Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Pediatr Blood Cancer ; 70(3): e30193, 2023 03.
Article in English | MEDLINE | ID: covidwho-20233114

ABSTRACT

Vulnerable patient populations have seen decreased rates of vaccination against SARS-CoV2-19 (COVID-19) due to hesitancies and distrust, magnified by a paucity of data for certain populations. The rate of COVID-19 vaccination in children with sickle cell disease (SCD) remains low despite the risk for severe complications, resulting in continued infections and hospitalizations from COVID-19. We sought to describe vaccine reactions, including vaso-occlusive crises, emergency department visits, and hospitalizations, in children with SCD. Our findings will start to provide the necessary vaccine side effect data to inform patients, caregivers, and clinicians considering the COVID-19 primary vaccination series.


Subject(s)
Anemia, Sickle Cell , COVID-19 Vaccines , COVID-19 , Drug-Related Side Effects and Adverse Reactions , Child , Humans , Anemia, Sickle Cell/therapy , Anemia, Sickle Cell/drug therapy , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/complications , COVID-19 Vaccines/adverse effects , RNA, Viral/therapeutic use , SARS-CoV-2
2.
Mult Scler Relat Disord ; 74: 104715, 2023 Jun.
Article in English | MEDLINE | ID: covidwho-2305245

ABSTRACT

INTRODUCTION: An association between intercurrent viral respiratory infections and exacerbations of Multiple Sclerosis (MS) disease activity has been proposed by several studies. Considering the rapid spread of SARS-CoV2 worldwide and the systematic effort to immediately detect all incident cases with specific diagnostic tests, the pandemic can represent an interesting experimental model to assess the relationship between viral respiratory infections and MS disease activity. AIMS AND METHODS: In this study, we have performed a propensity score matched case-control study with a prospective clinical/MRI follow-up, on a cohort of relapsing-remitting MS (RRMS) patients who tested positive for SARS-CoV2 in the period 2020-2022, with the aim to evaluate if the SARS-CoV2 infection influences the short-term risk of disease activity. Controls (RRMS patients not exposed to SARS-CoV-2, using 2019 as the reference period) were matched 1:1 with cases for age, EDSS, sex and disease-modifying treatment (DMT) (moderate efficacy vs high efficacy). Differences in relapses, MRI disease activity and confirmed disabilty worsening (CDW) between cases in the 6 months following the SARS-CoV-2 infection, and controls in a similar 6 months reference period in 2019 were compared. RESULTS: We identified 150 cases of SARS-CoV2 infection in the period March 2020 - March 2022, out of a total population of approximately 1500 MS patients, matched with 150 MS patients not exposed to SARS-CoV2 (controls). Mean age was 40.9 ± 12.0 years in cases and 42.0 ± 10.9 years in controls, mean EDSS was 2.54±1.36 in cases and 2.60±1.32 in controls. All patients were treated with a DMT, and a considerable proportion with a high efficacy DMT (65.3% in cases and 66% in controls), reflecting a typical real world RRMS population. 52.8% of patients in this cohort had been vaccinated with a mRNA Covid-19 vaccine. We did not observe a significant difference in relapses (4.0% cases, 5.3% controls; p = 0.774), MRI disease activity (9.3% cases, 8.0% controls; p = 0.838), CDW (5.3% cases, 6.7% controls; p = 0.782) in the 6 months after SARS-CoV-2 infection between cases and controls. CONCLUSION: Using a propensity score matching design and including both clinical and MRI data, this study does not suggest an increased risk of MS disease activity following SARS-CoV-2 infection. All MS patients in this cohort were treated with a DMT, and a considerable number with a high efficacy DMT. These results therefore may not be applicable to untreated patients, for which the risk of increased MS disease activity after SARS-CoV-2 infection may not be excluded. A possible hypothesis explaining these results could be that SARS-CoV2 is less prone, compared to other viruses, to induce exacerbations of MS disease activity; another possible interpretation of these data might be that DMT is able to effectively suppress the increase of disease activity triggered by SARS-CoV2 infection.


Subject(s)
COVID-19 , Multiple Sclerosis , Humans , Adult , Middle Aged , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/epidemiology , Multiple Sclerosis/drug therapy , SARS-CoV-2 , COVID-19 Vaccines , Case-Control Studies , COVID-19/epidemiology , Prospective Studies , Pandemics , Propensity Score , RNA, Viral/therapeutic use , Recurrence
3.
J Clin Lipidol ; 17(2): 219-224, 2023.
Article in English | MEDLINE | ID: covidwho-2227889

ABSTRACT

BACKGROUND: The effect of SARS-CoV-2 infection in blood lipids of homozygous familial hypercholesterolemia (HoFH) has not been explored. CASE SUMMARY: We report a case of a 43-year-old male patient with -/-LDLR HoFH with previous history of premature coronary artery disease, coronary artery bypass graft (CABG) and surgical repair of aortic valve stenosis. He presented with an abrupt decrease of his blood lipid levels during acute infection with SARS-CoV2 and subsequently a rebound increase above pre-infection levels, refractory to treatment including LDL-apheresis, statin, ezetimibe and lomitapide up-titration to maximum tolerated doses. Markers of liver stiffness were closely monitored, increased at 9 months and decreased at 18 months after the infection. Potential interactions of hypolipidemic treatment with the viral replication process during the acute phase, as well as therapeutic dilemmas occurring in the post infection period are discussed.


