Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.573
2.
Viruses ; 16(4)2024 Mar 26.
Article En | MEDLINE | ID: mdl-38675850

Respiratory viral infections (RVIs) are common reasons for healthcare consultations. The inpatient management of RVIs consumes significant resources. From 2009 to 2014, we assessed the costs of RVI management in 4776 hospitalized children aged 0-18 years participating in a quality improvement program, where all ILI patients underwent virologic testing at the National Reference Centre followed by detailed recording of their clinical course. The direct (medical or non-medical) and indirect costs of inpatient management outside the ICU ('non-ICU') versus management requiring ICU care ('ICU') added up to EUR 2767.14 (non-ICU) vs. EUR 29,941.71 (ICU) for influenza, EUR 2713.14 (non-ICU) vs. EUR 16,951.06 (ICU) for RSV infections, and EUR 2767.33 (non-ICU) vs. EUR 14,394.02 (ICU) for human rhinovirus (hRV) infections, respectively. Non-ICU inpatient costs were similar for all eight RVIs studied: influenza, RSV, hRV, adenovirus (hAdV), metapneumovirus (hMPV), parainfluenza virus (hPIV), bocavirus (hBoV), and seasonal coronavirus (hCoV) infections. ICU costs for influenza, however, exceeded all other RVIs. At the time of the study, influenza was the only RVI with antiviral treatment options available for children, but only 9.8% of influenza patients (non-ICU) and 1.5% of ICU patients with influenza received antivirals; only 2.9% were vaccinated. Future studies should investigate the economic impact of treatment and prevention of influenza, COVID-19, and RSV post vaccine introduction.


Cost of Illness , Hospitalization , Respiratory Tract Infections , Humans , Child, Preschool , Child , Infant , Respiratory Tract Infections/economics , Respiratory Tract Infections/virology , Respiratory Tract Infections/therapy , Germany/epidemiology , Adolescent , Male , Female , Infant, Newborn , Hospitalization/economics , COVID-19/epidemiology , COVID-19/economics , COVID-19/therapy , Inpatients , Virus Diseases/economics , Virus Diseases/therapy , SARS-CoV-2 , Health Care Costs
3.
J Pediatric Infect Dis Soc ; 13(Supplement_1): S49-S57, 2024 Feb 28.
Article En | MEDLINE | ID: mdl-38417086

Viral infections are a major source of morbidity and mortality in the context of immune deficiency and immunosuppression following allogeneic hematopoietic cell (allo-HCT) and solid organ transplantation (SOT). The pharmacological treatment of viral infections is challenging and often complicated by limited efficacy, the development of resistance, and intolerable side effects. A promising strategy to rapidly restore antiviral immunity is the adoptive transfer of virus-specific T cells (VST). This therapy involves the isolation and ex vivo expansion or direct selection of antigen-specific T cells from healthy seropositive donors, followed by infusion into the patient. This article provides a practical guide to VST therapy by reviewing manufacturing techniques, donor selection, and treatment indications. The safety and efficacy data of VSTs gathered in clinical trials over nearly 30 years is summarized. Current challenges and limitations are discussed, as well as opportunities for further research and development.


Hematopoietic Stem Cell Transplantation , Organ Transplantation , Sepsis , Virus Diseases , Humans , T-Lymphocytes , Virus Diseases/therapy , Adoptive Transfer/methods , Hematopoietic Stem Cell Transplantation/adverse effects
4.
SLAS Discov ; 29(3): 100140, 2024 Apr.
Article En | MEDLINE | ID: mdl-38182043

The use of recombinant antibodies developed through phage display technology offers a promising approach for combating viral infectious diseases. By specifically targeting antigens on viral surfaces, these antibodies have the potential to reduce the severity of infections or even prevent them altogether. With the emergence of new and more virulent strains of viruses, it is crucial to develop innovative methods to counteract them. Phage display technology has proven successful in generating recombinant antibodies capable of targeting specific viral antigens, thereby providing a powerful tool to fight viral infections. In this mini-review article, we examine the development of these antibodies using phage display technology, and discuss the associated challenges and opportunities in developing novel treatments for viral infectious diseases. Furthermore, we provide an overview of phage display technology. As these methods continue to evolve and improve, novel and sophisticated tools based on phage display and peptide display systems are constantly emerging, offering exciting prospects for solving scientific, medical, and technological problems related to viral infectious diseases in the near future.


