Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
2.
Nat Commun ; 13(1): 460, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075154

ABSTRACT

The SARS-CoV-2 Delta variant has spread rapidly worldwide. To provide data on its virological profile, we here report the first local transmission of Delta in mainland China. All 167 infections could be traced back to the first index case. Daily sequential PCR testing of quarantined individuals indicated that the viral loads of Delta infections, when they first become PCR-positive, were on average ~1000 times greater compared to lineage A/B infections during the first epidemic wave in China in early 2020, suggesting potentially faster viral replication and greater infectiousness of Delta during early infection. The estimated transmission bottleneck size of the Delta variant was generally narrow, with 1-3 virions in 29 donor-recipient transmission pairs. However, the transmission of minor iSNVs resulted in at least 3 of the 34 substitutions that were identified in the outbreak, highlighting the contribution of intra-host variants to population-level viral diversity during rapid spread.


Subject(s)
COVID-19/transmission , Contact Tracing/methods , Disease Outbreaks/prevention & control , SARS-CoV-2/isolation & purification , Animals , COVID-19/epidemiology , COVID-19/virology , Chlorocebus aethiops , Humans , RNA-Seq/methods , Reverse Transcriptase Polymerase Chain Reaction , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Time Factors , Vero Cells , Viral Load/genetics , Viral Load/physiology , Virus Replication/genetics , Virus Replication/physiology , Virus Shedding/genetics , Virus Shedding/physiology
3.
J Med Virol ; 94(4): 1402-1411, 2022 04.
Article in English | MEDLINE | ID: mdl-34766661

ABSTRACT

Patients with COVID-19 may be recurrence positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA after being cured and discharged from the hospital. The aim of this study was to explore independent influencing factors as markers for predicting positive SARS-CoV-2 RNA recurrence. The study included 601 COVID-19 patients who were cured and discharged from the Public and Health Clinic Centre of Chengdu from January 2020 to March 2021, and the recurrence positive of patients within 6 weeks after SARS-CoV-2 RNA turned negative was followed up. We used propensity score matching to eliminate the influence of confounding factors, and multivariate Logistic regression analysis was used to determine the independent influencing factors for positive SARS-CoV-2 RNA recurrence. Multivariate Logistic regression showed that the elevated serum potassium (odds ratio [OR] = 6.537, 95% confidence interval [CI]: 1.864-22.931, p = 0.003), elevated blood chlorine (OR = 1.169, 95% CI: 1.032-1.324, p = 0.014) and elevated CD3+ CD4+ count (OR = 1.003, 95% CI: 1.001-1.004, p < 0.001) were identified as independent risk factors for positive SARS-CoV-2 RNA recurrence (p < 0.05). The difference in virus shedding duration (OR = 1.049, 95% CI: 1.000-1.100, p = 0.05) was borderline statistically significant. For sensitivity analysis, we included virus shedding duration as a categorical variable in the model again and found that the OR value related to recurrence positively increased with delayed virus shedding duration, and the trend test showed a statistical difference (P trend = 0.03). Meanwhile, shortening of activated partial prothrombinase time (OR = 0.908, 95% CI: 0.824-1.000, p = 0.049) was identified as an independent protection factor for SARS-CoV-2 RNA recurrence positive. We have identified independent factors that affect the recurrence of SARS-CoV-2 RNA positive. It is recommended that doctors pay attention to these indicators when first admitted to the hospital.


Subject(s)
COVID-19/virology , RNA, Viral/isolation & purification , SARS-CoV-2/physiology , Virus Shedding/physiology , Adult , Antiviral Agents/administration & dosage , COVID-19/epidemiology , China , Female , Hospitalization , Humans , Logistic Models , Male , Middle Aged , Propensity Score , Recurrence , Retrospective Studies , Risk Factors , Virus Shedding/drug effects
4.
JCI Insight ; 7(2)2022 01 25.
Article in English | MEDLINE | ID: mdl-34871181