Subject(s)
Anticholesteremic Agents , COVID-19 , Homozygous Familial Hypercholesterolemia , Hypercholesterolemia , Hyperlipoproteinemia Type II , Adult , Humans , Male , Anticholesteremic Agents/therapeutic use , Homozygote , Hypercholesterolemia/drug therapy , Hyperlipoproteinemia Type II/complications , Hyperlipoproteinemia Type II/genetics , Lipids , RNA, Viral/therapeutic use , SARS-CoV-2
4.
Reumatol Clin (Engl Ed) ; 19(2): 67-73, 2023 Feb.
Article in English | MEDLINE | ID: covidwho-2221312

ABSTRACT

BACKGROUND: Rheumatological manifestations following COVID-19 are various, including Reactive Arthritis (ReA), which is a form of asymmetric oligoarthritis mainly involving the lower limbs, with or without extra-articular features. The current case series describes the clinical profile and treatment outcome of 23 patients with post-COVID-19 ReA. METHODS: A retrospective, observational study of patients with post-COVID-19 arthritis over one year was conducted at a tertiary care centre in India. Patients (n=23) with either a positive polymerase chain reaction test for SARS-CoV2 or an anti-COVID-19 antibody test were included. Available demographic details, musculoskeletal symptoms, inflammatory markers, and treatment given were documented. RESULTS: Sixteen out of 23 patients were female. The mean age of the patients was 42.8 years. Nineteen patients had had symptomatic COVID-19 infection in the past. The duration between onset of COVID-19 symptoms and arthritis ranged from 5 to 52 days with a mean of 25.9 days. The knee was the most involved joint (16 out of 23 cases). Seven patients had inflammatory lower back pain and nine had enthesitis. Most patients were treated with non-steroidal anti-inflammatory drugs (NSAIDs) and steroids - either depot injection or a short oral course. Three patients required treatment with hydroxychloroquine and methotrexate which were eventually stopped. No relapse was reported in any of the patients. CONCLUSION: On combining our data with 21 other case reports of ReA, a lower limb predominant, oligoarticular, asymmetric pattern of arthritis was seen with a female preponderance. The mean number of joints involved was 2.8. Axial symptoms and enthesitis were often coexistent. Treatment with NSAIDs and intra-articular steroids was effective. However, whether COVID-19 was the definitive aetiology of the arthritis is yet to be proven.


Subject(s)
Arthritis, Reactive , COVID-19 , Humans , Female , Adult , Male , Arthritis, Reactive/diagnosis , Arthritis, Reactive/drug therapy , Arthritis, Reactive/etiology , Tertiary Care Centers , Retrospective Studies , RNA, Viral/therapeutic use , COVID-19/complications , SARS-CoV-2 , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Observational Studies as Topic
5.
Arch Razi Inst ; 77(6): 2181-2186, 2022 12.
Article in English | MEDLINE | ID: covidwho-2226511

ABSTRACT

It is estimated that more than 400 million people worldwide are suffering from diabetes. There are two types of diabetes. Type 1 diabetes is the result of insufficient insulin secretion into the bloodstream, most often due to an autoimmune attack on the pancreas glands. Type 2 diabetes is caused by the inability of the surface ligands to adsorb the insulin from the bloodstream. The conventional medicines for diabetes mellitus include sulfonylureas, biguanide, thiazolidinediones, alpha-glucosidase inhibitors, and meglitinide. By February 2022, Severe acute respiratory syndrome coronavirus 2 (SARSCoV2) had infected more than 391 million people worldwide, claiming 5.7 million lives and imposing heavy costs on the healthcare system. The present study aimed to assess the potential use of this non-structural SARS-CoV-2 protein in the treatment of type 2 diabetes mellitus. The nsp10 was structurally aligned with GoDrugBank therapeutic agents, and lixisenatide was found to have the most similar chemical structure. This drug is a glucagon-like peptide-1 (GLP1) receptor agonist used for the treatment of type 2 diabetes mellitus. The best molecular docking energy score for these two proteins was -301.47, and the ligand root mean square deviation was calculated to be 107.93 Å. The molecular dynamics for the stability of the nsp10 and GLP1R binding in triplicate for 150 ns demonstrated that the nsp10-GLP1R remained bound for more than 80 ns. This study indicated that the nsp10 protein can be further studied to be used as an antidiabetic medication.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 2 , Humans , Blood Glucose , COVID-19/veterinary , Diabetes Mellitus, Type 2/drug therapy , Molecular Docking Simulation , RNA, Viral/therapeutic use , SARS-CoV-2
6.
J Neurooncol ; 161(1): 67-76, 2023 Jan.
Article in English | MEDLINE | ID: covidwho-2174681