Cell Surface Display Techniques , Recombinant Proteins , Virus Diseases , Humans , Virus Diseases/immunology , Virus Diseases/therapy , Cell Surface Display Techniques/methods , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Antibodies, Neutralizing/immunology , Peptide Library , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Animals , Antigens, Viral/immunology , Antigens, Viral/genetics
5.
WIREs Mech Dis ; 16(3): e1640, 2024.
Article En | MEDLINE | ID: mdl-38253964

Liver disease represents a significant global burden, placing individuals at a heightened risk of developing cirrhosis and liver cancer. Viral infections act as a primary cause of liver diseases on a worldwide scale. Infections involving hepatitis viruses, notably hepatitis B, C, and E viruses, stand out as the most prevalent contributors to acute and chronic intrahepatic adverse outcome, although the hepatitis C virus (HCV) can be effectively cured with antiviral drugs, but no preventative vaccination developed. Hepatitis B virus (HBV) and HCV can lead to both acute and chronic liver diseases, including liver cirrhosis and hepatocellular carcinoma (HCC), which are principal causes of worldwide morbidity and mortality. Other viruses, such as Epstein-Barr virus (EBV) and cytomegalovirus (CMV), are capable of causing liver damage. Therefore, it is essential to recognize that virus infections and liver diseases are intricate and interconnected processes. A profound understanding of the underlying relationship between virus infections and liver diseases proves pivotal in the effective prevention, diagnosis, and treatment of these conditions. In this review, we delve into the mechanisms by which virus infections induce liver diseases, as well as explore the pathogenesis, diagnosis, and treatment of liver diseases. This article is categorized under: Infectious Diseases > Biomedical Engineering.


Liver Diseases , Humans , Liver Diseases/virology , Liver Diseases/diagnosis , Liver Diseases/etiology , Liver Diseases/therapy , Virus Diseases/diagnosis , Virus Diseases/therapy , Virus Diseases/virology , Antiviral Agents/therapeutic use , Animals , Liver Neoplasms/diagnosis , Liver Neoplasms/virology , Liver Neoplasms/etiology , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/therapy
6.
JAMA Netw Open ; 6(12): e2346769, 2023 Dec 01.
Article En | MEDLINE | ID: mdl-38060222

Importance: Pediatric readiness is essential for all emergency departments (EDs). Children's experience of care may differ according to operational challenges in children's hospitals, community hospitals, and rural EDs caused by recurring and sometimes unpredictable viral illness surges. Objective: To describe wait times, lengths of stay (LOS), and ED revisits across diverse EDs participating in a statewide quality collaborative during a surge in visits in 2022. Design, Setting, and Participants: This retrospective cohort study included 25 EDs from the Michigan Emergency Department Improvement Collaborative data registry from January 1, 2021, through December 31, 2022. Pediatric (patient age <18 years) encounters for viral and respiratory conditions were analyzed, comparing wait times, LOS, and ED revisit rates for children's hospital, urban pediatric high-volume (≥10% of overall visits), urban pediatric low-volume (<10% of overall visits), and rural EDs. Exposures: Surge in ED visit volumes for children with viral and respiratory illnesses from September 1 through December 31, 2022. Main Outcomes and Measures: Prolonged ED visit wait times (arrival to clinician assigned, >4 hours), prolonged LOS (arrival to departure, >12 hours), and ED revisit rate (ED discharge and return within 72 hours). Results: A total of 2 761 361 ED visits across 25 EDs in 2021 and 2022 were included. From September 1 to December 31, 2022, there were 301 688 pediatric visits for viral and respiratory illness, an increase of 71.8% over the 4 preceding months and 15.7% over the same period in 2021. At children's hospitals during the surge, 8.0% of visits had prolonged wait times longer than 4 hours, 8.6% had prolonged LOS longer than 12 hours, and 42 revisits occurred per 1000 ED visits. Prolonged wait times were rare among other sites. However, prolonged LOS affected 425 visits (2.2%) in urban high-pediatric volume EDs, 133 (2.6%) in urban pediatric low-volume EDs, and 176 (3.1%) in rural EDs. High visit volumes were associated with increased ED revisits across sites. Conclusions and Relevance: In this cohort study of more than 2.7 million ED visits, a pediatric viral illness surge was associated with different pediatric acute care across EDs in the state. Clinical management pathways and quality improvement efforts may more effectively mitigate dangerous clinical conditions with strong collaborative relationships across EDs and setting of care.