ABSTRACT

Isolation guidelines for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are largely derived from data collected prior to the emergence of the delta variant. We followed a cohort of ambulatory patients with postvaccination breakthrough SARS-CoV-2 infections with longitudinal collection of nasal swabs for SARS-CoV-2 viral load quantification, whole-genome sequencing, and viral culture. All delta variant infections in our cohort were symptomatic, compared with 64% of non-delta variant infections. Symptomatic delta variant breakthrough infections were characterized by higher initial viral load, longer duration of virologic shedding by PCR, greater likelihood of replication-competent virus at early stages of infection, and longer duration of culturable virus compared with non-delta variants. The duration of time since vaccination was also correlated with both duration of PCR positivity and duration of detection of replication-competent virus. Nonetheless, no individuals with symptomatic delta variant infections had replication-competent virus by day 10 after symptom onset or 24 hours after resolution of symptoms. These data support US CDC isolation guidelines as of November 2021, which recommend isolation for 10 days or until symptom resolution and reinforce the importance of prompt testing and isolation among symptomatic individuals with delta breakthrough infections. Additional data are needed to evaluate these relationships among asymptomatic and more severe delta variant breakthrough infections.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/genetics , COVID-19/metabolism , SARS-CoV-2/physiology , Virus Replication , Virus Shedding/physiology , Adult , COVID-19/prevention & control , Female , Humans , Male , Middle Aged , Time Factors
5.
Sci Rep ; 11(1): 24183, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34921199

ABSTRACT

COVID-19 has restricted singing in communal worship. We sought to understand variations in droplet transmission and the impact of wearing face masks. Using rapid laser planar imaging, we measured droplets while participants exhaled, said 'hello' or 'snake', sang a note or 'Happy Birthday', with and without surgical face masks. We measured mean velocity magnitude (MVM), time averaged droplet number (TADN) and maximum droplet number (MDN). Multilevel regression models were used. In 20 participants, sound intensity was 71 dB for speaking and 85 dB for singing (p < 0.001). MVM was similar for all tasks with no clear hierarchy between vocal tasks or people and > 85% reduction wearing face masks. Droplet transmission varied widely, particularly for singing. Masks decreased TADN by 99% (p < 0.001) and MDN by 98% (p < 0.001) for singing and 86-97% for other tasks. Masks reduced variance by up to 48%. When wearing a mask, neither singing task transmitted more droplets than exhaling. In conclusion, wide variation exists for droplet production. This significantly reduced when wearing face masks. Singing during religious worship wearing a face mask appears as safe as exhaling or talking. This has implications for UK public health guidance during the COVID-19 pandemic.


Subject(s)
COVID-19/transmission , Disease Transmission, Infectious/prevention & control , Face , Masks , Singing/physiology , Adult , COVID-19/epidemiology , COVID-19/virology , Cross-Sectional Studies , Exhalation/physiology , Female , Humans , Male , Pandemics/prevention & control , Risk Factors , SARS-CoV-2/physiology , Virus Shedding/physiology
6.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34625475

ABSTRACT

The pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global threat to human health and life. A useful pathological animal model accurately reflecting human pathology is needed to overcome the COVID-19 crisis. In the present study, COVID-19 cynomolgus monkey models including monkeys with underlying diseases causing severe pathogenicity such as metabolic disease and elderly monkeys were examined. Cynomolgus macaques with various clinical conditions were intranasally and/or intratracheally inoculated with SARS-CoV-2. Infection with SARS-CoV-2 was found in mucosal swab samples, and a higher level and longer period of viral RNA was detected in elderly monkeys than in young monkeys. Pneumonia was confirmed in all of the monkeys by computed tomography images. When monkeys were readministrated SARS-CoV-2 at 56 d or later after initial infection all of the animals showed inflammatory responses without virus detection in swab samples. Surprisingly, in elderly monkeys reinfection showed transient severe pneumonia with increased levels of various serum cytokines and chemokines compared with those in primary infection. The results of this study indicated that the COVID-19 cynomolgus monkey model reflects the pathophysiology of humans and would be useful for elucidating the pathophysiology and developing therapeutic agents and vaccines.