ABSTRACT

PURPOSE: Glioblastoma (GBM) is the most common and malignant primary brain tumor in adults with a median overall survival of only 14.6 months despite aggressive treatment. While immunotherapy has been successful in other cancers, its benefit has been proven elusive in GBM, mainly due to a markedly immunosuppressive tumor microenvironment. SARS-CoV-2 has been associated with the development of a pronounced central nervous system (CNS) inflammatory response when infecting different cells including astrocytes, endothelial cells, and microglia. While SARS-CoV2 entry factors have been described in different tissues, their presence and implication on GBM aggressiveness or microenvironment has not been studied on appropriate preclinical models. METHODS: We evaluated the presence of crucial SARS-CoV-2 entry factors: ACE2, TMPRSS2, and NRP1 in matched surgically-derived GBM tissue, cells lines, and organoids; as well as in human brain derived specimens using immunohistochemistry, confocal pixel line intensity quantification, and transcriptome analysis. RESULTS: We show that patient derived-GBM tissue and cell cultures express SARS-CoV2 entry factors, being NRP1 the most crucial facilitator of SARS-CoV-2 infection in GBM. Moreover, we demonstrate that, receptor expression remains present in our GBM organoids, making them an adequate model to study the effect of this virus in GBM for the potential development of viral therapies in the future. CONCLUSION: Our findings suggest that the SARS-CoV-2 virus entry factors are expressed in primary tissues and organoid models and could be potentially utilized to study the susceptibility of GBM to this virus to target or modulate the tumor microenviroment.


Subject(s)
COVID-19 , Glioblastoma , Adult , Humans , Glioblastoma/pathology , SARS-CoV-2 , RNA, Viral/metabolism , RNA, Viral/therapeutic use , Endothelial Cells/metabolism , Organoids/metabolism , Organoids/pathology , Tumor Microenvironment
7.
Epilepsy Behav ; 139: 109070, 2023 02.
Article in English | MEDLINE | ID: covidwho-2165962

ABSTRACT

OBJECTIVE: This study aimed to identify seizure outcomes in people with epilepsy (PWE) following severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) messenger RNA vaccination. METHODS: We examined PWE (n = 332, age ≥ 14 years) treated in four tertiary hospitals between 2021 and 2022 to assess the incidence of seizure worsening following vaccination using closed questions. We identified the clinical factors associated with worsening and 6-month vaccination outcomes. We also conducted a nationwide survey on self-reported seizure worsening using open questions, to which 261 general practitioners from 99 institutes contributed. RESULTS: Of the 282 PWE vaccinated in the four hospitals, 16 (5.7%) exhibited seizure worsening; most of them emerged within 48 h of vaccination and were not sustained. Thus, all PWE were at baseline condition 6 months after their vaccination. PWE with seizure worsening were more significantly associated with focal impaired awareness seizures (p < 0.001), high seizure frequency (p = 0.025), and drug-resistant epilepsy (p = 0.007) at baseline compared to PWE without worsening. Multivariate logistic regression analysis revealed that focal impaired awareness seizures were independently associated with worsening (odds ratio, 7.0; 95% confidence interval, 1.50-32.77). A nationwide survey of 5156 PWE data (real-world data) confirmed an extremely low incidence rate of self-reported seizure worsening (0.43%). SIGNIFICANCE: Some PWE, particularly refractory focal epilepsy, exhibit seizure worsening. However, the worsening events were infrequent, non-sustainable, and probably under-reported by PWE, suggesting that there is little evidence that worsening seizures discourage current and future vaccinations.


Subject(s)
COVID-19 , Epilepsies, Partial , Epilepsy , Humans , Adolescent , RNA, Viral/therapeutic use , SARS-CoV-2 , COVID-19/prevention & control , Seizures/etiology , Epilepsy/epidemiology
8.
PLoS One ; 17(11): e0271574, 2022.
Article in English | MEDLINE | ID: covidwho-2119192

ABSTRACT

BACKGROUND: While vaccination is the most important way to combat the SARS-CoV-2 pandemic, there may still be a need for early outpatient treatment that is safe, inexpensive, and currently widely available in parts of the world that do not have access to the vaccine. There are in-silico, in-vitro, and in-tissue data suggesting that metformin inhibits the viral life cycle, as well as observational data suggesting that metformin use before infection with SARS-CoV2 is associated with less severe COVID-19. Previous observational analyses from single-center cohorts have been limited by size. METHODS: Conducted a retrospective cohort analysis in adults with type 2 diabetes (T2DM) for associations between metformin use and COVID-19 outcomes with an active comparator design of prevalent users of therapeutically equivalent diabetes monotherapy: metformin versus dipeptidyl-peptidase-4-inhibitors (DPP4i) and sulfonylureas (SU). This took place in the National COVID Cohort Collaborative (N3C) longitudinal U.S. cohort of adults with +SARS-CoV-2 result between January 1 2020 to June 1 2021. Findings included hospitalization or ventilation or mortality from COVID-19. Back pain was assessed as a negative control outcome. RESULTS: 6,626 adults with T2DM and +SARS-CoV-2 from 36 sites. Mean age was 60.7 +/- 12.0 years; 48.7% male; 56.7% White, 21.9% Black, 3.5% Asian, and 16.7% Latinx. Mean BMI was 34.1 +/- 7.8kg/m2. Overall 14.5% of the sample was hospitalized; 1.5% received mechanical ventilation; and 1.8% died. In adjusted outcomes, compared to DPP4i, metformin had non-significant associations with reduced need for ventilation (RR 0.68, 0.32-1.44), and mortality (RR 0.82, 0.41-1.64). Compared to SU, metformin was associated with a lower risk of ventilation (RR 0.5, 95% CI 0.28-0.98, p = 0.044) and mortality (RR 0.56, 95%CI 0.33-0.97, p = 0.037). There was no difference in unadjusted or adjusted results of the negative control. CONCLUSIONS: There were clinically significant associations between metformin use and less severe COVID-19 compared to SU, but not compared to DPP4i. New-user studies and randomized trials are needed to assess early outpatient treatment and post-exposure prophylaxis with therapeutics that are safe in adults, children, pregnancy and available worldwide.