Emergency Medical Services , Virus Diseases , Child , Humans , Adolescent , Cohort Studies , Retrospective Studies , Emergency Service, Hospital , Emergency Treatment , Virus Diseases/epidemiology , Virus Diseases/therapy
7.
Front Immunol ; 14: 1267774, 2023.
Article En | MEDLINE | ID: mdl-37928543

In the setting of viral challenge, natural killer (NK) cells play an important role as an early immune responder against infection. During this response, significant changes in the NK cell population occur, particularly in terms of their frequency, location, and subtype prevalence. In this review, changes in the NK cell repertoire associated with several pathogenic viral infections are summarized, with a particular focus placed on changes that contribute to NK cell dysregulation in these settings. This dysregulation, in turn, can contribute to host pathology either by causing NK cells to be hyperresponsive or hyporesponsive. Hyperresponsive NK cells mediate significant host cell death and contribute to generating a hyperinflammatory environment. Hyporesponsive NK cell populations shift toward exhaustion and often fail to limit viral pathogenesis, possibly enabling viral persistence. Several emerging therapeutic approaches aimed at addressing NK cell dysregulation have arisen in the last three decades in the setting of cancer and may prove to hold promise in treating viral diseases. However, the application of such therapeutics to treat viral infections remains critically underexplored. This review briefly explores several therapeutic approaches, including the administration of TGF-ß inhibitors, immune checkpoint inhibitors, adoptive NK cell therapies, CAR NK cells, and NK cell engagers among other therapeutics.


Killer Cells, Natural , Virus Diseases , Humans , Virus Diseases/therapy , Immunotherapy, Adoptive
8.
Sci Rep ; 13(1): 18168, 2023 10 24.
Article En | MEDLINE | ID: mdl-37875555

Canine primary lung cancer with metastasis has a poor prognosis with no effective treatment. We previously generated a recombinant measles virus (MV) that lost binding affinity to a principal receptor, SLAM, to eliminate its virulence as a new cancer treatment strategy. The virus, rMV-SLAMblind, targets nectin-4, recently listed as a tumor marker, and exerts antitumor activity against nectin-4-positive canine mammary cancer and urinary bladder transitional cell carcinoma cells. However, the effectivity of rMV-SLAMblind for other types of canine cancers is still unknown. Here we evaluated the antitumor effect of rMV-SLAMblind to canine lung cancer. Nectin-4 is expressed on three canine lung cancer cell lines (CLAC, AZACL1, AZACL2) and rMV-SLAMblind was able to infect these cell lines. CLAC cells showed reduced cell viability after virus infection. In the CLAC xenograft nude mouse model, intratumoral administration of rMV-SLAMblind significantly suppressed tumor growth. In rMV-SLAMblind-treated mice, natural killer cells were activated, and Cxcl10 and Il12a levels were significantly increased in comparison with levels in the control group. In addition, the depletion of NK cells reduced the anti-tumor effect. To understand difference in efficacy among canine lung cancer cell lines, we compared virus growth and gene expression pattern after virus treatment in the three canine lung cancer cell lines; virus growth was highest in CLAC cells compared with the other cell lines and the induction of interferon (IFN)-beta and IFN-stimulated genes was at lower levels in CLAC cells. These results suggested that rMV-SLAMblind exhibits oncolytic effect against some canine lung cancer cells and the cellular response after the virus infection may influence its efficacy.


