187. Oosthuizen K, Steyn EC, Tucker L, et al. SARS-CoV-2 Encephalitis Presenting as a Clinical Cerebellar Syndrome: A Case Report. Neurology 2021; 97:27.
  188. Sotgiu S, Uzzau S, Pippia A, et al. Expanding the Spectrum of Acute Cerebellitis due to SARS-Cov-2. Pediatr Neurol 2021; 121:1.
  189. Zanin L, Saraceno G, Panciani PP, et al. SARS-CoV-2 can induce brain and spine demyelinating lesions. Acta Neurochir (Wien) 2020; 162:1491.
  190. Reichard RR, Kashani KB, Boire NA, et al. Neuropathology of COVID-19: a spectrum of vascular and acute disseminated encephalomyelitis (ADEM)-like pathology. Acta Neuropathol 2020; 140:1.
  191. Delamarre L, Gollion C, Grouteau G, et al. COVID-19-associated acute necrotising encephalopathy successfully treated with steroids and polyvalent immunoglobulin with unusual IgG targeting the cerebral fibre network. J Neurol Neurosurg Psychiatry 2020; 91:1004.
  192. Krett JD, Jewett GAE, Elton-Lacasse C, et al. Hemorrhagic encephalopathy associated with COVID-19. J Neuroimmunol 2020; 346:577326.
  193. Poyiadji N, Shahin G, Noujaim D, et al. COVID-19-associated Acute Hemorrhagic Necrotizing Encephalopathy: Imaging Features. Radiology 2020; 296:E119.
  194. Dixon L, Varley J, Gontsarova A, et al. COVID-19-related acute necrotizing encephalopathy with brain stem involvement in a patient with aplastic anemia. Neurol Neuroimmunol Neuroinflamm 2020; 7.
  195. Danoun OA, Zillgitt A, Hill C, et al. Outcomes of seizures, status epilepticus, and EEG findings in critically ill patient with COVID-19. Epilepsy Behav 2021; 118:107923.
  196. Emami A, Fadakar N, Akbari A, et al. Seizure in patients with COVID-19. Neurol Sci 2020; 41:3057.
  197. Asadi-Pooya AA, Kouhanjani MF, Nemati H, et al. A follow-up study of patients with COVID-19 presenting with seizures. Epilepsy Behav 2021; 122:108207.
  198. Anand P, Al-Faraj A, Sader E, et al. Seizure as the presenting symptom of COVID-19: A retrospective case series. Epilepsy Behav 2020; 112:107335.
  199. Dono F, Nucera B, Lanzone J, et al. Status epilepticus and COVID-19: A systematic review. Epilepsy Behav 2021; 118:107887.
  200. Rábano-Suárez P, Bermejo-Guerrero L, Méndez-Guerrero A, et al. Generalized myoclonus in COVID-19. Neurology 2020; 95:e767.
  201. Franceschi AM, Ahmed O, Giliberto L, Castillo M. Hemorrhagic Posterior Reversible Encephalopathy Syndrome as a Manifestation of COVID-19 Infection. AJNR Am J Neuroradiol 2020; 41:1173.
  202. Kaya Y, Kara S, Akinci C, Kocaman AS. Transient cortical blindness in COVID-19 pneumonia; a PRES-like syndrome: Case report. J Neurol Sci 2020; 413:116858.
  203. Kishfy L, Casasola M, Banankhah P, et al. Posterior reversible encephalopathy syndrome (PRES) as a neurological association in severe Covid-19. J Neurol Sci 2020; 414:116943.
  204. Princiotta Cariddi L, Tabaee Damavandi P, Carimati F, et al. Reversible Encephalopathy Syndrome (PRES) in a COVID-19 patient. J Neurol 2020; 267:3157.
  205. Parauda SC, Gao V, Gewirtz AN, et al. Posterior reversible encephalopathy syndrome in patients with COVID-19. J Neurol Sci 2020; 416:117019.
  206. Sadeghizadeh A, Pourmoghaddas Z, Zandifar A, et al. Reversible Cerebral Vasoconstriction Syndrome and Multisystem Inflammatory Syndrome in Children With COVID-19. Pediatr Neurol 2022; 129:1.