Subject(s)
COVID-19 Drug Treatment , Diabetes Mellitus, Type 2 , Dipeptidyl-Peptidase IV Inhibitors , Metformin , Adult , Child , Male , Humans , Middle Aged , Aged , Female , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Retrospective Studies , RNA, Viral/therapeutic use , SARS-CoV-2 , Treatment Outcome , Sulfonylurea Compounds/therapeutic use , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Metformin/therapeutic use , Cohort Studies
9.
J Clin Neurophysiol ; 39(7): 575-582, 2022 Nov 01.
Article in English | MEDLINE | ID: covidwho-2107710

ABSTRACT

PURPOSE: Corona virus disease 2019 (COVID-19) refers to coronavirus disease secondary to SARS-CoV2 infection mainly affecting the human respiratory system. The SARS-CoV2 has been reported to have neurotropic and neuroinvasive features and neurological sequalae with wide range of reported neurological manifestations, including cerebrovascular disease, skeletal muscle injury, meningitis, encephalitis, and demyelination, as well as seizures and focal status epilepticus. In this case series, we analyzed the continuous video-EEGs of patients with COVID-19 infection to determine the presence of specific EEG features or epileptogenicity. METHODS: All continuous video-EEG tracings done on SARS-CoV2-positive patients during a 2-week period from April 5, 2020, to April 19, 2020, were reviewed. The demographics, clinical characteristics, imaging, and EEG features were analyzed and presented. RESULTS: Of 23 patients undergoing continuous video-EEG, 16 were COVID positive and were included. Continuous video-EEG monitoring was ordered for "altered mental status" in 11 of 16 patients and for "clinical seizure" in 5 of 16 patients. None of the patients had seizures or status epilepticus as a presenting symptom of COVID-19 infection. Instead, witnessed clinical seizures developed as results of COVID-19-related medical illness(es): anoxic brain injury, stroke/hemorrhage, lithium (Li) toxicity (because of kidney failure), hypertension, and renal disease. Three patients required therapeutic burst suppression because of focal nonconvulsive status epilepticus, status epilepticus/myoclonus secondary to anoxic injury from cardiac arrest, and one for sedation (and with concomitant EEG abnormalities secondary to Li toxicity). CONCLUSIONS: In this observational case series of 16 patients with COVID-19 who were monitored with continuous video-EEG, most patients experienced a nonspecific encephalopathy. Clinical seizures and electrographic status epilepticus were the second most commonly observed neurological problem.


Subject(s)
COVID-19 , Status Epilepticus , Humans , COVID-19/complications , RNA, Viral/therapeutic use , SARS-CoV-2 , Status Epilepticus/diagnosis , Seizures/diagnosis , Seizures/etiology , Seizures/drug therapy , Electroencephalography/methods
10.
Semin Neurol ; 42(4): 512-522, 2022 Aug.
Article in English | MEDLINE | ID: covidwho-2096904

ABSTRACT

Medication overuse headache (MOH), new daily persistent headache (NDPH), and persistent refractory headache attributed to severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection represent a significant burden in terms of disability and quality of life, and a challenge in terms of definition, pathophysiology, and treatment. Regarding MOH, prevention without withdrawal is not inferior to prevention with withdrawal. Preventive medications like topiramate, onabotulinumtoxinA, and calcitonin gene-related peptide (CGRP) monoclonal antibodies improve chronic migraine with MOH regardless of withdrawal. The differential diagnosis of NDPH is broad and should be carefully examined. There are no guidelines for the treatment of NDPH, but options include a short course of steroids, nerve blocks, topiramate, nortriptyline, gabapentin, CGRP monoclonal antibodies, and onabotulinumtoxinA. The persistence of headache 3 months after SARS-CoV2 infection is a predictor of poor prognosis.