Subject(s)
COVID-19/immunology , Disease Models, Animal , Macaca fascicularis/immunology , Primate Diseases/immunology , SARS-CoV-2/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19/virology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lung/diagnostic imaging , Lung/immunology , Lung/virology , Macaca fascicularis/virology , Male , Primate Diseases/virology , SARS-CoV-2/physiology , Tomography, X-Ray Computed/methods , Virus Shedding/immunology , Virus Shedding/physiology
7.
Viruses ; 13(10)2021 10 17.
Article in English | MEDLINE | ID: mdl-34696519

ABSTRACT

This study investigated the infectivity of severe acute respiratory syndrome (SARS-CoV-2) in individuals who re-tested positive for SARS-CoV-2 RNA after recovering from their primary illness. We investigated 295 individuals with re-positive SARS-CoV-2 polymerase chain reaction (PCR) test results and 836 of their close contacts. We attempted virus isolation in individuals with re-positive SARS-CoV-2 PCR test results using cell culture and confirmed the presence of neutralizing antibodies using serological tests. Viral culture was negative in all 108 individuals with re-positive SARS-CoV-2 PCR test results in whom viral culture was performed. Three new cases of SARS-CoV-2 infection were identified among household contacts using PCR. Two of the three new cases had had contact with the index patient during their primary illness, and all three had antibody evidence of past infection. Thus, there was no laboratory evidence of viral shedding and no epidemiological evidence of transmission among individuals with re-positive SARS-CoV-2 PCR test results.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/immunology , Reinfection/virology , SARS-CoV-2/immunology , Virus Shedding/physiology , Adolescent , Adult , Aged , COVID-19/diagnosis , COVID-19/transmission , COVID-19 Serological Testing , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Polymerase Chain Reaction , Reinfection/immunology , Republic of Korea , Retrospective Studies , SARS-CoV-2/isolation & purification , Severity of Illness Index , Young Adult
8.
J Clin Lab Anal ; 35(9): e23923, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34390043

ABSTRACT

BACKGROUND: The dynamic alteration and comparative study of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA shedding pattern during treatment are limited. This study explores the potential risk factors influencing prolonged viral shedding in COVID-19. METHODS: A total of 126 COVID-19 patients were enrolled in this retrospective longitudinal study. A multivariate logistic regression analysis was carried out to estimate the potential risk factors. RESULTS: 38.1% (48/126) cases presented prolonged respiratory tract viral shedding, and 30 (23.8%) cases presented prolonged rectal swab viral shedding. Obesity (OR, 3.31; 95% CI, 1.08-10.09), positive rectal swab (OR, 3.43; 95% CI, 1.53-7.7), treatment by lopinavir/ritonavir with chloroquine phosphate (OR, 2.5; 95% CI, 1.04-6.03), the interval from onset to antiviral treatment more than 7 days (OR, 2.26; 95% CI, 1.04-4.93), lower CD4+ T cell (OR, 0.92; 95% CI, 0.86-0.99) and higher NK cells (OR, 1.11; 95% CI, 1.02-1.20) were significantly associated with prolonged respiratory tract viral shedding. CD3-CD56+ NK cells (OR, 0.87; 95% CI, 0.76-0.99) were related with prolonged fecal shedding. CONCLUSIONS: Obesity, delayed antiviral treatment, and positive SARS-CoV-2 for stool were independent risk factors for prolonged SARS-CoV-2 RNA shedding of the respiratory tract. A combination of LPV/r and abidol as the initial antiviral regimen was effective in shortening the duration of viral shedding compared with LPV/r combined with chloroquine phosphate. CD4+ T cell and NK cells were significantly associated with prolonged viral shedding, and further studies are to be warranted to determine the mechanism of immunomodulatory response in virus clearance.