  207. Ray S, Kamath VV, Raju P A, et al. Fulminant Reversible Cerebral Vasoconstriction Syndrome in Breakthrough COVID 19 Infection. J Stroke Cerebrovasc Dis 2022; 31:106238.
  208. Arandela K, Samudrala S, Abdalkader M, et al. Reversible Cerebral Vasoconstriction Syndrome in Patients with Coronavirus Disease: A Multicenter Case Series. J Stroke Cerebrovasc Dis 2021; 30:106118.
  209. Goërtz YMJ, Van Herck M, Delbressine JM, et al. Persistent symptoms 3 months after a SARS-CoV-2 infection: the post-COVID-19 syndrome? ERJ Open Res 2020; 6.
  210. Huang C, Huang L, Wang Y, et al. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet 2021; 397:220.
  211. Xiong Q, Xu M, Li J, et al. Clinical sequelae of COVID-19 survivors in Wuhan, China: a single-centre longitudinal study. Clin Microbiol Infect 2021; 27:89.
  212. Neurology and COVID-19: Scientific brief, 29 September 2021 https://www.who.int/publications/i/item/WHO-2019-nCoV-Sci-Brief-Neurology-2021.1.
  213. Kim Y, Bitna-Ha, Kim SW, et al. Post-acute COVID-19 syndrome in patients after 12 months from COVID-19 infection in Korea. BMC Infect Dis 2022; 22:93.
  214. Petersen MS, Kristiansen MF, Hanusson KD, et al. Long COVID in the Faroe Islands: A Longitudinal Study Among Nonhospitalized Patients. Clin Infect Dis 2021; 73:e4058.
  215. Centers for Disease Control and Prevention. Background: Evaluating and caring for patients with post-COVID conditions: Interim guidance. https://www.cdc.gov/coronavirus/2019-ncov/hcp/clinical-care/post-covid-background.html (Accessed on June 17, 2021).
  216. Havervall S, Rosell A, Phillipson M, et al. Symptoms and Functional Impairment Assessed 8 Months After Mild COVID-19 Among Health Care Workers. JAMA 2021; 325:2015.
  217. Office of National Statistics. Prevalence of ongoing symptoms following coronavirus (COVID-19) infection in the UK: 1 April 2021. https://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/conditionsanddiseases/bulletins/prevalenceofongoingsymptomsfollowingcoronaviruscovid19infectionintheuk/1april2021 (Accessed on June 17, 2021).
  218. Chevinsky JR, Tao G, Lavery AM, et al. Late Conditions Diagnosed 1-4 Months Following an Initial Coronavirus Disease 2019 (COVID-19) Encounter: A Matched-Cohort Study Using Inpatient and Outpatient Administrative Data-United States, 1 March-30 June 2020. Clin Infect Dis 2021; 73:S5.
  219. Hernandez-Romieu AC, Leung S, Mbanya A, et al. Health Care Utilization and Clinical Characteristics of Nonhospitalized Adults in an Integrated Health Care System 28-180 Days After COVID-19 Diagnosis - Georgia, May 2020-March 2021. MMWR Morb Mortal Wkly Rep 2021; 70:644.
  220. Pavli A, Theodoridou M, Maltezou HC. Post-COVID Syndrome: Incidence, Clinical Spectrum, and Challenges for Primary Healthcare Professionals. Arch Med Res 2021; 52:575.
  221. Cabrera Martimbianco AL, Pacheco RL, Bagattini ÂM, Riera R. Frequency, signs and symptoms, and criteria adopted for long COVID-19: A systematic review. Int J Clin Pract 2021; 75:e14357.
  222. Lund LC, Hallas J, Nielsen H, et al. Post-acute effects of SARS-CoV-2 infection in individuals not requiring hospital admission: a Danish population-based cohort study. Lancet Infect Dis 2021; 21:1373.
  223. Groff D, Sun A, Ssentongo AE, et al. Short-term and Long-term Rates of Postacute Sequelae of SARS-CoV-2 Infection: A Systematic Review. JAMA Netw Open 2021; 4:e2128568.