Subject(s)
Botulinum Toxins, Type A , COVID-19 Drug Treatment , COVID-19 , Headache Disorders, Secondary , Headache Disorders , Humans , Calcitonin Gene-Related Peptide/therapeutic use , Botulinum Toxins, Type A/therapeutic use , Quality of Life , Topiramate/therapeutic use , RNA, Viral/therapeutic use , COVID-19/complications , SARS-CoV-2 , Headache Disorders, Secondary/diagnosis , Headache Disorders, Secondary/drug therapy , Headache/diagnosis , Headache/drug therapy , Antibodies, Monoclonal/therapeutic use
11.
Cell Rep Med ; 3(9): 100735, 2022 09 20.
Article in English | MEDLINE | ID: covidwho-1984242

ABSTRACT

We here investigate the impact of antiviral treatments such as remdesivir on intra-host genomic diversity and emergence of SARS-CoV2 variants in patients with a prolonged course of infection. Sequencing and variant analysis performed in 112 longitudinal respiratory samples from 14 SARS-CoV2-infected patients with severe disease progression show that major frequency variants do not generally arise during prolonged infection. However, remdesivir treatment can increase intra-host genomic diversity and result in the emergence of novel major variant species harboring fixed mutations. This is particularly evident in a patient with B cell depletion who rapidly developed mutations in the RNA-dependent RNA polymerase gene following remdesivir treatment. Remdesivir treatment-associated emergence of novel variants is of great interest in light of current treatment guidelines for hospitalized patients suffering from severe SARS-CoV2 disease, as well as the potential use of remdesivir to preventively treat non-hospitalized patients at high risk for severe disease progression.


Subject(s)
COVID-19 Drug Treatment , Coronavirus Infections , Pneumonia, Viral , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Antiviral Agents/adverse effects , Betacoronavirus , Coronavirus Infections/drug therapy , Disease Progression , Humans , Pandemics , Pneumonia, Viral/chemically induced , RNA, Viral/therapeutic use , RNA-Dependent RNA Polymerase , SARS-CoV-2/genetics
12.
An Pediatr (Engl Ed) ; 97(1): 22-29, 2022 Jul.
Article in English | MEDLINE | ID: covidwho-1944180

ABSTRACT

BACKGROUND AND AIMS: To face the rapid spread of SARS-CoV2 coronavirus pandemic, home lockdown in Spain was decreed on 15th March 2020. The main objective of this study is to evaluate the impact of this constraint on glycemic control in children and adolescents with type 1 diabetes mellitus (T1D). PATIENTS AND METHODS: Observational, retrospective study in children and adolescents with T1D users of interstitial glucose monitoring systems. The following information corresponding to the last 2 weeks of lockdown was collected for subsequent comparison with data of 2 weeks prior to quarantine: daily insulin needs, mean interstitial glucose, estimated HbA1c, coefficient of variation (CV), time in range (70-180mg/dl), hypoglycemia (<70 and <54mg/dl) and hyperglycemia (>180 and> 250mg/dl), sensor use and number of blood glucose measurements. Data about meal routines, physical exercise, need for adjustments in therapy, acute complications and lockdown of caregivers were assessed via a survey. RESULTS: 80 patients were studied (mean age 12.61±3.32 years, mean time of evolution of the disease 5.85±3.92 years), 66.2% treated with an insulin pump, users of following glucose monitoring systems: Guardian 3 (65%), FreeStyle Libre (18.8%) and Dexcom G6 (16.2%). Time in range in the cohort increased significantly during confinement (72.1±10.5 vs 74.8±10.5%; P=0.011) with lower time in hypoglycemia both <70mg/dl (4.6±3.2 vs 3.2±2.7%; P<0.001) and <54mg/dl (1.2±1.6 vs 0.7±1.2%; P<0.001) and hyperglycemia >250mg/dl (4.6±3.9 vs 3.7±3.7%; P=0.038). CV also decreased (35.8±6.3 vs 33.1±6.1%; P<0.001). Patients treated with multiple doses of insulin and poorer baseline glycemic control experienced greatest improvement. Daily insulin requirements remained stable. Regular practice of physical exercise and caregivers' confinement did not have a significant impact. CONCLUSIONS: Glycemic control in children and adolescents with T1D improved during quarantine, particularly in those with worse baseline control.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 1 , Hyperglycemia , Hypoglycemia , Adolescent , Blood Glucose , Blood Glucose Self-Monitoring , Child , Communicable Disease Control , Diabetes Mellitus, Type 1/drug therapy , Humans , Hypoglycemia/epidemiology , Hypoglycemia/prevention & control , Insulin/therapeutic use , Insulin, Regular, Human/therapeutic use , RNA, Viral/therapeutic use , Retrospective Studies , SARS-CoV-2
13.
Headache ; 62(7): 903-907, 2022 07.
Article in English | MEDLINE | ID: covidwho-1879033