Subject(s)
COVID-19/virology , Feces/virology , SARS-CoV-2/physiology , Virus Shedding/physiology , Adult , Animals , Antiviral Agents/administration & dosage , CD4 Lymphocyte Count , COVID-19/epidemiology , Chloroquine/administration & dosage , Chloroquine/adverse effects , Chloroquine/analogs & derivatives , Female , Humans , Killer Cells, Natural , Longitudinal Studies , Lopinavir/administration & dosage , Lynx , Male , Obesity/epidemiology , Respiratory System/virology , Retrospective Studies , Risk Factors , Ritonavir/administration & dosage , Time Factors , Virus Shedding/drug effects
9.
Vet Microbiol ; 261: 109204, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34399298

ABSTRACT

Whether subclinical shedding of canine parvovirus (CPV) by cats might contribute to the epidemiology of canine CPV infections, particularly in facilities housing both cats and dogs, requires clarification. Conflicting results are reported to date. Using conventional PCR (cPCR) to amplify the VP2 gene, shedding of the CPV variants (CPV-2a, 2b, 2c) by healthy cats in multi-cat environments was reportedly common in Europe but rare in Australia. The aim of this study was to determine whether low-level faecal CPV shedding occurs in multi-cat environments in Australia and Italy using a TaqMan real-time PCR to detect Carnivore protoparvovirus 1 (CPV and feline parvovirus, FPV) DNA, and minor-groove binder probe real-time PCR assay to differentiate FPV and CPV types and to characterize CPV variants. In total, 741 non-diarrhoeic faecal samples from shelters in Australia (n = 263) and from shelters or cat colonies in Italy (n = 478) were tested. Overall, Carnivore protoparvovirus 1 DNA was detected in 49 of 741 (6.61 %) samples. Differentiation was possible for 31 positive samples. FPV was most common among positive samples (28/31, 90.3 %). CPV was detected in 4/31 samples (12.9 %) including CPV-2a in one sample, CPV-2b in another and co-infections of FPV/CPV-2b and CPV-2a/CPV-2b in the remaining two samples. A high rate of subclinical FPV infection was detected in one shelter during an outbreak of feline panleukopenia, during which 21 of 22 asymptomatic cats (95.5 %) sampled were shedding FPV. Faecal shedding of CPV by cats in multi-cat environments is uncommon suggesting that domestic cats are not significant reservoirs of CPV.


Subject(s)
Dog Diseases/epidemiology , Feces/virology , Parvoviridae Infections/veterinary , Parvovirus, Canine/physiology , Virus Shedding/physiology , Animals , Capsid Proteins/genetics , Cat Diseases/pathology , Cat Diseases/virology , Cats , Disease Reservoirs/veterinary , Dog Diseases/transmission , Dog Diseases/virology , Dogs , Parvoviridae Infections/transmission , Parvoviridae Infections/virology , Parvovirus, Canine/genetics , Real-Time Polymerase Chain Reaction
10.
Am J Epidemiol ; 190(12): 2536-2543, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34216204

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection worldwide, but reports of temporal changes in the risk of transmission among close contacts has been scarce. This study aimed to examine an association between the viral load trajectory and transmission risk to develop a better control strategy for the disease spread. We conducted a household-based prospective cohort study in Biliran Province, the Philippines, and enrolled 451 participants to observe the development of acute respiratory infection. Including the cases found at the health-care facility, we analyzed the data of viral loads with symptom records obtained from 172 followed participants who had household member positive for RSV with a rapid test during an RSV outbreak in 2018-2019. We developed a model estimating a temporal change in the viral shedding from the infection and evaluated transmission dynamics. We found that most transmission events occurred within approximately 7 days of the household exposure, including potential presymptomatic transmissions. The inferred risk of infection among those younger than 5 years was 3.5 times higher than that of those older than 5 years. This finding suggested that the initial week after the household exposure is particularly important for preventing RSV spread.


Subject(s)
Family Characteristics , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/transmission , Viral Load/physiology , Virus Shedding/physiology , Age Factors , Child, Preschool , Female , Humans , Infant , Male , Models, Theoretical , Philippines/epidemiology , Prospective Studies
11.
Emerg Microbes Infect ; 10(1): 1156-1168, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34060982

ABSTRACT

ABSTRACTThe risk of secondary infection with SARS-CoV-2 and influenza A virus is becoming a practical problem that must be addressed as the flu season merges with the COVID-19 pandemic. As SARS-CoV-2 and influenza A virus have been found in patients, understanding the in vivo characteristics of the secondary infection between these two viruses is a high priority. Here, hACE2 transgenic mice were challenged with the H1N1 virus at a nonlethal dose during the convalescent stage on 7 and 14 days post SARS-CoV-2 infection, and importantly, subsequent H1N1 infection showed enhanced viral shedding and virus tissue distribution. Histopathological observation revealed an extensive pathological change in the lungs related to H1N1 infection in mice recovered from SARS-CoV-2 infection, with severe inflammation infiltration and bronchiole disruption. Moreover, upon H1N1 exposure on 7 and 14 dpi of SARS-CoV-2 infection, the lymphocyte population activated at a lower level with T cell suppressed in both PBMC and lung. These findings will be valuable for evaluating antiviral therapeutics and vaccines as well as guiding public health work.