  224. Taquet M, Dercon Q, Luciano S, et al. Incidence, co-occurrence, and evolution of long-COVID features: A 6-month retrospective cohort study of 273,618 survivors of COVID-19. PLoS Med 2021; 18:e1003773.
  225. Sampaio Rocha-Filho PA, Albuquerque PM, Carvalho LCLS, et al. Headache, anosmia, ageusia and other neurological symptoms in COVID-19: a cross-sectional study. J Headache Pain 2022; 23:2.
  226. Kosugi EM, Lavinsky J, Romano FR, et al. Incomplete and late recovery of sudden olfactory dysfunction in COVID-19. Braz J Otorhinolaryngol 2020; 86:490.
  227. Karamali K, Elliott M, Hopkins C. COVID-19 related olfactory dysfunction. Curr Opin Otolaryngol Head Neck Surg 2022; 30:19.
  228. Balcom EF, Nath A, Power C. Acute and chronic neurological disorders in COVID-19: potential mechanisms of disease. Brain 2021; 144:3576.
  229. Del Brutto OH, Wu S, Mera RM, et al. Cognitive decline among individuals with history of mild symptomatic SARS-CoV-2 infection: A longitudinal prospective study nested to a population cohort. Eur J Neurol 2021; 28:3245.
  230. Liu YH, Chen Y, Wang QH, et al. One-Year Trajectory of Cognitive Changes in Older Survivors of COVID-19 in Wuhan, China: A Longitudinal Cohort Study. JAMA Neurol 2022; 79:509.
  231. Kanjwal K, Jamal S, Kichloo A, Grubb BP. New-onset Postural Orthostatic Tachycardia Syndrome Following Coronavirus Disease 2019 Infection. J Innov Card Rhythm Manag 2020; 11:4302.
  232. Miglis MG, Prieto T, Shaik R, et al. A case report of postural tachycardia syndrome after COVID-19. Clin Auton Res 2020; 30:449.
  233. Blitshteyn S, Whitelaw S. Postural orthostatic tachycardia syndrome (POTS) and other autonomic disorders after COVID-19 infection: a case series of 20 patients. Immunol Res 2021; 69:205.
  234. Mazza MG, De Lorenzo R, Conte C, et al. Anxiety and depression in COVID-19 survivors: Role of inflammatory and clinical predictors. Brain Behav Immun 2020; 89:594.
  235. Islam MF, Cotler J, Jason LA. Post-viral fatigue and COVID-19: lessons from past epidemics. Fatigue 2020.
  236. Townsend L, Dyer AH, Jones K, et al. Persistent fatigue following SARS-CoV-2 infection is common and independent of severity of initial infection. PLoS One 2020; 15:e0240784.
  237. Afari N, Buchwald D. Chronic fatigue syndrome: a review. Am J Psychiatry 2003; 160:221.
  238. Norrie J, Heitger M, Leathem J, et al. Mild traumatic brain injury and fatigue: a prospective longitudinal study. Brain Inj 2010; 24:1528.
  239. Marshall M. The lasting misery of coronavirus long-haulers. Nature 2020; 585:339.
  240. Douaud G, Lee S, Alfaro-Almagro F, et al. SARS-CoV-2 is associated with changes in brain structure in UK Biobank. Nature 2022; 604:697.
  241. Hanson BA, Visvabharathy L, Ali ST, et al. Plasma Biomarkers of Neuropathogenesis in Hospitalized Patients With COVID-19 and Those With Postacute Sequelae of SARS-CoV-2 Infection. Neurol Neuroimmunol Neuroinflamm 2022; 9.
  242. Živković SA, Gruener G, Narayanaswami P, AANEM Quality and Patient Safety Committee. Doctor-Should I get the COVID-19 vaccine? Infection and immunization in individuals with neuromuscular disorders. Muscle Nerve 2021; 63:294.
  243. National Multiple Sclerosis Society. COVID-19 Vaccine Guidance for People Living with MS. https://www.nationalmssociety.org/coronavirus-covid-19-information/multiple-sclerosis-and-coronavirus/covid-19-vaccine-guidance (Accessed on February 12, 2021).
  244. Kim YE, Huh K, Park YJ, et al. Association Between Vaccination and Acute Myocardial Infarction and Ischemic Stroke After COVID-19 Infection. JAMA 2022; 328:887.
  245. Rutkove SB, Betensky RA. No, it's not 1976 all over again. Ann Neurol 2021; 90:189.
  246. Ashrani AA, Crusan DJ, Petterson T, et al. Age- and Sex-Specific Incidence of Cerebral Venous Sinus Thrombosis Associated With Ad26.COV2.S COVID-19 Vaccination. JAMA Intern Med 2022; 182:80.
  247. Patone M, Handunnetthi L, Saatci D, et al. Neurological complications after first dose of COVID-19 vaccines and SARS-CoV-2 infection. Nat Med 2021; 27:2144.
  248. Wise J. Covid-19: European countries suspend use of Oxford-AstraZeneca vaccine after reports of blood clots. BMJ 2021; 372:n699.
  249. European Medicines Agency. Signal assessment report on embolic and thrombotic events (SMQ) with COVID-19 Vaccine (ChAdOx1-S [recombinant]) – COVID-19 Vaccine AstraZeneca (Other viral vaccines). https://www.ema.europa.eu/en/documents/prac-recommendation/signal-assessment-report-embolic-thrombotic-events-smq-covid-19-vaccine-chadox1-s-recombinant-covid_en.pdf (Accessed on April 02, 2021).
  250. Schultz NH, Sørvoll IH, Michelsen AE, et al. Thrombosis and Thrombocytopenia after ChAdOx1 nCoV-19 Vaccination. N Engl J Med 2021; 384:2124.
  251. Greinacher A, Thiele T, Warkentin TE, et al. Thrombotic Thrombocytopenia after ChAdOx1 nCov-19 Vaccination. N Engl J Med 2021; 384:2092.
  252. Joint CDC and FDA Statement on Johnson & Johnson COVID-19 Vaccine https://www.cdc.gov/media/releases/2021/s0413-JJ-vaccine.html.
  253. European Medicines Agency safety committee report https://www.ema.europa.eu/en/news/astrazenecas-covid-19-vaccine-ema-finds-possible-link-very-rare-cases-unusual-blood-clots-low-blood.
  254. Muir KL, Kallam A, Koepsell SA, Gundabolu K. Thrombotic Thrombocytopenia after Ad26.COV2.S Vaccination. N Engl J Med 2021; 384:1964.
  255. Bayas A, Menacher M, Christ M, et al. Bilateral superior ophthalmic vein thrombosis, ischaemic stroke, and immune thrombocytopenia after ChAdOx1 nCoV-19 vaccination. Lancet 2021; 397:e11.
  256. See I, Su JR, Lale A, et al. US Case Reports of Cerebral Venous Sinus Thrombosis With Thrombocytopenia After Ad26.COV2.S Vaccination, March 2 to April 21, 2021. JAMA 2021; 325:2448.
  257. Blauenfeldt RA, Kristensen SR, Ernstsen SL, et al. Thrombocytopenia with acute ischemic stroke and bleeding in a patient newly vaccinated with an adenoviral vector-based COVID-19 vaccine. J Thromb Haemost 2021; 19:1771.
  258. Pavord S, Scully M, Hunt BJ, et al. Clinical Features of Vaccine-Induced Immune Thrombocytopenia and Thrombosis. N Engl J Med 2021; 385:1680.
  259. Sánchez van Kammen M, Aguiar de Sousa D, Poli S, et al. Characteristics and Outcomes of Patients With Cerebral Venous Sinus Thrombosis in SARS-CoV-2 Vaccine-Induced Immune Thrombotic Thrombocytopenia. JAMA Neurol 2021; 78:1314.
  260. Perry RJ, Tamborska A, Singh B, et al. Cerebral venous thrombosis after vaccination against COVID-19 in the UK: a multicentre cohort study. Lancet 2021; 398:1147.
  261. European Medicines Agency COVID-19 vaccine safety update 14 July 2021 ema.europa.eu/en/documents/covid-19-vaccine-safety-update/covid-19-vaccine-safety-update-covid-19-vaccine-janssen-14-july-2021_en.pdf.