ABSTRACT

BACKGROUND: Headache can be a prominent feature of Post-Acute Sequelae of SARS-Cov2 infection (PASC) and previous studies have centered around PASC headaches that have resolved within a month of infection. METHODS: We performed a retrospective chart review of 31 adults evaluated at the Stanford Headache Clinic between September 2020 and January 2022 who developed new or worsening headaches after COVID-19 infection that were unresolved at time of evaluation for demographics, medical history, and headache diagnosis. RESULTS: Headache had been present for a mean duration of 7.4±4.8 months after infection. Notably, 25/31 (81%) had a previous history of headache. The specific features of the headache varied considerably, but 23/31 (74%) met International Classification of Headache Disorders, Third Edition (ICHD-3) criteria for migraine, with 20/31 (65%) meeting ICHD-3 criteria for chronic migraine, while only 5/31 (16%) met these criteria before COVID infection. Additionally, full-time employment decreased from 25/31 (81%) to 17/31 (55%). Prior to establishing care at our clinic, 13/18 (72%) of the patients who were started on preventive medications currently indicated for migraine management, reported a decrease in frequency and/or severity of headaches. CONCLUSIONS: Our study presents a group of patients with protracted headache after COVID-19 infection that includes both patients with a previously lower headache burden who largely exhibited chronification from episodic to chronic migraine, as well as patients with no previous history of headache who meet ICHD-3 criteria for headache attributed to a systemic viral illness, mostly with a migrainous phenotype.


Subject(s)
COVID-19 , Migraine Disorders , COVID-19/complications , Headache/epidemiology , Headache/etiology , Humans , Migraine Disorders/diagnosis , Migraine Disorders/epidemiology , Migraine Disorders/etiology , RNA, Viral/therapeutic use , Retrospective Studies , SARS-CoV-2
14.
Horm Metab Res ; 54(8): 540-548, 2022 Aug.
Article in English | MEDLINE | ID: covidwho-1788343

ABSTRACT

The use of standard procedures for the diagnosis of osteoporosis and assessment of fracture risk significantly decreased during the COVID-19 pandemic, while the incidence of fragility fractures was mostly unaltered. Both COVID-19 per se and its treatments are associated with a negative impact on bone health. Preclinical models show that mice infected with SARS-CoV2 even without symptoms display loss of trabecular bone mass two weeks post infection, due to increased numbers of osteoclasts. Osteoporosis medications do not aggravate the clinical course of COVID-19, while preclinical data suggests possible beneficial effects of some therapies. While vitamin D deficiency is clearly associated with a worse clinical course of COVID-19, evidence of improved patient outcome with vitamin D supplementation is lacking. Osteoporosis treatment should not be generally discontinued, and recommendations for substituting therapies are available. Osteoporosis therapies do not interfere with the efficacy or side-effect profiles of COVID-19 vaccines and should not be stopped or indefinitely delayed because of vaccination.


Subject(s)
COVID-19 , Fractures, Bone , Osteoporosis , Animals , COVID-19 Vaccines , Fractures, Bone/complications , Humans , Mice , Osteoporosis/drug therapy , Pandemics , RNA, Viral/therapeutic use , SARS-CoV-2 , Vitamin D/therapeutic use
15.
Rev Esp Enferm Dig ; 114(9): 567-568, 2022 09.
Article in English | MEDLINE | ID: covidwho-1776738

ABSTRACT

SARS-CoV2 infection and vaccination against this virus have been related to the development of autoimmune diseases. We report a case of autoimmune hepatitis (AIH) after SARS-COV2 vaccine. Male, 76 years old, with a history of hepatic cirrhosis secondary to primary biliary cholangitis (PBC), compensated, treated with ursodeoxycholic acid and obeticholic acid. The patient received the third dose of the SARS-CoV2 vaccine (BioNTech/Pfizer) in December 2021. In subsequent analytical control, the patient presented altered liver test, with elevation of ALT and AST. Ultrasound was performed, without alterations, and viral causes were ruled out. IgG elevation and positive antinuclear antibodies were observed. A liver biopsy was performed, with findings of intense interface and lobular hepatitis and areas of centrilobular necrosis. The inflammation was predominantly lymphoplasmacytic. The patient was diagnosed with AIH and initiated therapy with steroids and azathioprine, currently with an adequate response. AIH is an immune-mediated disease of uncertain etiology. Cases of AIH with SARS-CoV2 vaccination as a possible trigger have recently been published, with characteristics similar to ours. Some of them had a history of autoimmune pathology, such as this case (PBC). Therefore, it is suggested that vaccination can induce the development of autoimmune pathology in patients at risk. Our reported case reinforces the hypothesis of an association between AIH and the SARS-CoV2 vaccine.


Subject(s)
COVID-19 Vaccines , COVID-19 , Hepatitis, Autoimmune , Liver Cirrhosis, Biliary , Aged , COVID-19 Vaccines/adverse effects , Hepatitis, Autoimmune/drug therapy , Hepatitis, Autoimmune/etiology , Humans , Liver Cirrhosis, Biliary/drug therapy , Male , RNA, Viral/therapeutic use , SARS-CoV-2
16.
Neurol Neurochir Pol ; 56(2): 163-170, 2022.
Article in English | MEDLINE | ID: covidwho-1753880