Subject(s)
Acute Lung Injury/pathology , Angiotensin-Converting Enzyme 2/genetics , COVID-19/pathology , Orthomyxoviridae Infections/pathology , Acute Lung Injury/virology , Animals , COVID-19/therapy , Coinfection/pathology , Coinfection/virology , Cytokines/blood , Disease Models, Animal , Female , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Lung/pathology , Lymphocyte Count , Lymphocytes/immunology , Mice , Mice, Transgenic , Orthomyxoviridae Infections/therapy , SARS-CoV-2/isolation & purification , Viral Load , Virus Replication/physiology , Virus Shedding/physiology
12.
mSphere ; 6(3)2021 05 19.
Article in English | MEDLINE | ID: mdl-34011679

ABSTRACT

Information regarding the infectivity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in asymptomatic carriers is scarce. In order to determine the duration of infectivity and its correlation with reverse transcription-PCR (RT-PCR) results and time since initial positive PCR test in this population, we evaluated SARS-CoV-2 cell infectivity in nasopharyngeal samples longitudinally obtained from asymptomatic carriers who disembarked from a cruise ship during a COVID-19 outbreak. Of 166 nasopharyngeal samples collected from 39 asymptomatic carriers every 48 h until two consecutive negative PCR test results were obtained, SARS-CoV-2 was successfully isolated from 9 PCR-positive samples which were obtained from 7 persons (18%; 7/39). Viable viruses were isolated predominantly within 7 days after the initial positive PCR test, except for one person who shed viable virus until day 15. The median crossing point (Cp) value of RT-PCR of culture-positive samples was 24.6 (interquartile range [IQR], 20.4 to 25.8; range, 17.9 to 30.3), and Cp values were significantly associated with isolation of viable virus (odds ratio, 0.496; 95% confidence interval [CI], 0.329 to 0.747; P value, 0.001), which was consistent with existing data for symptomatic patients. Genome sequence analysis of SARS-CoV-2 samples consecutively obtained from a person who shed viable virus for 15 days identified the emergence of two novel single nucleotide variants (C8626T transition and C18452T transition) in the sample collected on day 15, with the latter corresponding to an amino acid substitution in nonstructural protein 14. The impact of these mutations on prolonged viable-virus shedding is unclear. These findings underscore the potential role of asymptomatic carriers in transmission.IMPORTANCE A growing number of studies suggest the potential role of asymptomatic SARS-CoV-2 carriers as a major driver of the COVID-19 pandemic; however, virological assessment of asymptomatic infection has largely been limited to reverse transcription-PCR (RT-PCR), which can be persistently positive without necessarily indicating the presence of viable virus (e.g., replication-competent virus). Here, we evaluated the infectivity of asymptomatic SARS-CoV-2 carriers by detecting SARS-CoV-2-induced cytopathic effects on Vero cells using longitudinally obtained nasopharyngeal samples from asymptomatic carriers. We show that asymptomatic carriers can shed viable virus until 7 days after the initial positive PCR test, with one outlier shedding until day 15. The crossing point (Cp) value of RT-PCR was the leading predictive factor for virus viability. These findings provide additional insights into the role of asymptomatic carriers as a source of transmission and highlight the importance of universal source control measures, along with isolation policy for asymptomatic carriers.