Lung Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Virus Diseases , Humans , Animals , Dogs , Mice , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Lung Neoplasms/metabolism , Measles virus/metabolism , Oncolytic Virotherapy/methods , Nectins/metabolism , Cell Line, Tumor , Cell Adhesion Molecules/metabolism , Virus Diseases/therapy , Xenograft Model Antitumor Assays , Oncolytic Viruses/genetics
9.
Viruses ; 15(10)2023 10 05.
Article En | MEDLINE | ID: mdl-37896827

Shrimp aquaculture has become a vital industry, meeting the growing global demand for seafood. Shrimp viral diseases have posed significant challenges to the aquaculture industry, causing major economic losses worldwide. Conventional treatment methods have proven to be ineffective in controlling these diseases. However, recent advances in RNA interference (RNAi) technology have opened new possibilities for combating shrimp viral diseases. This cutting-edge technology uses cellular machinery to silence specific viral genes, preventing viral replication and spread. Numerous studies have shown the effectiveness of RNAi-based therapies in various model organisms, paving the way for their use in shrimp health. By precisely targeting viral pathogens, RNAi has the potential to provide a sustainable and environmentally friendly solution to combat viral diseases in shrimp aquaculture. This review paper provides an overview of RNAi-based therapy and its potential as a game-changer for shrimp viral diseases. We discuss the principles of RNAi, its application in combating viral infections, and the current progress made in RNAi-based therapy for shrimp viral diseases. We also address the challenges and prospects of this innovative approach.


RNAi Therapeutics , Virus Diseases , Animals , RNA Interference , Virus Diseases/genetics , Virus Diseases/therapy , Crustacea , Aquaculture
10.
RNA Biol ; 20(1): 847-858, 2023 01.
Article En | MEDLINE | ID: mdl-37882652

Circular RNA (circRNA) forms closed loops via back-splicing in precursor mRNA, resisting exonuclease degradation. In higher eukaryotes, protein-coding genes create circRNAs through exon back-splicing. Unlike mRNAs, circRNAs possess unique production and structural traits, bestowing distinct cellular functions and biomedical potential. In this review, we explore the pivotal roles of viral circRNAs and associated RNA in various biological processes. Analysing the interactions between viral circRNA and host cellular machinery yields fresh insights into antiviral immunity, catalysing the development of potential therapeutics. Furthermore, circRNAs serve as enduring biomarkers in viral diseases due to their stable translation within specific tissues. Additionally, a deeper understanding of translational circRNA could expedite the establishment of circRNA-based expression platforms, meeting the rising demand for broad-spectrum viral vaccines. We also highlight the applications of circular RNA in biomarker studies as well as circRNA-based therapeutics. Prospectively, we expect a technological revolution in combating viral infections using circRNA.


MicroRNAs , Virus Diseases , Humans , RNA, Circular/genetics , RNA, Circular/metabolism , RNA/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA Splicing , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Diseases/diagnosis , Virus Diseases/genetics , Virus Diseases/therapy , MicroRNAs/genetics
11.
Microb Pathog ; 182: 106234, 2023 Sep.
Article En | MEDLINE | ID: mdl-37442216

Growing antimicrobial resistance (AMR) is a threat to human and animal populations citing the limited available options. Alternative antimicrobial options or functional enhancement of currently available antimicrobials remains only options. One of the potential options seems stem cells especially the mesenchymal stem cells (MSCs) that show antimicrobial properties. These cells additionally have pro-healing effects that may plausibly improve healing outcomes. MSCs antimicrobial actions are mediated either through direct cell-cell contact or their secretome that enhances innate immune mediated antimicrobial activities. These cells synergistically enhance efficacy of currently available antimicrobials especially against the biofilms. Reciprocal action from antimicrobials on the MSCs functionality remains poorly understood. Currently, the main limitation with MSCs based therapy is their limited efficacy. This demands further understanding and can be enhanced through biotechnological interventions. One of the interventional options is the 'priming' to enhance MSCs resistance and specific expression potential. The available literature shows potential antimicrobial actions of MSCs both ex vivo as well as in vivo. The studies on veterinary species are very promising although limited by number and extensiveness in details for their utility as standard therapeutic agents. The current review aims to discuss the role of animals in AMR and the potential antimicrobial actions of MSCs in veterinary medicine. The review also discusses the limitations in their utilization as standard therapeutics.


Bacterial Infections , Cell- and Tissue-Based Therapy , Mesenchymal Stem Cells , Animals , Animals, Domestic , Bacterial Infections/therapy , Bacterial Infections/veterinary , Cell- and Tissue-Based Therapy/veterinary , Drug Resistance , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Virus Diseases/therapy , Virus Diseases/veterinary , Secretome , Antimicrobial Peptides/metabolism
12.
Front Cell Infect Microbiol ; 13: 1173505, 2023.
Article En | MEDLINE | ID: mdl-37465759

The inflammasome is a multiprotein complex that further regulates cell pyroptosis and inflammation by activating caspase-1. The assembly and activation of inflammasome are associated with a variety of diseases. Accumulative studies have shown that inflammasome is a key modulator of the host's defense response to viral infection. Indeed, it has been established that activation of inflammasome occurs during viral infection. At the same time, the host has evolved a variety of corresponding mechanisms to inhibit unnecessary inflammasome activation. Therefore, here, we review and summarize the latest research progress on the interaction between inflammosomes and viruses, highlight the assembly and activation of inflammosome in related cells after viral infection, as well as the corresponding molecular regulatory mechanisms, and elucidate the effects of this activation on virus immune escape and host innate and adaptive immune defenses. Finally, we also discuss the potential therapeutic strategies to prevent and/or ameliorate viral infection-related diseases via targeting inflammasomes and its products.


Host Microbial Interactions , Inflammasomes , Virus Diseases , Viruses , Humans , Inflammasomes/immunology , Virus Diseases/immunology , Virus Diseases/therapy , Viruses/immunology , Host Microbial Interactions/immunology , Animals
14.
Semin Hematol ; 60(1): 10-19, 2023 01.
Article En | MEDLINE | ID: mdl-37080705

Hematopoietic stem cell transplantation (HSCT) has been used as a curative standard of care for moderate to severe primary immunodeficiency disorders as well as relapsed hematologic malignancies for over 50 years [1,2]. However, chronic and refractory viral infections remain a leading cause of morbidity and mortality in the immune deficient period following HSCT, where use of available antiviral pharmacotherapies is limited by toxicity and emerging resistance [3]. Adoptive immunotherapy using virus-specific T cells (VSTs) has been explored for over 2 decades [4,5] in patients post-HSCT and has been shown prior phase I-II studies to be safe and effective for treatment or preventions of viral infections including cytomegalovirus, Epstein-Barr virus, BK virus, and adenovirus with minimal toxicity and low risk of graft vs host disease [6-9]. This review summarizes methodologies to generate VSTs the clinical results utilizing VST therapeutics and the challenges and future directions for the field.


Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Virus Diseases , Humans , T-Lymphocytes/transplantation , Herpesvirus 4, Human , Neoplasm Recurrence, Local , Virus Diseases/therapy , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods
16.
Viruses ; 15(2)2023 01 26.
Article En | MEDLINE | ID: mdl-36851568

The myocardium/heart is the most mitochondria-rich tissue in the human body with mitochondria comprising approximately 30% of total cardiomyocyte volume. As the resident "powerhouse" of cells, mitochondria help to fuel the high energy demands of a continuously beating myocardium. It is no surprise that mitochondrial dysfunction underscores the pathogenesis of many cardiovascular ailments, including those of viral origin such as virus-induced myocarditis. Enteroviruses have been especially linked to injuries of the myocardium and its sequelae dilated cardiomyopathy for which no effective therapies currently exist. Intriguingly, recent mechanistic insights have demonstrated viral infections to directly damage mitochondria, impair the mitochondrial quality control processes of the cell, such as disrupting mitochondrial antiviral innate immune signaling, and promoting mitochondrial-dependent pathological inflammation of the infected myocardium. In this review, we briefly highlight recent insights on the virus-mitochondria crosstalk and discuss the therapeutic implications of targeting mitochondria to preserve heart function and ultimately combat viral myocarditis.


Myocarditis , Virus Diseases , Humans , Myocarditis/therapy , Myocardium , Virus Diseases/therapy , Myocytes, Cardiac , Mitochondria
17.
Haematologica ; 108(8): 2080-2090, 2023 08 01.
Article En | MEDLINE | ID: mdl-36794500

Therapy-resistant viral reactivations contribute significantly to mortality after hematopoietic stem cell transplantation. Adoptive cellular therapy with virus-specific T cells (VST) has shown efficacy in various single-center trials. However, the scalability of this therapy is hampered by laborious production methods. In this study we describe the in-house production of VST in a closed system (CliniMACS Prodigy® system, Miltenyi Biotec). In addition, we report the efficacy in 26 patients with viral disease following hematopoietic stem cell transplantation in a retrospective analysis (adenovirus, n=7; cytomegalovirus, n=8; Epstein-Barr virus, n=4; multi-viral, n=7). The production of VST was successful in 100% of cases. The safety profile of VST therapy was favorable (n=2 grade 3 and n=1 grade 4 adverse events; all three were reversible). A response was seen in 20 of 26 patients (77%). Responding patients had a significantly better overall survival than patients who did not respond (P<0.001). Virus-specific symptoms were reduced or resolved in 47% of patients. The overall survival of the whole cohort was 28% after 6 months. This study shows the feasibility of automated VST production and safety of application. The scalability of the CliniMACS Prodigy® device increases the accessibility of VST treatment.


Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Virus Diseases , Humans , T-Lymphocytes , Epstein-Barr Virus Infections/therapy , Retrospective Studies , Herpesvirus 4, Human , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Virus Diseases/etiology , Virus Diseases/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Stem Cell Transplantation
18.
Transplant Cell Ther ; 29(5): 305-310, 2023 05.
Article En | MEDLINE | ID: mdl-36736781

Infections with double-stranded DNA viruses are a common complication after hematopoietic stem cell transplantation (HSCT) and cause significant morbidity and mortality in the post-transplantation period. Both donor-derived (DD) and third-party (TP) virus-specific T cells (VSTs) have shown efficacy and safety in viral management following HSCT in children and young adults. Owing to a greater degree of HLA matching between the recipient and stem cell donor, DD VSTs potentially persist longer in circulation compared to TP VSTs, because they are collected from a well-matched donor. However, TP VSTs are more easily accessible, particularly for smaller transplantation centers that do not have VST manufacturing capabilities, and more economical than creating a customized product for each transplant recipient. We conducted the present study to compare clinical efficacy and safety outcomes for DD VSTs and TP VSTs in a large cohort of pediatric and young adult HSCT recipients and to determine whether DD VSTs are associated with improved outcomes owing to potentially longer persistence in the recipient's circulation. This retrospective cohort study included 145 patients who received VSTs at Cincinnati Children's Hospital Medical Center (CCHMC) between 2017 and 2021 for the treatment of adenovirus, BK virus, cytomegalovirus, and/or Epstein-Barr virus. Viruses were detected using quantitative polymerase chain reaction. Patients received VSTs on a DD (NCT02048332) or TP (NCT02532452) protocol, and VST products for both protocols were manufactured in an identical fashion. The primary study outcome was clinical response to VSTs, evaluated 4 weeks after VST administration, defined as decrease in viral load to under the inclusion thresholds, or resolution of symptoms of invasive viral infection, without the need for additional conventional antiviral medication following VST administration. Secondary outcomes included graft-versus-host-disease, transplant-associated thrombotic microangiopathy, renal function, hospital length of stay, and overall survival at 30 days and 100 days after VST administration and 1 year after HSCT. Statistical analysis was performed using the Fisher exact test or chi-square test. An unpaired t test was used to compare continuous variables. The study group comprised 77 patients in the DD cohort and 68 patients in the TP cohort. Eighteen patients in the TP cohort underwent HSCT at CCHMC, and the other 50 underwent HSCT at other institutions and presented to CCHMC solely for VST administration. There was no statistically significant difference in clinical response rates between DD and TP cohorts (65.6% versus 62.7%; odds ratio [OR], 1.162; 95% confidence interval [CI], .619 to 2.164; P = .747). There were no significant differences in secondary outcomes between the 2 cohorts. The percentage of patients requiring multiple infusions for a clinical response did not differ significantly between the DD and TP cohorts (38.2% versus 32.5%; OR, .780; 95% CI, .345 to 1.805; P = .666). We found no significant difference in clinical response rate between DD VSTs and TP VSTs and a similar safety profile. Our data suggest that TP VSTs may be sufficient to control viral infection until immune reconstitution occurs despite the potential for more rapid VST clearance compared to DD VSTs. The lack of significant differences between DD VSTs and TP VSTs is an important finding, indicating that it is not necessary for every transplant center to manufacture customized DD VSTs, and that TP VSTs are a satisfactory substitute.


Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Virus Diseases , Child , Humans , Young Adult , Hematopoietic Stem Cell Transplantation/adverse effects , Herpesvirus 4, Human , Retrospective Studies , T-Lymphocytes , Transplantation, Homologous , Virus Diseases/etiology , Virus Diseases/therapy
19.
Anaesthesia ; 78(5): 626-635, 2023 05.
Article En | MEDLINE | ID: mdl-36633460

Viral infections form a substantial part of the intensive care workload, even before the recent and ongoing COVID-19 pandemic. The growing availability of molecular diagnostics for viral infections has led to increased recognition of these pathogens. This additional information, however, provides new challenges for interpretation and management. As the SARS-CoV-2 pandemic has amply demonstrated, the emergence and global spread of novel viruses are likely to provide continued challenges for critical care physicians into the future. This article will provide an overview of viral infections relevant to the critical care physician, discussing the diagnosis and management of respiratory viral infections, blood borne and enteric viruses. We will also discuss herpesviridae complications, commonly seen due to reactivation of latent infections. Further, we explore some rarer and emerging viruses, including recognition of viral haemorrhagic fevers, and briefly discuss post-viral syndromes which may present to the intensive care unit. Finally, we will discuss infection control and its importance in preventing nosocomial viral transmission.


COVID-19 , Virus Diseases , Humans , COVID-19/prevention & control , SARS-CoV-2 , Pandemics , Virus Diseases/diagnosis , Virus Diseases/therapy , Critical Care
20.
Methods Mol Biol ; 2614: 139-149, 2023.
Article En | MEDLINE | ID: mdl-36587124

Oncolytic viruses (OVs) rapidly and specifically replicate in and kill tumor cells. OV-targeted infection of malignant cells has the potential to create an "inflammatory storm" that stimulates both innate and adaptive anti-tumor immune responses. The generation of anti-tumor immunity following OV treatment has been shown to be crucial for effective therapy. Therefore, establishing methodologies to measure the generation of anti-tumor T cell responses following OV infection in in vitro assays, which better mimic the complexity of the human tumor microenvironment (TME), will be critical to harness the full potential of OV therapy. Such experimental platforms will accelerate the development of next-generation OVs that are capable of overcoming immunosuppressive networks found within the tumor microenvironment. Here we describe a method that was designed to test the generation and quantification of human tumor-specific T cells following OV infection of 3D tumor spheroids cultured with or without fibroblasts.


Antineoplastic Agents , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Virus Diseases , Humans , Oncolytic Virotherapy/methods , Neoplasms/therapy , T-Lymphocytes , Tumor Microenvironment , Virus Diseases/therapy
...