ABSTRACT

INTRODUCTION: The aim of this study was to assess the clinical profiles and outcomes of patients with confirmed COVID-19 infection and acute ischaemic stroke (AIS) treated with mechanical thrombectomy (MT) at the Comprehensive Stroke Centre (CSC) of the University Hospital in Krakow. CLINICAL RATIONALE FOR THE STUDY: COVID-19 is a risk factor for AIS and worsens prognosis in patients with large artery occlusions. During the pandemic, the global number of MT has dropped. At the same time, studies assessing outcomes of this treatment in COVID-19-associated AIS have produced divergent results. MATERIAL AND METHODS: In this single-centre study, we retrospectively analysed and compared the clinical profiles (age, sex, presence of cardiovascular risk factors, neurological deficit at admission), stroke size (measured using postprocessing analysis of perfusion CT with RAPID software), time from stroke onset to arrival at the CSC, time from arrival at the CSC to groin puncture, treatment with intravenous thrombolysis, length of hospitalisation, laboratory test results, and short-term outcomes (measured with Thrombolysis in Cerebral Infarction scale, modified Rankin Scale and National Health Institute Stroke Scale) in patients with AIS treated with MT during the pandemic. A comparison between patients with and without concomitant SARS-CoV2 infection was then performed. RESULTS: There were no statistically significant differences between 15 COVID (+) and 167 COVID (-) AIS patients treated with AIS with respect to clinical profiles (p > 0.05), stroke size (p > 0.05) or outcomes (NIHSS at discharge, 8.1 (SD = 7.1) vs. 8.8 (SD = 9.6), p = 0.778, mRS at discharge 2.9 (SD = 2) vs. 3.1 (SD = 2.1), p = 0.817, death rate 6.7% vs. 12.6%, p = 0.699). There was a significant difference between patients with and without COVID-19 concerning time from arrival at the CSC to groin puncture [104.27 (SD = 51.47) vs. 97.63 (SD = 156.94) min., p = 0.044] and the length of hospitalisation [23.7 (SD = 11.9) vs. 10.5 (SD = 6.9) days, p < 0.001]. CONCLUSION: In AIS patients treated with MT, concomitant SARS-CoV2 infection did not affect the outcome. Our observations need to be confirmed in larger, and preferably multicentre, studies.


Subject(s)
Brain Ischemia , COVID-19 , Ischemic Stroke , Stroke , Brain Ischemia/diagnostic imaging , Brain Ischemia/surgery , COVID-19/complications , Humans , Ischemic Stroke/diagnostic imaging , Ischemic Stroke/etiology , Ischemic Stroke/surgery , RNA, Viral/therapeutic use , Retrospective Studies , SARS-CoV-2 , Stroke/diagnostic imaging , Stroke/etiology , Stroke/surgery , Thrombectomy/methods , Treatment Outcome
17.
Dermatol Ther ; 35(5): e15430, 2022 05.
Article in English | MEDLINE | ID: covidwho-1731140

ABSTRACT

SARS-CoV2 vaccines were approved without long-term monitoring due to emergent situations. This has raised some issues about timing and protocol of receiving vaccines in specific situations including patients with chronic inflammatory disorders such as psoriasis. Here, we present different aspects of SARS-CoV-2 infection and vaccination in psoriasis patients and aim to provide solutions to overcome the potential challenges. In brief, the benefits of vaccination outweigh the potential risk; vaccine-triggered de novo or flares of psoriasis is uncommon. As such, all psoriasis patients, especially those receiving systemic treatments including anti tumor necrosis factor agents, are strongly recommended to get SARS-CoV-2 vaccines. It is recommended that new immunosuppressive/immunomodulatory therapies be initiated at least 1 week after the second SARS-CoV-2 vaccine dose, if possible. In addition, in severe and active forms of psoriasis, it is better to delay vaccination until stabilization of the disease.


Subject(s)
COVID-19 , Psoriasis , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , Humans , Psoriasis/drug therapy , RNA, Viral/therapeutic use , SARS-CoV-2 , Vaccination
18.
Eur J Neurol ; 29(5): 1538-1544, 2022 05.
Article in English | MEDLINE | ID: covidwho-1691579

ABSTRACT

BACKGROUND AND PURPOSE: SARS-CoV2 vaccination is recommended for patients with multiple sclerosis (pwMS), but response may be limited by disease-modifying-treatments (DMTs). The aim of this study was to compare the rates of humoral immune response and safety of SARS-CoV-2 vaccines in pwMS and healthy controls (HCs). METHODS: In this multicenter prospective study on 456 pwMS and 116 HCs, SARS-CoV-2-IgG response was measured 3 months after the first vaccine dose. The primary endpoint was defined as proportion of patients developing antibodies (seroconversion). Secondary endpoints included antibody level, safety and efficacy. RESULTS: Compared to 97.4% in HCs, seroconversion occurred in 96.7% (88/91) untreated pwMS, 97.1% of patients (135/139) on immunomodulatory DMTs and 61.1% (138/226; p < 0.001) on immunosuppressive DMTs. Seroconversion was lowest in patients on antiCD20 monoclonal antibodies (CD20 mAbs; 52.6%) followed by sphingosine-1-phosphate-receptor-modulators (S1PMs; 63.6%). In the S1PM subgroup, seroconversion increased with lymphocyte count (odds ratio [OR] 1.31 per 0.1 G/L; p = 0.035). In pwMS on CD20 mAbs, B-cell depletion decreased seroconversion (OR 0.52; p = 0.038), whereas time since last DMT did not. Safety of SARS-CoV-2 vaccines in pwMS was excellent. CONCLUSIONS: Humoral response to SARS-CoV2 vaccines in pwMS is generally excellent. While reduced by immunosuppressive DMTs, most importantly by B-cell-depleting CD20 mAbs and S1PMs, seroconversion is still expected in the majority of patients. SARS-CoV2 vaccination should be offered to every MS patient.