Subject(s)
Asymptomatic Infections/epidemiology , COVID-19/transmission , Virus Shedding/physiology , Adolescent , Adult , Aged , Animals , COVID-19 Nucleic Acid Testing/methods , Cell Line , Child , Chlorocebus aethiops , Female , Genome, Viral/genetics , Humans , Male , Middle Aged , Nasopharynx/virology , SARS-CoV-2/genetics , Vero Cells , Whole Genome Sequencing , Young Adult
13.
J Med Virol ; 93(7): 4570-4575, 2021 07.
Article in English | MEDLINE | ID: mdl-33830520

ABSTRACT

Inpatient coronavirus disease 2019 (COVID-19) cases present enormous costs to patients and health systems in the United States. Many hospitalized patients may continue testing COVID-19 positive even after the resolution of symptoms. Thus, a pressing concern for clinicians is the safety of discharging these asymptomatic patients if they have any remaining infectivity. This case report explores the viral viability in a patient with persistent COVID-19 over the course of a 2-month hospitalization. Positive nasopharyngeal swab samples were collected and isolated in the laboratory and analyzed by quantitative reverse-transcription polymerase chain reactions (qRT-PCR), and serology was tested for neutralizing antibodies throughout the hospitalization period. The patient experienced waning symptoms by hospital day 40 and had no viable virus growth by hospital day 41, suggesting no risk of infectivity, despite positive RT-PCR results which prolonged his hospital stay. Notably, this case showed infectivity for at least 24 days after disease onset, which is longer than the discontinuation of transmission-based precautions recommended by the Center for Disease Control and Prevention. Thus, our findings suggest that the timeline for discontinuing transmission-based precautions may need to be extended for patients with severe and prolonged COVID-19 disease. Additional large-scale studies are needed to draw definitive conclusions on the appropriate clinical management for these patients. ​.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19 Nucleic Acid Testing , COVID-19/diagnosis , Virus Shedding/physiology , Aged , Asymptomatic Infections , Humans , Male , RNA, Viral/analysis , Reverse Transcriptase Polymerase Chain Reaction , SARS-CoV-2/genetics , SARS-CoV-2/immunology
15.
Eur J Cancer ; 147: 154-160, 2021 04.
Article in English | MEDLINE | ID: mdl-33676266

ABSTRACT

The worldwide spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the associated infectious coronavirus disease (COVID-19) has posed a unique challenge to medical staff, patients and their families. Patients with cancer, particularly those with haematologic malignancies, have been identified to be at high risk to develop severe COVID-19. Since publication of our previous guideline on evidence-based management of COVID-19 in patients with cancer, research efforts have continued and new relevant data has come to light, maybe most importantly in the field of vaccination studies. Therefore, an update of our guideline on several clinically important topics is warranted. Here, we provide a concise update of evidence-based recommendations for rapid diagnostics, viral shedding, vaccination and therapy of COVID-19 in patients with cancer. This guideline update was prepared by the Infectious Diseases Working Party (AGIHO) of the German Society for Haematology and Medical Oncology by critically reviewing the currently available data on these topics applying evidence-based medicine criteria.


Subject(s)
COVID-19 Testing/standards , COVID-19 Vaccines/therapeutic use , COVID-19 , Neoplasms , SARS-CoV-2/physiology , Virus Shedding/physiology , Antiviral Agents/therapeutic use , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/therapy , COVID-19/virology , COVID-19 Testing/methods , Evidence-Based Medicine/standards , Evidence-Based Medicine/statistics & numerical data , Germany/epidemiology , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/epidemiology , Hematologic Neoplasms/therapy , Hematologic Neoplasms/virology , Hematology/organization & administration , Hematology/standards , Humans , Immunization, Passive/methods , Immunization, Passive/standards , Infectious Disease Medicine/organization & administration , Infectious Disease Medicine/standards , Medical Oncology/organization & administration , Medical Oncology/standards , Neoplasms/diagnosis , Neoplasms/epidemiology , Neoplasms/therapy , Neoplasms/virology , SARS-CoV-2/immunology , Societies, Medical/standards , Vaccination/methods , Vaccination/standards , COVID-19 Serotherapy
16.
Am J Epidemiol ; 190(5): 701-706, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33564827

ABSTRACT

The emergence of the novel coronavirus disease 2019 (COVID-19) presented the field of reproductive medicine with many challenges due to an absence of data to guide clinical decision-making and inform patient counseling and management in the early days of the pandemic. Epidemiological studies rapidly filled key gaps in our understanding of the susceptibility of reproductive-aged women to the virus, transmission dynamics during pregnancy and lactation, and the effect of infection during the prenatal, pregnancy, and postpartum periods. This data guided the development of clinical guidelines written by the American Society for Reproductive Medicine as patients and clinicians navigated reproductive decisions during a time of uncertainty. We present a review of epidemiologic studies published between March and December 2020 that have directly informed prenatal and fertility care during the COVID-19 pandemic. Despite a significant increase in our knowledge base over the past year, many questions remain about the impact of COVID-19 on conception, pregnancy, fetal development, and lactation. In the future, a commitment toward inclusion of pregnant persons and those attempting pregnancy in the design of observational and interventional trials is necessary to gain earlier insights about outcomes and assist providers and patients in making data-driven decisions.


Subject(s)
COVID-19/epidemiology , COVID-19/prevention & control , Adolescent , Adult , Breast Feeding , COVID-19/therapy , COVID-19/transmission , Female , Humans , Infectious Disease Transmission, Vertical/prevention & control , Middle Aged , Pandemics , Pregnancy , Pregnancy Complications, Infectious/epidemiology , Prenatal Care , Risk Factors , SARS-CoV-2 , Virus Shedding/physiology , Young Adult
17.
PLoS Comput Biol ; 17(1): e1008609, 2021 01.
Article in English | MEDLINE | ID: mdl-33513139

ABSTRACT

A key parameter in epidemiological modeling which characterizes the spread of an infectious disease is the generation time, or more generally the distribution of infectiousness as a function of time since infection. There is increasing evidence supporting a prolonged viral shedding window for COVID-19, but the transmissibility in this phase is unclear. Based on this, we develop a generalized Susceptible-Exposed-Infected-Resistant (SEIR) model including an additional compartment of chronically infected individuals who can stay infectious for a longer duration than the reported generation time, but with infectivity reduced to varying degrees. Using the incidence and fatality data from different countries, we first show that such an assumption also yields a plausible model in explaining the data observed prior to the easing of the lockdown measures (relaxation). We then test the predictive power of this model for different durations and levels of prolonged infectiousness using the incidence data after the introduction of relaxation in Switzerland, and compare it with a model without the chronically infected population to represent the models conventionally used. We show that in case of a gradual easing on the lockdown measures, the predictions of the model including the chronically infected population vary considerably from those obtained under a model in which prolonged infectiousness is not taken into account. Although the existence of a chronically infected population still remains largely hypothetical, we believe that our results provide tentative evidence to consider a chronically infected population as an alternative modeling approach to better interpret the transmission dynamics of COVID-19.


Subject(s)
COVID-19 , Communicable Disease Control , Models, Statistical , Virus Shedding/physiology , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/transmission , COVID-19/virology , Communicable Disease Control/methods , Communicable Disease Control/statistics & numerical data , Computational Biology , Humans , SARS-CoV-2 , Switzerland
18.
Sci Rep ; 11(1): 2209, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33500518

ABSTRACT

Oncolytic virotherapy is an emerging strategy that uses replication-competent viruses to kill tumor cells. We have reported the oncolytic effects of TG6002, a recombinant oncolytic vaccinia virus, in preclinical human xenograft models and canine tumor explants. To assess the safety, biodistribution and shedding of TG6002 administered by the intravenous route, we conducted a study in immune-competent healthy dogs. Three dogs each received a single intravenous injection of TG6002 at 105 PFU/kg, 106 PFU/kg or 107 PFU/kg, and one dog received three intravenous injections at 107 PFU/kg. The injections were well tolerated without any clinical, hematological or biochemical adverse events. Viral genomes were only detected in blood at the earliest sampling time point of one-hour post-injection at 107 PFU/kg. Post mortem analyses at day 35 allowed detection of viral DNA in the spleen of the dog which received three injections at 107 PFU/kg. Viral genomes were not detected in the urine, saliva or feces of any dogs. Seven days after the injections, a dose-dependent antibody mediated immune response was identified. In conclusion, intravenous administration of TG6002 shows a good safety profile, supporting the initiation of clinical trials in canine cancer patients as well as further development as a human cancer therapy.


Subject(s)
Dogs , Oncolytic Viruses , Vaccinia virus , Virus Shedding , Animals , Dogs/blood , Dogs/immunology , Dogs/virology , Male , Body Temperature , Body Weight , DNA, Viral/blood , Immunity/immunology , Injections, Intravenous , Leukocyte Count , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/physiology , Organ Specificity , Tissue Distribution , Vaccinia virus/physiology , Virus Shedding/physiology
19.
J Med Virol ; 93(1): 506-512, 2021 01.
Article in English | MEDLINE | ID: mdl-32644223

ABSTRACT

To investigate the factors associated with the duration of severe acute respiratory syndrome coronavirus 2 RNA shedding in patients with coronavirus disease 2019 (COVID-19). A retrospective cohort of COVID-19 patients admitted to a designated hospital in Beijing was analyzed to study the factors affecting the duration of viral shedding. The median duration of viral shedding was 11 days (IQR, 8-14.3 days) as measured from illness onset. Univariate regression analysis showed that disease severity, corticosteroid therapy, fever (temperature>38.5°C), and time from onset to hospitalization were associated with prolonged duration of viral shedding (P < .05). Multivariate regression analysis showed that fever (temperature>38.5°C) (OR, 5.1, 95%CI: 1.5-18.1), corticosteroid therapy (OR, 6.3, 95%CI: 1.5-27.8), and time from onset to hospitalization (OR, 1.8, 95%CI: 1.19-2.7) were associated with increased odds of prolonged duration of viral shedding. Corticosteroid treatment, fever (temperature>38.5°C), and longer time from onset to hospitalization were associated with prolonged viral shedding in COVID-19 patients.


Subject(s)
COVID-19/virology , SARS-CoV-2/physiology , Virus Shedding/physiology , Adrenal Cortex Hormones/therapeutic use , Adult , COVID-19/pathology , Female , Humans , Male , Middle Aged , RNA, Viral/isolation & purification , Risk Factors , Time Factors , COVID-19 Drug Treatment
20.
Clin Exp Immunol ; 203(3): 472-479, 2021 03.
Article in English | MEDLINE | ID: mdl-33270222

ABSTRACT

Genital cytomegalovirus (CMV) reactivation is common during the third trimester of pregnancy. We hypothesized that cervical CMV shedding may increase risk of spontaneous preterm birth (sPTB) through the release of inflammatory cytokines in the cervix. We conducted a nested case-control analysis to determine the relationship between CMV shedding and sPTB using data and samples from a prospective cohort study in western Kenya. Women who delivered between 28 + 0 and 33 + 6 weeks gestation were matched by gestational age at sample collection to controls who delivered ≥ 37 + 0 weeks. Levels of CMV DNA and interleukin (IL)-1 beta (ß), IL-6, IL-8 and tumor necrosis factor (TNF)-α were measured in cervical swabs. We used conditional logistic regression to assess relationships between CMV shedding, cervical cytokine levels and sPTB. Among 86 cases and 86 matched controls, cervical CMV levels were not significantly associated with sPTB [odds ratio (OR) = 1·23, 95% confidence interval (CI) = 0·59-2·56], but were significantly associated with higher levels of cervical IL-6 (ß = 0·15, 95% CI = 0·02-0·29) and TNF-α (ß = 0·14, 95% CI = 0·01-0·27). In univariate analysis, higher odds of sPTB was associated with higher cervical IL-6 levels (OR = 1·54, 95% CI = 1·00-2·38), but not with other cervical cytokines. In this cohort of Kenyan women, we did not find a significant association between cervical CMV shedding and sPTB before 34 weeks.


Subject(s)
Cervix Uteri/metabolism , Cervix Uteri/virology , Cytokines/metabolism , Cytomegalovirus/physiology , Virus Activation/physiology , Virus Shedding/physiology , Adult , Case-Control Studies , Female , Gestational Age , Humans , Infant, Newborn , Kenya , Logistic Models , Pregnancy , Pregnancy Trimester, Third , Premature Birth/physiopathology , Prospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...