  262. Allen CM, Ramsamy S, Tarr AW, et al. Guillain-Barré Syndrome Variant Occurring after SARS-CoV-2 Vaccination. Ann Neurol 2021; 90:315.
  263. Maramattom BV, Krishnan P, Paul R, et al. Guillain-Barré Syndrome following ChAdOx1-S/nCoV-19 Vaccine. Ann Neurol 2021; 90:312.
  264. Woo EJ, Mba-Jonas A, Dimova RB, et al. Association of Receipt of the Ad26.COV2.S COVID-19 Vaccine With Presumptive Guillain-Barré Syndrome, February-July 2021. JAMA 2021; 326:1606.
  265. Hanson KE, Goddard K, Lewis N, et al. Incidence of Guillain-Barré Syndrome After COVID-19 Vaccination in the Vaccine Safety Datalink. JAMA Netw Open 2022; 5:e228879.
  266. Needham EJ, Chou SH, Coles AJ, Menon DK. Neurological Implications of COVID-19 Infections. Neurocrit Care 2020; 32:667.
  267. Brownlee W, Bourdette D, Broadley S, et al. Treating multiple sclerosis and neuromyelitis optica spectrum disorder during the COVID-19 pandemic. Neurology 2020; 94:949.
  268. Sormani MP, Italian Study Group on COVID-19 infection in multiple sclerosis. An Italian programme for COVID-19 infection in multiple sclerosis. Lancet Neurol 2020; 19:481.
  269. International MG/COVID-19 Working Group, Jacob S, Muppidi S, et al. Guidance for the management of myasthenia gravis (MG) and Lambert-Eaton myasthenic syndrome (LEMS) during the COVID-19 pandemic. J Neurol Sci 2020; 412:116803.
  270. Solé G, Salort-Campana E, Pereon Y, et al. Guidance for the care of neuromuscular patients during the COVID-19 pandemic outbreak from the French Rare Health Care for Neuromuscular Diseases Network. Rev Neurol (Paris) 2020; 176:507.
  271. Disanto G, Sacco R, Bernasconi E, et al. Association of Disease-Modifying Treatment and Anti-CD20 Infusion Timing With Humoral Response to 2 SARS-CoV-2 Vaccines in Patients With Multiple Sclerosis. JAMA Neurol 2021; 78:1529.
  272. Louapre C, Ibrahim M, Maillart E, et al. Anti-CD20 therapies decrease humoral immune response to SARS-CoV-2 in patients with multiple sclerosis or neuromyelitis optica spectrum disorders. J Neurol Neurosurg Psychiatry 2022; 93:24.
  273. Apostolidis SA, Kakara M, Painter MM, et al. Cellular and humoral immune responses following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-CD20 therapy. Nat Med 2021; 27:1990.
  274. Brill L, Rechtman A, Zveik O, et al. Humoral and T-Cell Response to SARS-CoV-2 Vaccination in Patients With Multiple Sclerosis Treated With Ocrelizumab. JAMA Neurol 2021; 78:1510.
  275. Tallantyre EC, Vickaryous N, Anderson V, et al. COVID-19 Vaccine Response in People with Multiple Sclerosis. Ann Neurol 2022; 91:89.
  276. Muppidi S, Guptill JT, Jacob S, et al. COVID-19-associated risks and effects in myasthenia gravis (CARE-MG). Lancet Neurol 2020; 19:970.
  277. Anand P, Slama MCC, Kaku M, et al. COVID-19 in patients with myasthenia gravis. Muscle Nerve 2020; 62:254.
  278. Rodrigues CL, de Freitas HC, Lima PRO, et al. Myasthenia gravis exacerbation and myasthenic crisis associated with COVID-19: case series and literature review. Neurol Sci 2022; 43:2271.
  279. Gungor Tuncer O, Deymeer F. Clinical course and outcome of an outpatient clinic population with myasthenia gravis and COVID-19. Muscle Nerve 2022; 65:447.
  280. Businaro P, Vaghi G, Marchioni E, et al. COVID-19 in patients with myasthenia gravis: Epidemiology and disease course. Muscle Nerve 2021; 64:206.
  281. Neykova KK, Milanova M, Ignatov PN. Myasthenia gravis and covid-19 in pregnancy: a review of the literature and case series report. J Matern Fetal Neonatal Med 2022; 35:8308.
  282. Jakubíková M, Týblová M, Tesař A, et al. Predictive factors for a severe course of COVID-19 infection in myasthenia gravis patients with an overall impact on myasthenic outcome status and survival. Eur J Neurol 2021; 28:3418.
  283. Yang X, Yu Y, Xu J, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med 2020; 8:475.
  284. Guan WJ, Ni ZY, Hu Y, et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med 2020; 382:1708.
  285. Zhang JJ, Dong X, Cao YY, et al. Clinical characteristics of 140 patients infected with SARS-CoV-2 in Wuhan, China. Allergy 2020; 75:1730.
  286. Qin C, Zhou L, Hu Z, et al. Clinical Characteristics and Outcomes of COVID-19 Patients With a History of Stroke in Wuhan, China. Stroke 2020; 51:2219.
  287. Kummer BR, Klang E, Stein LK, et al. History of Stroke Is Independently Associated With In-Hospital Death in Patients With COVID-19. Stroke 2020; 51:3112.
  288. Yang J, Zheng Y, Gou X, et al. Prevalence of comorbidities and its effects in patients infected with SARS-CoV-2: a systematic review and meta-analysis. Int J Infect Dis 2020; 94:91.
  289. Azar KMJ, Shen Z, Romanelli RJ, et al. Disparities In Outcomes Among COVID-19 Patients In A Large Health Care System In California. Health Aff (Millwood) 2020; 39:1253.
  290. Tal Y, Adini A, Eran A, Adini I. Racial disparity in Covid-19 mortality rates - A plausible explanation. Clin Immunol 2020; 217:108481.
  291. Siahaan YMT, Ketaren RJ, Hartoyo V, Hariyanto TI. Epilepsy and the risk of severe coronavirus disease 2019 outcomes: A systematic review, meta-analysis, and meta-regression. Epilepsy Behav 2021; 125:108437.
  292. Benussi A, Pilotto A, Premi E, et al. Clinical characteristics and outcomes of inpatients with neurologic disease and COVID-19 in Brescia, Lombardy, Italy. Neurology 2020; 95:e910.
  293. Solé G, Mathis S, Friedman D, et al. Impact of Coronavirus Disease 2019 in a French Cohort of Myasthenia Gravis. Neurology 2021; 96:e2109.
  294. Louapre C, Collongues N, Stankoff B, et al. Clinical Characteristics and Outcomes in Patients With Coronavirus Disease 2019 and Multiple Sclerosis. JAMA Neurol 2020; 77:1079.
  295. Simpson-Yap S, De Brouwer E, Kalincik T, et al. Associations of Disease-Modifying Therapies With COVID-19 Severity in Multiple Sclerosis. Neurology 2021; 97:e1870.
  296. Clark JR, Batra A, Shlobin NA, et al. Acute-care hospital reencounters in COVID-19 patients. Geroscience 2021; 43:2041.
  297. Azarpazhooh MR, Amiri A, Morovatdar N, et al. Correlations between COVID-19 and burden of dementia: An ecological study and review of literature. J Neurol Sci 2020; 416:117013.
  298. Cabezudo-García P, Ciano-Petersen NL, Mena-Vázquez N, et al. Incidence and case fatality rate of COVID-19 in patients with active epilepsy. Neurology 2020; 95:e1417.
  299. Sanchez-Larsen A, Conde-Blanco E, Viloria-Alebesque A, et al. COVID-19 prevalence and mortality in people with epilepsy: A nation-wide multicenter study. Epilepsy Behav 2021; 125:108379.
  300. Puteikis K, Jasionis A, Mameniškienė R. Recalling the COVID-19 lockdown: Insights from patients with epilepsy. Epilepsy Behav 2021; 115:107573.
  301. French JA, Brodie MJ, Caraballo R, et al. Keeping people with epilepsy safe during the COVID-19 pandemic. Neurology 2020; 94:1032.
Topic 128153 Version 29.0

References