Subject(s)
COVID-19 , Multiple Sclerosis , Antibodies, Viral , Austria , COVID-19/prevention & control , COVID-19 Vaccines/therapeutic use , Humans , Immunity, Humoral , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis/drug therapy , Prospective Studies , RNA, Viral/therapeutic use , SARS-CoV-2
19.
J Neuroophthalmol ; 42(2): 251-255, 2022 06 01.
Article in English | MEDLINE | ID: covidwho-1596393

ABSTRACT

BACKGROUND: The opsoclonus-myoclonus-ataxia syndrome (OMAS) represents a pathophysiology and diagnostic challenge. Although the diverse etiologies likely share a common mechanism to generate ocular, trunk, and limb movements, the underlying cause may be a paraneoplastic syndrome, as the first sign of cancer, or may be a postinfectious complication, and thus, the outcome depends on identifying the trigger mechanism. A recent hypothesis suggests increased GABAA receptor sensitivity in the olivary-oculomotor vermis-fastigial nucleus-premotor saccade burst neuron circuit in the brainstem. Therefore, OMAS management will focus on immunosuppression and modulation of GABAA hypersensitivity with benzodiazepines. METHODS: We serially video recorded the eye movements at the bedside of 1 patient with SARS-CoV-2-specific Immunoglobulin G (IgG) serum antibodies, but twice-negative nasopharyngeal reverse transcription polymerase chain reaction (RT-PCR). We tested cerebrospinal fluid (CSF), serum, and nasopharyngeal samples. After brain MRI and chest, abdomen, and pelvis CT scans, we treated our patient with clonazepam and high-dose Solu-MEDROL, followed by a rituximab infusion after her formal eye movement analysis 10 days later. RESULTS: The recordings throughout her acute illness demonstrated different eye movement abnormalities. While on high-dose steroids and clonazepam, she initially had macrosaccadic oscillations, followed by brief ocular flutter during convergence the next day; after 10 days, she had bursts of opsoclonus during scotopic conditions with fixation block but otherwise normal eye movements. Concern for a suboptimal response to high-dose Solu-MEDROL motivated an infusion of rituximab, which induced remission. An investigation for a paraneoplastic etiology was negative. CSF testing showed elevated neuron-specific enolase. Serum IgG to Serum SARS-CoV2 IgG was elevated with negative RT-PCR nasopharyngeal testing. CONCLUSION: A recent simulation model of macrosaccadic oscillations and OMAS proposes a combined pathology of brainstem and cerebellar because of increased GABAA receptor sensitivity. In this case report, we report 1 patient with elevated CSF neuronal specific enolase, macrosaccadic oscillations, ocular flutter, and OMAS as a SARS-CoV-2 postinfectious complication. Opsoclonus emerged predominantly with fixation block and suppressed with fixation, providing support to modern theories on the mechanism responsible for these ocular oscillations involving cerebellar-brainstem pathogenesis.


Subject(s)
COVID-19 , Cerebellar Ataxia , Ocular Motility Disorders , Opsoclonus-Myoclonus Syndrome , COVID-19/complications , Cerebellar Ataxia/complications , Clonazepam/therapeutic use , Female , Humans , Immunoglobulin G , Methylprednisolone Hemisuccinate/therapeutic use , Ocular Motility Disorders/diagnosis , Ocular Motility Disorders/drug therapy , Ocular Motility Disorders/etiology , Opsoclonus-Myoclonus Syndrome/diagnosis , Opsoclonus-Myoclonus Syndrome/drug therapy , Opsoclonus-Myoclonus Syndrome/etiology , RNA, Viral/therapeutic use , Receptors, GABA-A/therapeutic use , Rituximab/therapeutic use , SARS-CoV-2
20.
Cancer Invest ; 40(5): 406-412, 2022 May.
Article in English | MEDLINE | ID: covidwho-1364664

ABSTRACT

BACKGROUND: The impact of systemic anticancer treatments on SARS-CoV-2-related mortality is still debatable. METHODS: By a retrospective analysis of patients with non-small-cell lung cancer (NSCLC) treated with first-line Pembrolizumab or in combination with chemotherapy (ChT) during the first surge of the pandemic. RESULTS: The adjusted risk of death was higher in patients treated with ChT + Pembrolizumab (HR 4.6, 1.2-17.4, p = 0.02). The SARS-CoV-2-related mortality rate was higher in patients treated with ChT + Pembrolizumab (p = 0.03), ≥70 years (p = 0.03) and current smokers (p = 0.17). CONCLUSIONS: The addition of ChT to immunotherapy could be associated with increased risk of mortality and higher SARS-CoV-2-related mortality rate.


Subject(s)
COVID-19 , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Immunotherapy , Lung Neoplasms/drug therapy , RNA, Viral/therapeutic use , Retrospective Studies , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL